Scolaris Content Display Scolaris Content Display

Administración de suplementos de vitamina D para las hepatopatías crónicas en adultos

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Abu‐Mouch 2011 {published data only}

Abu‐Mouch S, Fireman Z, Jarchovsky J, Zeina AR, Assy N. Vitamin D supplementation improves sustained virologic response in chronic hepatitis C (genotype 1)‐naïve patients. World Journal of Gastroenterology 2011;17(47):5184‐90. CENTRAL

Atsukawa 2016 {published data only}

Atsukawa M, Tsubota A, Shimada N, Yoshizawa K, Abe H, Asano T, et al. Effect of native vitamin D3 supplementation on refractory chronic hepatitis C patients in simeprevir with pegylated interferon/ribavirin. Hepatology Research 2016;46(5):450‐8. CENTRAL

Barchetta 2016 {published data only}

Barchetta I, Del Ben M, Angelico F, Di Martino M, Fraioli A, La Torre G, et al. No effects of oral vitamin D supplementation on non‐alcoholic fatty liver disease in patients with type 2 diabetes: a randomized, double‐blind, placebo‐controlled trial. BMC Medicine 2016;14:92. CENTRAL

Esmat 2015 {published data only}

Esmat G, El Raziky M, Elsharkawy A, Sabry D, Hassany M, Ahmed A, et al. Impact of vitamin D supplementation on sustained virological response in chronic hepatitis C genotype 4 patients treated by pegylated interferon/ribavirin. Journal of Interferon & Cytokine Research 2015;35(1):49‐54. CENTRAL

Foroughi 2016 {published data only}

Foroughi M, Maghsoudi Z, Askari G. The effect of vitamin D supplementation on blood sugar and different indices of insulin resistance in patients with non‐alcoholic fatty liver disease (NAFLD). Iranian Journal of Nursing and Midwifery Research 2016;21(1):100‐4. CENTRAL
Foroughi M, Maghsoudi Z, Ghiasvand R, Iraj B, Askari G. Effect of vitamin D supplementation on C‐reactive protein in patients with nonalcoholic fatty liver. International Journal of Preventive Medicine 2014;5(8):969‐75. CENTRAL

Lorvand Amiri 2016 {published data only}

Lorvand Amiri H, Agah S, Mousavi SN, Hosseini AF, Shidfar F. Regression of non‐alcoholic fatty liver by vitamin D supplement: a double‐blind randomized controlled clinical trial. Archives of Iranian Medicine 2016;19(9):631‐8. CENTRAL
Lorvand Amiri H, Agah S, Tolouei Azar J, Hosseini S, Shidfar F, Mousavi SN. Effect of daily calcitriol supplementation with and without calcium on disease regression in non‐alcoholic fatty liver patients following an energy‐restricted diet: randomized, controlled, double‐blind trial. Clinical Nutrition 2016;16:31260‐2. CENTRAL

Mobarhan 1984 {published data only}

Mobarhan SA, Russell RM, Recker RR, Posner DB, Iber FL, Miller P. Metabolic bone disease in alcoholic cirrhosis: a comparison of the effect of vitamin D2, 25‐hydroxyvitamin D, or supportive treatment. Hepatology 1984;4(2):266‐73. CENTRAL

Nimer 2012 {published data only}

Nimer A, Mouch A. Vitamin D improves viral response in hepatitis C genotype 2‐3 naïve patients. World Journal of Gastroenterology 2012;18(8):800‐5. CENTRAL

Pilz 2016 {published data only}

Pilz S, Putz‐Bankuti C, Gaksch M, Spindelboeck W, Haselberger M, Rainer F, et al. Effects of vitamin D supplementation on serum 25‐Hydroxyvitamin D concentrations in cirrhotic patients: a randomized controlled trial. Nutrients 2016;8(5):pii: E278. CENTRAL

Sharifi 2014 {published data only}

Sharifi N, Amani R, Hajiani E, Cheraghian B. Does vitamin D improve liver enzymes, oxidative stress, and inflammatory biomarkers in adults with non‐alcoholic fatty liver disease? A randomized clinical trial. Endocrine 2014;47(1):70‐80. CENTRAL

Shiomi 1999a {published data only}

Shiomi S, Masaki K, Habu D, Takeda T, Nishiguchi S, Kuroki T, et al. Calcitriol for bone disease in patients with cirrhosis of the liver. Journal of Gastroenterology and Hepatology 1999;14(6):547‐52. CENTRAL

Shiomi 1999b {published data only}

Shiomi S, Masaki K, Habu D, Takeda T, Nishiguchi S, Kuroki T, et al. Calcitriol for bone loss in patients with primary biliary cirrhosis. Journal of Gastroenterology 1999;34(2):241‐5. CENTRAL

Vosoghinia 2016 {published data only}

Vosoghinia H, Esmaeilzadeh A, Ganji A, Hosseini SM, Jamehdar SA, Salehi M, et al. Vitamin D in standard HCV regimen (PEG‐Interferon plus Ribavirin), its effect on the early virologic r response rate: a clinical trial. Razavi Int J Med 2016;4(2):e36632. CENTRAL

Xing 2013 {published data only}

Xing T, Qiu G, Zhong L, Ling L, Huang L, Peng Z. Calcitriol reduces the occurrence of acute cellular rejection of liver transplants: a prospective controlled study. Pharmazie 2013;68(10):821‐6. CENTRAL

Yokoyama 2014 {published data only}

Yokoyama S, Takahashi S, Kawakami Y, Hayes CN, Kohno H, Kohno H, et al. Effect of vitamin D supplementation on pegylated interferon/ribavirin therapy for chronic hepatitis C genotype 1b: a randomized controlled trial. Journal of Viral Hepatitis 2014;21(5):348‐56. CENTRAL

Referencias de los estudios excluidos de esta revisión

Atsukawa 2013 {published data only}

Atsukawa M, Tsubota A, Shimada N, Kondo C, Itokawa N, Nakagawa A, et al. Efficacy of alfacalcidol on PEG‐IFN/ribavirin combination therapy for elderly patients with chronic hepatitis C: a pilot study. Hepatitis Monthly 2013;13(12):e14872. CENTRAL

Benetti 2008 {published data only}

Benetti A, Crosignani A, Varenna M, Giussani CS, Allocca M, Zuin M, et al. Primary biliary cirrhosis is not an additional risk factor for bone loss in women receiving regular calcium and vitamin D supplementation: a controlled longitudinal study. Journal of Clinical Gastroenterology 2008;42(3):306‐11. CENTRAL

Bitetto 2010 {published data only}

Bitetto D, Fabris C, Fornasiere E, Pipan C, Fumolo E, Cussigh A, et al. Vitamin D supplementation improves response to antiviral treatment for recurrent hepatitis C. Transplant International 2011;24(1):43‐50. CENTRAL

Fernández Fernández 2016 {published data only}

Fernández Fernández N, Linares Torres P, Joáo Matias D, Jorquera Plaza F, Olcoz Goñi JL. Vitamin D deficiency in chronic liver disease, clinical‐epidemiological analysis and report after vitamin d supplementation [Déficit de vitamina D en la enfermedad hepática crónica, análisis clínico epidemiológico y tras aporte vitamínico]. Gastroenterologia y Hepatologia 2016;39(5):305‐10. CENTRAL

Floreani 2007 {published data only}

Floreani A, Carderi I, Ferrara F, Rizzotto ER, Luisetto G, Camozzi V, et al. A 4‐year treatment with clodronate plus calcium and vitamin D supplements does not improve bone mass in primary biliary cirrhosis. Digestive and Liver Disease 2007;39(6):544‐8. CENTRAL

Kitson 2016 {published data only}

Kitson MT, Pham A, Gordon A, Kemp W, Roberts SK. High‐dose vitamin D supplementation and liver histology in NASH. Gut 2016;65(4):717‐8. CENTRAL

Kondo 2013 {published data only}

Kondo Y, Kato T, Kimura O, Iwata T, Ninomiya M, Kakazu E, et al. 1(OH) vitamin D3 supplementation improves the sensitivity of the immune‐response during Peg‐IFN/RBV therapy in chronic hepatitis C patients‐case controlled trial. PloS One 2013;8(5):e63672. CENTRAL

Ladero 2013 {published data only}

Ladero JM, Torrejón MJ, Sánchez‐Pobre P, Suárez A, Cuenca F, de la Orden V, et al. Vitamin D deficiency and vitamin D therapy in chronic hepatitis C. Annals of Hepatology 2013;12(2):199‐204. CENTRAL

Long 1978 {published data only}

Long RG, Varghese Z, Meinhard EA, Skinner RK, Wills MR, Sherlock S. Parenteral 1,25‐dihydroxycholecalciferol in hepatic osteomalacia. British Medical Journal 1978;1(6105):75‐7. CENTRAL

Malham 2012 {published data only}

Malham M, Peter Jørgensen S, Lauridsen AL, Ott P, Glerup H, Dahlerup JF. The effect of a single oral megadose of vitamin D provided as either ergocalciferol (D2) or cholecalciferol (D3) in alcoholic liver cirrhosis. European Journal of Gastroenterology & Hepatology 2012;24(2):172‐8. CENTRAL

Papapostoli 2016 {published data only}

Papapostoli I, Lammert F, Stokes CS. Effect of short‐term vitamin D correction on hepatic steatosis as quantified by controlled attenuation parameter (CAP). Journal of Gastrointestinal and Liver Diseases 2016;25(2):175‐81. CENTRAL

Rode 2010 {published data only}

Rode A, Fourlanos S, Nicoll A. Oral vitamin D replacement is effective in chronic liver disease [Fréquence du déficit en vitamine D et effet de la supplémentation orale au cours des maladies chroniques du foie]. Gastroentérologie Clinique et Biologique 2010;34(11):618‐20. CENTRAL

Stokes 2016 {published data only}

Stokes CS, Grünhage F, Baus C, Volmer DA, Wagenpfeil S, Riemenschneider M, et al. Vitamin D supplementation reduces depressive symptoms in patients with chronic liver disease. Clinical Nutrition (Edinburgh, Scotland) 2016;35(4):950‐7. CENTRAL

Terrier 2015 {published data only}

Terrier B, Lapidus N, Pol S, Serfaty L, Ratziu V, Asselah T, et al. Vitamin D in addition to peg‐interferon‐alpha/ribavirin in chronic hepatitis C virus infection: ANRS‐HC25‐VITAVIC study. World Journal of Gastroenterology 2015;21(18):5647‐53. CENTRAL

IRCT2016020326342N1 {published data only}

Effectiveness of Vitamin D Supplementation on Severity of Cirrhosis Based on CHILD and MELD Scores in Patients with Decompensate Cirrhosis.. Ongoing studyMarch 2016..

NCT02779465 {published data only}

Study of Oral Vitamin D Treatment for the Prevention of Hepatocellular Carcinoma in Patients with Chronic Hepatitis B.. Ongoing studyJune 2016..

Arteh 2010

Arteh J, Narra S, Nair S. Prevalence of vitamin D deficiency in chronic liver disease. Digestive Diseases and Sciences 2010;55(9):2624‐8.

Atef 2017

Atef SH. Vitamin D assays in clinical laboratory: past, present and future challenges. Journal of Steroid Biochemistry and Molecular Biology 2017 Feb 24 [Epub ahead of print].

Autier 2014

Autier P, Boniol M, Mullie P. Vitamin D status and ill health: a systematic review. The Lancet. Diabetes & Endocrinology 2014;2(1):76‐89.

Autier 2016

Autier P. Vitamin D status as a synthetic biomarker of health status. Endocrine 2016;51(2):201‐2.

Begg 1994

Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics 1994;50(4):1088‐101.

Bischoff‐Ferrari 2009

Bischoff‐Ferrari H. Vitamin D: what is an adequate vitamin D level and how much supplementation is necessary. Best Practice & Research. Clinical Rheumatology 2009;23(6):789‐95.

Bitetto 2011

Bitetto D, Fabris C, Falleti E, Toniutto P. Vitamin D deficiency and HCV chronic infection: what comes first. Journal of Hepatology 2011;55(4):944‐5.

Bitetto 2012

Bitetto D, Fabris C, Fornasiere E, Pipan C, Fumolo E, Cussigh A, et al. Vitamin D supplementation improves response to antiviral treatment for recurrent hepatitis C. Transplant International 2012;24(1):43‐50.

Bjelakovic 2014a

Bjelakovic G, Gluud LL, Nikolova D, Whitfield K, Wetterslev J, Simonetti RG, et al. Vitamin D supplementation for prevention of mortality in adults. Cochrane Database of Systematic Reviews 2014, Issue 1. [DOI: 10.1002/14651858.CD007470.pub3]

Bjelakovic 2014b

Bjelakovic G, Gluud LL, Nikolova D, Whitfield K, Krstic G, Wetterslev J, et al. Vitamin D supplementation for prevention of cancer in adults. Cochrane Database of Systematic Reviews 2014, Issue 6. [DOI: 10.1002/14651858.CD007469.pub2]

Bolland 2006

Bolland MJ, Grey AB, Ames RW, Mason BH, Horne AM, Gamble GD, et al. Determinants of vitamin D status in older men living in a subtropical climate. Osteoporosis International 2006;17(12):1742‐8.

Bolland 2014

Bolland MJ, Grey A, Gamble GD, Reid IR. The effect of vitamin D supplementation on skeletal, vascular, or cancer outcomes: a trial sequential meta‐analysis. Lancet, Diabetes Endocrinology 2014;2(4):307‐20.

Borenstein 2009

Borenstein M, Hedges LV, Higgins T, Rothstein HR. Introduction to Meta‐Analysis. Chichester (UK): John Wiley & Sons, 2009.

Bradburn 2007

Bradburn MJ, Deeks JJ, Berlin JA, Russell Localio A. Much ado about nothing: a comparison of the performance of meta‐analytical methods with rare events. Statistics in Medicine 2007;26:53‐77.

Brok 2008

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

Brok 2009

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

Cacopardo 2012

Cacopardo B, Camma C, Petta S, Pinzone MR, Cappellani A, Zanghi A, et al. Diagnostic and therapeutical role of vitamin D in chronic hepatitis C virus infection. Frontiers in Bioscience (Elite Edition) 2012;4:1276‐86.

Chan 2004

Chan AW, Hróbjartsson A, Haahr MT, Gøtzsche PC, Altman DG. Empirical evidence for selective reporting of outcomes in randomized trials: comparison of protocols to published articles. JAMA 2004;291(20):2457‐65.

Chen 2014

Chen EQ, Shi Y, Tang H. New insight of vitamin D in chronic liver diseases. Hepatobiliary & Pancreatic Diseases International 2014;13(6):580‐5.

Chiang 2011

Chiang KC, Yeh CN, Chen MF, Chen TC. Hepatocellular carcinoma and vitamin D: a review. Journal of Gastroenterology and Hepatology 2011;26(11):1597‐603.

Cholongitas 2012

Cholongitas E, Theocharidou E, Goulis J, Tsochatzis E, Akriviadis E, Burroughs K. Review article: the extra‐skeletal effects of vitamin D in chronic hepatitis C infection. Alimentary Pharmacology & Therapeutics 2012;35(6):634‐46.

Cohen 1960

Cohen J. A coefficient of agreement for nominal scales. Educational and Psychological Measurement 1960;20:37‐46.

Corey 2012

Corey KE, Zheng H, Mendez‐Navarro J, Delgado‐Borrego A, Dienstag JL, Chung RT, et al. Serum vitamin D levels are not predictive of the progression of chronic liver disease in hepatitis C patients with advanced fibrosis. PloS One 2012;7(2):e27144.

Davis 2007

Davis CD, Dwyer JT. The "sunshine vitamin": benefits beyond bone?. Journal of the National Cancer Institute 2007;99(21):1563‐5.

Dawson‐Hughes 2005

Dawson‐Hughes B, Heaney RP, Holick MF, Lips P, Meunier PJ, Vieth R. Estimates of optimal vitamin D status. Osteoporosis International 2005;16(7):713‐6.

Dawson‐Hughes 2010

Dawson‐Hughes B, Mithal A, Bonjour JP, Boonen S, Burckhardt P, Fuleihan GE, et al. IOF position statement: vitamin D recommendations for older adults. Osteoporosis International 2010;21(7):1151‐4.

DeMets 1987

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

DerSimonian 1986

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

Egger 1997

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

Elangovan 2017

Elangovan H, Chahal S, Gunton JE. Vitamin D in liver disease: current evidence and potential directions. Biochimica et Biophysica Acta 2017;1863(4):907‐16.

Eliades 2013

Eliades M, Spyrou E, Agrawal N, Lazo M, Brancati FL, Potter JJ, et al. Meta‐analysis: vitamin D and non‐alcoholic fatty liver disease. Alimentary Pharmacology & Therapeutics 2013;38(3):246‐54.

Eliades 2015

Eliades M, Spyrou E. Vitamin D: a new player in non‐alcoholic fatty liver disease. World Journal of Gastroenterology 2015;21(6):1718‐27.

Farnik 2013

Farnik H, Bojunga J, Berger A, Allwinn R, Waidmann O, Kronenberger B, et al. Low vitamin D serum concentration is associated with high levels of hepatitis B virus replication in chronically infected patients. Hepatology 2013;58(4):1270‐6.

Finkelmeier 2014

Finkelmeier F, Kronenberger B, Köberle V, Bojunga J, Zeuzem S, Trojan J, et al. Severe 25‐hydroxyvitamin D deficiency identifies a poor prognosis in patients with hepatocellular carcinoma ‐ a prospective cohort study. Alimentary Pharmacology & Therapeutics 2014;39(10):1204‐12.

Finkelmeier 2015

Finkelmeier F, Kronenberger B, Zeuzem S, Piiper A, Waidmann O. Low 25‐hydroxyvitamin D levels are associated with infections and mortality in patients with cirrhosis. PloS One 2015;10(6):e0132119.

Fortmann 2013

Fortmann SP, Burda BU, Senger CA, Lin JS, Whitlock EP. Vitamin and mineral supplements in the primary prevention of cardiovascular disease and cancer: an updated systematic evidence review for the U.S. Preventive Services Task Force. Annals of Internal Medicine 2013;159(12):824‐34.

Furukawa 2007

Furukawa TA, Watanabe N, Omori IM, Montori VM, Guyatt GH. Association between unreported outcomes and effect size estimates in Cochrane meta‐analyses. JAMA 2007;297(5):468‐70.

Garattini 2016

Garattini S, Jakobsen JC, Wetterslev J, Bertelé 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.

Geier 2011

Geier A. Shedding new light on vitamin D and fatty liver disease. Journal of Hepatology 2011;55(2):273‐5.

Gluud 2006

Gluud C. The culture of designing hepato‐biliary randomised trials. Journal of Hepatology 2006;44(3):607‐15. [MEDLINE: 16434120]

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 2008

Gluud C, Hilden J. Povl Heiberg's 1897 methodological study on the statistical method as an aid in therapeutic trials, 2008. www.jameslindlibrary.org/articles/povl‐heibergs‐1897‐methodological‐study‐on‐the‐statistical‐method‐as‐an‐aid‐in‐therapeutic‐trials/ (accessed 17 July 2017).

Gluud 2017

Gluud C, Nikolova D, Klingenberg SL. Cochrane Hepato‐Biliary Group. About The Cochrane Collaboration (Cochrane Review Groups (CRGs)). 2017; Issue 1. Art. No.: LIVER.

GRADEpro [Computer program]

GRADE Working Group, McMaster University. GRADEpro. Hamilton (ON): GRADE Working Group, McMaster University, 2014.

Grey 2010

Grey A, Bolland M. Vitamin D: a place in the sun. Archives of Internal Medicine 2010;170(13):1099‐100.

Guallar 2010

Guallar E, Miller ER, Ordovas JM, Stranges S. Vitamin D supplementation in the age of lost innocence. Annals of Internal Medicine 2010;152(5):327‐9.

Guañabens 2010

Guañabens N, Parés A. Liver and bone. Archives of Biochemistry and Biophysics 2010;503(1):84‐94.

Gutierrez 2011

Gutierrez JA, Parikh N, Branch AD. Classical and emerging roles of vitamin D in hepatitis C virus infection. Seminars in Liver Disease 2011;31(4):387‐98.

Han 2013

Han YP, Kong M, Zheng S, Ren Y, Zhu L, Shi H, et al. Vitamin D in liver diseases: from mechanisms to clinical trials. Journal of Gastroenterology and Hepatology 2013;28(Suppl 1):49‐55.

Harvey 2012

Harvey NC, Cooper C. Vitamin D: some perspective please. BMJ 2012;345:e4695. [DOI: 10.1136/bmj.e4695]

Higgins 2002

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

Higgins 2003

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

Higgins 2011

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

Hilger 2014

Hilger J, Friedel A, Herr R, Rausch T, Roos F, Wahl DA, et al. A systematic review of vitamin D status in populations worldwide. British Journal of Nutrition 2014;111(1):23‐45.

Hoan 2016

Hoan NX, Khuyen N, Binh MT, Giang DP, Van Tong H, Hoan PQ, et al. Association of vitamin D deficiency with hepatitis B virus‐related liver diseases. BMC Infectious Diseases 2016;16(1):507.

Holick 2007

Holick MF. Vitamin D deficiency. New England Journal of Medicine 2007;357(3):266‐81.

Holick 2011

Holick MF, Binkley NC, Bischoff‐Ferrari HA, Gordon CM, Hanley DA, Heaney RP, et al. Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology 2011;96(7):1911‐30.

Hollis 1999

Hollis S, Campbell F. What is meant by intention to treat analysis? Survey of published randomised controlled trials. BMJ 1999;319(7211):670‐4.

Hollis 2008

Hollis BW. Measuring 25‐hydroxyvitamin D in a clinical environment: challenges and needs. American Journal of Clinical Nutrition 2008;88(2):507S‐10S.

ICH‐GCP 1997

International Committee on Harmonization. Code of Federal Regulations & Guidelines. Vol. 1, Philadelphia, US: Barnett International/PAREXEL1997.

Ioannidis 2009

Ioannidis JP. Adverse events in randomized trials: neglected, restricted, distorted, and silenced. Archives of Internal Medicine 2009;169(19):1737‐9.

IOM 2011

Institute of Medicine. Dietary Reference Intakes for Calcium and Vitamin D. Washington, DC: National Academies Press, 2011.

Iruzubieta 2014

Iruzubieta P, Terán Á, Crespo J, Fábrega E. Vitamin D deficiency in chronic liver disease. World Journal of Hepatology 2014;6(12):901‐15.

Jakobsen 2014

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

Jakobsen 2017

Jakobsen JC, Nielsen EE, Feinberg J, Katakam KK, Fobian K, Hauser G, et al. Direct‐acting antivirals for chronic hepatitis C. Cochrane Database of Systematic Reviews 2017, Issue 9. [DOI: 10.1002/14651858.CD012143.pub3]

Johnson 2007

Johnson RT, Dickersin K. Publication bias against negative results from clinical trials: three of the seven deadly sins. Nature Clinical Practice. Neurology 2007;3(11):590‐1.

Keus 2010

Keus F, Wetterslev J, Gluud C, van Laarhoven CJ. Evidence at a glance: error matrix approach for overviewing available evidence. BMC Medical Research Methodology 2010;10:90.

Kitson 2012

Kitson MT, Roberts SK. D‐livering the message: the importance of vitamin D status in chronic liver disease. Journal of Hepatology 2012;57(4):897‐909.

Kitson 2014

Kitson MT, Sarrazin C, Toniutto P, Eslick GD, Roberts SK. Vitamin D level and sustained virologic response to interferon‐based antiviral therapy in chronic hepatitis C: a systematic review and meta‐analysis. Journal of Hepatology 2014;61(6):1247‐52.

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.

Konstantakis 2016

Konstantakis C, Tselekouni P, Kalafateli M, Triantos C. Vitamin D deficiency in patients with liver cirrhosis. Annals of Gastroenterology 2016;29(3):297‐306.

Kovesdy 2013

Kovesdy Csaba P, Quarles LD. Fibroblast growth factor‐23: what we know, what we don't know, and what we need to know. Nephrology Dialysis Transplantation 2013;28(9):2228‐36.

Kupferschmidt 2012

Kupferschmidt K. Uncertain verdict as vitamin D goes on trial. Science 2012;337(6101):1476‐8.

Kwok 2013

Kwok RM, Torres DM, Harrison SA. Vitamin D and nonalcoholic fatty liver disease (NAFLD): is it more than just an association. Hepatology 2013;58(3):1166‐74.

Lange 2013

Lange CM, Miki D, Ochi H, Nischalke HD, Bojunga J, Bibert S, et al. Genetic analyses reveal a role for vitamin D insufficiency in HCV‐associated hepatocellular carcinoma development. PloS One 2013;8(5):e64053.

Lesser 2007

Lesser LI, Ebbeling CB, Goozner M, Wypij D, Ludwig DS. Relationship between funding source and conclusion among nutrition‐related scientific articles. PLoS Medicine 2007;4(1):e5.

Li 2013

Li YJ, Tang YW, Shi YQ, Han S, Wang JB, Zhou XM, et al. Polymorphisms in the vitamin D receptor gene and risk of primary biliary cirrhosis: a meta‐analysis. Journal of Gastroenterology and Hepatology 2013;29(4):706‐15.

Lim 2012

Lim LY, Chalasani N. Vitamin D deficiency in patients with chronic liver disease and cirrhosis. Current Gastroenterology Reports 2012;14(1):67‐73.

Lips 2004

Lips P. Which circulating level of 25‐hydroxyvitamin D is appropriate. Journal of Steroid Biochemistry and Molecular Biology 2004;89‐90(1‐5):611‐4.

Lips 2006

Lips P. Vitamin D physiology. Progress in Biophysics and Molecular Biology 2006;92(1):4‐8.

Lips 2010

Lips P. Worldwide status of vitamin D nutrition. Journal of Steroid Biochemistry and Molecular Biology 2010;121(1‐2):297‐300.

Lucas 2005

Lucas JA, Bolland MJ, Grey AB, Ames RW, Mason BH, Horne AM, et al. Determinants of vitamin D status in older women living in a subtropical climate. Osteoporosis International 2005;16(12):1641‐8.

Lundh 2017

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

Luong 2012

Loung KV, Nguyen LT. Theoretical basis of a beneficial role for vitamin D in viral hepatitis. World Journal of Gastroenterology 2012;18(38):5338‐50.

Luong 2013a

Luong KV, Nguyen LT. The role of vitamin D in autoimmune hepatitis. Journal of Clinical Medicine Research 2013;5(6):407‐15.

Luong 2013b

Luong KV, Nguyen LT. The role of vitamin D in primary biliary cirrhosis: possible genetic and cell signaling mechanisms. Gastroenterology Research and Practice 2013;2013:602321.

Luxon 2011

Luxon BA. Bone disorders in chronic liver diseases. Current Gastroenterology Reports 2011;13(1):40‐8.

Mahamid 2013

Mahamid M, Nseir W, Abu Elhija O, Shteingart S, Mahamid A, Smamra M, et al. Normal vitamin D levels are associated with spontaneous hepatitis B surface antigen seroclearance. World Journal of Gastroenterology 2013;5(6):328‐31.

Malham 2011

Malham M, Jørgensen SP, Ott P, Agnholt J, Vilstrup H, Borre M, et al. Vitamin D deficiency in cirrhosis relates to liver dysfunction rather than aetiology. World Journal of Gastroenterology 2011;17(7):922‐5.

Moher 1998

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

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG, the PRISMA Group. Preferred reporting items for systematic reviews and meta‐analyses: the PRISMA Statement. PLoS Medicine 2009;6(7):e1000097.

Nordic Trial Alliance 2015

Nordic Trial Alliance working group on transparency and registration. Transparency and registration in clinical research in the Nordic countries. 07 Group, 2015.

Oliveira 2017

Oliveira KD, Buss C, Tovo CV. Is there an association between vitamin D and liver fibrosis in patients with chronic hepatitis C. Arquivos de Gastroenterologia 2017;54(1):57‐9.

Paternostro 2017

Paternostro R, Wagner D, Reiberger T, Mandorfer M, Schwarzer R, Ferlitsch M, et al. Low 25‐OH‐vitamin D levels reflect hepatic dysfunction and are associated with mortality in patients with liver cirrhosis. Wiener Klinische Wochenschrift 2017;129(1‐2):8‐15.

Petta 2010

Petta S, Cammà C, Scazzone C, Tripodo C, Di Marco V, Bono A, et al. Low vitamin D serum level is related to severe fibrosis and low responsiveness to interferon‐based therapy in genotype 1 chronic hepatitis C. Hepatology 2010;51(4):1158‐67.

Putz‐Bankuti 2012

Putz‐Bankuti C, Pilz S, Stojakovic T, Scharnagl H, Pieber TR, Trauner M, et al. Association of 25‐hydroxyvitamin D levels with liver dysfunction and mortality in chronic liver disease. Liver International 2012;32(5):845‐51.

Reid 2014

Reid IR, Bolland MJ, Grey A. Effects of vitamin D supplements on bone mineral density: a systematic review and meta‐analysis. Lancet 2014;383(9912):146‐55.

RevMan 2014 [Computer program]

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

Royle 2003

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

SACN 2016

Public Health England. SACN vitamin D and health report, 2016. www.gov.uk/government/publications/sacn‐vitamin‐d‐and‐health‐report (accessed 17 July 2017).

Savović 2012a

Savović J, Jones H, Altman D, Harris R, Jűni P, Pildal J, et al. Influence of reported study design characteristics on intervention effect estimates from randomised controlled trials: combined analysis of meta‐epidemiological studies. Health Technology Assessment 2012;16(35):1‐82.

Savović 2012b

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

Sayiner 2016

Sayiner M, Koenig A, Henry L, Younossi ZM. Epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in the united states and the rest of the world. Clinics in Liver Disease 2016;20(2):205‐14.

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.

Siersma 2007

Siersma V, Als‐Nielsen B, Chen W, Hilden J, Gluud LL, Gluud C. Multivariable modelling for meta‐epidemiological assessment of the association between trial quality and treatment effects estimated in randomized clinical trials. Statistics in Medicine 2007;26(14):2745‐58.

Skaaby 2014

Skaaby T, Husemoen LL, Borglykke A, Jørgensen T, Thuesen BH, Pisinger C, et al. Vitamin D status, liver enzymes, and incident liver disease and mortality: a general population study. Endocrine 2014;47(1):213‐20.

Skaaby 2016

Skaaby T, Husemoen LL, Thuesen BH, Pisinger C, Hannemann A, Jørgensen T. Longitudinal associations between lifestyle and vitamin D: a general population study with repeated vitamin D measurements. Endocrine 2016;51:342‐50.

Stokes 2013

Stokes CS, Volmer DA, Grünhage F, Lammert F. Vitamin D in chronic liver disease. Liver International 2013;33(3):338‐52.

Stokes 2014

Stokes CS, Krawczyk M, Reichel C, Lammert F, Grünhage F. Vitamin D deficiency is associated with mortality in patients with advanced liver cirrhosis. European Journal of Clinical Investigation 2014;44(2):176‐83.

Storebø 2015

Storebø OJ, Krogh HB, Ramstad E, Moreira‐Maia CR, Holmskov M, Skoog M, et al. Methylphenidate for attention‐deficit/hyperactivity disorder in children and adolescents: Cochrane systematic review with meta‐analyses and trial sequential analyses of randomised clinical trials. BMJ 2015;351:h5203.

Sweeting 2004

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

Thorlund 2009

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

Thorlund 2011a

Thorlund K, Engstrøm J, Wetterslev J, Brok J, Imberger G, Gluud C. User Manual for Trial Sequential Analysis (TSA). Copenhagen Trial Unit, Centre for Clinical Intervention Research, Copenhagen, Denmark2011.

Thorlund 2011b

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

Trépo 2013

Trépo E, Ouziel R, Pradat P, Momozawa Y, Quertinmont E, Gervy C, et al. Marked 25‐hydroxyvitamin D deficiency is associated with poor prognosis in patients with alcoholic liver disease. Journal of Hepatology 2013;59(2):344‐50.

TSA 2017 [Computer program]

Copenhagen Trial Unit. Trial Sequential Analysis, version 0.9.5.5. Copenhagen Trial Unit, 2017.

Tsiaras 2011

Tsiaras WG, Weinstock MA. Factors influencing vitamin D status. Acta Dermato‐venereologica 2011;91(2):115‐24.

Turner 2013

Turner RM, Bird SM, Higgins JP. The impact of study size on meta‐analyses: examination of underpowered studies in Cochrane reviews. PloS One 2013;8(3):e59202.

van Schoor 2011

van Schoor NM, Lips P. Worldwide vitamin D status. Best Practice & Research. Clinical Endocrinology & Metabolism 2011;25(4):671‐80.

Villar 2013

Villar LM, Del Campo JA, Ranchal I, Lampe E, Romero‐Gomez M. Association between vitamin D and hepatitis C virus infection: a meta‐analysis. World Journal of Gastroenterology 2013;19(35):5917‐24.

Wang 2013

Wang JB, Abnet CC, Chen W, Dawsey SM, Fan JH, Yin LY, et al. Association between serum 25(OH) vitamin D, incident liver cancer and chronic liver disease mortality in the Linxian Nutrition Intervention Trials: a nested case‐control study. British Journal of Cancer 2013;109(7):1997‐2004.

Wang 2015

Wang X, Li W, Zhang Y, Yang Y, Qin G. Association between vitamin D and non‐alcoholic fatty liver disease/non‐alcoholic steatohepatitis: results from a meta‐analysis. International Journal of Clinical and Experimental Medicine 2015;8(10):17221‐34.

Wesley Pike 2005

Wesley Pike J, Shevde NK. The vitamin D receptor. In: Feldman D, Wesley Pike J, Glorieux FH editor(s). Vitamin D. 2nd Edition. Amsterdam (Netherlands): Elsevier Academic Press, 2005:167‐91.

Wetterslev 2008

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

Wetterslev 2009

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

Wetterslev 2017

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

Williamson 2005

Williamson PR, Gamble C, Altman DG, Hutton JL. Outcome selection bias in meta‐analysis. Statistical Methods in Medical Research 2005;14(5):515‐24.

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 2008;336(7644):601‐5.

Younossi 2016

Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease ‐ meta‐analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016;64(1):73‐84.

Zittermann 2014

Zittermann A, Ernst JB, Gummert JF, Börgermann J. Vitamin D supplementation, body weight and human serum 25‐hydroxyvitamin D response: a systematic review. European Journal of Nutrition 2014;53(2):367‐74.

Referencias de otras versiones publicadas de esta revisión

Bjelakovic 2015

Bjelakovic G, Nikolova D, Bjelakovic M, Gluud C. Vitamin D supplementation for chronic liver diseases in adults. Vitamin D supplementation for chronic liver diseases in adults. Cochrane Database of Systematic Reviews 2015, Issue 3. [DOI: 10.1002/14651858.CD011564]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abu‐Mouch 2011

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

72 participants (44% women), aged 18 to 65 years, mean age 47 years, with chronic HCV genotype 1.

Inclusion criteria: aged 18 to 65 years; chronic HCV genotype 1 infection; no previous treatment for HCV; seronegative for HBV, HDV, and HIV infections; absolute neutrophil count > 1500/mm3; platelet count > 90,000/mm3; and normal haemoglobin level.

Exclusion criteria: decompensated liver disease (cirrhosis with a Child‐Pugh score > 9), another cause of clinically significant liver disease, or presence of hepatocellular carcinoma.

Interventions

Intervention: PEG‐IFN‐α‐2b (1.5 μg/kg body weight) + oral ribavirin 1000 mg/day (for body weight < 75 kg) or 1200 mg/day (for body weight > 75 kg) and vitamin D3 2000 IU/day (n = 36).

Control: PEG‐IFN‐α‐2b (1.5 μg/kg body weight) + oral ribavirin 1000 mg/day (for body weight < 75 kg) or 1200 mg/day (for body weight > 75 kg) (n = 36).

For 48 weeks. All participants had ≥ 1 follow‐up visit at 24 weeks after completion of treatment.

Outcomes

Outcomes reported in abstract of publication.

Primary outcome: SVR defined as undetectable HCV‐RNA at 24 weeks' post‐treatment.

Secondary outcomes: treatment efficacy at weeks 4 (RVR), and 12 (EVR) during therapy, and 24 weeks after cessation of therapy (SVR).

Stated aim of study

To determine whether adding vitamin D improves HCV response to antiviral therapy.

Notes

No participant discontinued treatment. Vitamin D3 (Vitamidyne D, Fischer Pharmaceuticals, Israel) given by oral drops for 4 weeks before initiation of antiviral treatment and after serum levels reached > 32 ng/mL in all participants in the treatment group.

Registered at clinicaltrials.gov NCT00804752.

Additional information received through personal communication with authors on 8 February 2017.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence generation performed using computer random number generation.

Allocation concealment (selection bias)

Low risk

Participant allocations could not have been foreseen in advance of, or during, enrolment. Allocation sequence hidden in sequentially numbered, opaque, and sealed envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and assessment of outcomes likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding, and assessment of outcomes likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported fully.

For‐profit bias

Unclear risk

Trial may or may not have been free of for‐profit bias as no information on clinical trial support or sponsorship provided.

Other bias

Low risk

Trial appeared free of other factors that could put it at risk of bias.

Atsukawa 2016

Methods

Open‐label randomised clinical trial with parallel group design (two groups)

Participants

Number of participants randomised: 115 patients (50% women), aged 31 to 82 years, mean age 64 years, with chronic hepatitis C.

Inclusion criteria: HCV genotype 1b as determined by the conventional polymerase chain reaction (PCR)‐based method; IL28B SNP rs8099917 genotype TG or GG (designated as non‐TT); HCV RNA persistently detectable in serum by the real‐time PCR technique; white blood cell count of more than 2000/μ; platelet count of more than 50 000/μL; and haemoglobin levels of more than 9.0 g/dL at the time of enrolment. Patients could participate in the study regardless of whether they had received prior IFN‐based therapy. When patients had not received PEG IFN/ribavirin combination therapy, they were considered as naive patients.

Exclusion criteria: Decompensated liver cirrhosis, evidence of other forms of liver disease, presence of malignancy and other serious medical illness, evidence of hypercalcaemia or hyperparathyroidism, positive hepatitis B surface antigen and antibody to HIV type 1, medication with Chinese herbal medicine or other type of vitamin D, past medical history of interstitial pneumonia, pregnancy or possibility of pregnancy, lactating, and past medical history of allergy to biological preparations or antiviral agents.

Interventions

Intervention: lead‐in treatment with oral native vitamin D3 (Healthy Natural Products, Florence, KY, USA) at a dose of 2000 IU once daily for 4 weeks, followed by the addition of the vitamin D3 to the 12‐week triple therapy (PEG IFN‐α‐2a (Roche group‐Chugai, Tokyo, Japan), ribavirin (Chugai) and simeprevir (Janssen, Tokyo, Japan)), followed by 12 weeks of PEG IFN‐ α‐2a and ribavirin (n = 57);

Control: 12‐week triple therapy (PEG IFN‐α‐2a (Roche group‐Chugai, Tokyo, Japan), ribavirin (Chugai) and simeprevir (Janssen, Tokyo, Japan)) for 12 weeks, followed by 12 weeks of PEG IFN‐ α‐2a and ribavirin (n = 58).

PEG IFN‐α‐2a was administrated subcutaneously at a dose of 180 μg once weekly. Ribavirin was administrated orally twice daily, with doses adjusted according to bodyweight (600 mg daily for <60 kg, 800 mg daily for 60–80 kg and 1000 mg daily for >80 kg). Simeprevir was administrated orally once daily at a dose of 100 mg.

Because of the low likelihood of achieving an SVR and high likelihood of developing antiviral resistance, treatment was stopped for patients with serum HCV RNA decline from baseline of less than 3 log IU/mL at 4 weeks of treatment, detectable HCV RNA at 12 weeks of treatment or more than 2 log IU/mL increase in HCV RNA levels from the lowest levels during treatment (defined as viral breakthrough).

Outcomes

Primary outcome: sustainability undetectable viraemia 24 weeks after the end of treatment.

Stated aim of study

To clarify whether native vitamin D3 supplementation could improve SVR rate in PEG‐IFN/ribavirin therapy with simeprevir for people with treatment‐refractory genotype 1b HCV with the IL28B SNP rs8099917 non‐TT.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Used computer‐generated random number table.

Allocation concealment (selection bias)

Unclear risk

Method used to conceal the allocation not described so intervention allocations may have been foreseen before, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and outcome likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No blinding, and outcome measurement likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information to assess whether missing data in combination with method used to handle missing data were likely to induce bias.

Selective reporting (reporting bias)

Unclear risk

Unclear whether all predefined and clinically relevant and reasonably expected outcomes reported.

For‐profit bias

Unclear risk

Trial may or may not be free of for‐profit bias as no information provided on clinical trial support or sponsorship.

Other bias

Low risk

Trial appeared free of other factors that could put it at risk of bias.

Barchetta 2016

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

65participants (35% women), mean age 59 years, with NAFLD.

Inclusion criteria: men or women aged 25 to 70 years; diagnosis of type 2 diabetes according to American Diabetes Association 2009 criteria; presence of fatty liver detected by upper US and confirmed by MRI in people with clinical suspicion of NAFLD (increased serum transaminase levels in absence of known hepatic chronic disease, ALT > AST, presence of multiple components of metabolic syndrome); negative tests for hepatitis B surface antigen and antibody to HCV.

Exclusion criteria: history of alcohol abuse (defined by mean daily consumption of alcohol > 30 g/day in men and > 20 g/day in women), cirrhosis, autoimmune hepatitis and other causes of liver disease (haemochromatosis, Wilson's disease), chronic enteropathies, advanced renal failure, cancer, hyper/hypoparathyroidism, known hypersensitivity to cholecalciferol or any other excipients, hypercalcaemia, hypercalciuria, nephrolithiasis, nephrocalcinosis; ongoing/recent (previous 6 months) supplementation with vitamin D, calcium, multivitamin products; treatment with agents affecting bone and calcium/vitamin D metabolism (anticonvulsants, glucocorticoids, antacids containing aluminium, cholestyramine); ultraviolet radiation exposure; pregnancy and lactation; or severe psychiatric illnesses.

Interventions

Intervention: vitamin D3 2000 IU/day (n = 29).

Control: placebo (n = 36).

For 24 weeks.

Outcomes

Primary outcomes: reduction of hepatic fat fraction measured by MRI, changes in serum transaminases, CK18‐M30, N‐terminal procollagen III propeptide levels, and Fatty Liver Index.

Secondary outcomes: metabolic (fasting glycaemia, haemoglobin A1c, lipids, Homeostasis Model Assessment ‐ Insulin Resistance, Homeostasis Model Assessment ‐ beta cell function, adipose tissue insulin resistance, body fat distribution) and cardiovascular (ankle‐brachial index, intima‐media thickness, flow‐mediated dilatation) parameters.

Stated aim of study

To assess the efficacy and safety of 24‐week oral high‐dose vitamin D supplementation in people with type 2 diabetes and NAFLD.

Notes

Registered at www.clinicaltrialsregister.eu (number 2011‐003010‐17). Funded by research grants from the Sapienza University Ateneo Scientific Research (authors MGC and IB) and the Italian Minister of University and Research (authors MGC and MGB).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation performed by statistician using computer‐generated and centrally administered procedure.

Allocation concealment (selection bias)

Low risk

Participant allocations could not have been foreseen in advance of, or during, enrolment. Used central and independent randomisation unit controlled allocation.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants, investigators, clinical site staff, laboratory staff, and radiologists all masked to treatment assignment throughout study. Treatment and placebo provided in identical vials by an experienced independent pharmacist.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and unlikely that blinding could have been broken.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information to assess whether missing data in combination with method used to handle missing data were likely to introduce bias on the results.

Selective reporting (reporting bias)

Low risk

Study authors reported all predefined outcomes fully.

For‐profit bias

Low risk

Trial appeared free of industry sponsorship or other type of for‐profit support that could manipulate trial design, conductance, or trial results.

Other bias

Low risk

Trial appeared free of other factors that could put it at risk of bias.

Esmat 2015

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

101 participants (25% women) aged 18 to 60 years, mean age 40 years, with chronic HCV genotype 4.

Inclusion criteria: aged 18 to 60 years, chronic HCV infection genotype 4 for > 6 months by detectable serum quantitative HCV‐RNA, naive to treatment, compensated liver disease with the following minimum haematological and biochemical criteria (haemoglobin ≥ 12 g/dL for men and ≥ 11 g/dL for women, WBC > 3500/mm3, granulocyte count > 1500/mm3, platelet count > 75,000/mm3, albumin and thyroid function tests within normal limit, and antinuclear antibody ≤ 1:80). US‐guided liver biopsy within 12 months prior to study entry, using a semiautomatic true‐cut needle (16G).

Exclusion criteria: other liver diseases, decompensated liver cirrhosis, hepatocellular carcinoma, liver biopsy contraindication, unsuitable for combined IFN and ribavirin treatment due to persistent haematological abnormalities, receiving medications known to affect vitamin D3 level or metabolism (calcium, vitamin D supplementation, oestrogen, alendronate, isoniazid, thiazide diuretics, long‐term antacids, calcium channel blockers, cholestyramine, anticonvulsants, and orlistat), clinically evident osteomalacia (waddling gait, bone pain, and pathological fractures), renal diseases or parathyroid diseases, and BMI > 35.

Interventions

Intervention: vitamin D3 15,000 IU/week + PEG‐IFN‐α‐2b + ribavirin (n = 50).

Control: placebo + PEG‐IFN‐α‐2b + ribavirin (n = 51).

PEG‐IFN‐α‐2b (Peg‐Intron‐MSD) at 1.5 mg/kg subcutaneous injection once/week. Ribavirin (Rebetol, MSD) dose determined by body weight (< 75 kg = 1000 mg/day; ≥ 75 kg = 1200 mg/day in 2 separate oral doses after meals morning and night) for 48 weeks. Vitamin D3 given as oral solution with juice once weekly for 48 weeks.

Outcomes

Primary outcome: SVR.

Secondary outcome: stage of hepatic fibrosis.

Stated aim of study

To assess role of vitamin D supplementation on response to treatment in people with chronic HCV 4 and its possible relation to stage of hepatic fibrosis.

Notes

Additional information received through personal communication with authors on 23 January 2017.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of sequence generation not specified.

Allocation concealment (selection bias)

Low risk

Allocation sequence hidden in sequentially numbered, opaque, and sealed envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

No blinding, but we judged that outcomes were not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

No blinding of outcome assessment, but we judged that outcome measurements were not likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information to assess whether missing data in combination with the method used to handle missing data were likely to induce bias.

Selective reporting (reporting bias)

Unclear risk

Unclear whether all predefined and clinically relevant and reasonably expected outcomes reported.

For‐profit bias

Unclear risk

Trial may or may not have been free of for‐profit bias as trial did not provide any information on clinical trial support or sponsorship.

Other bias

Low risk

Trial appeared free of other factors that could put it at risk of bias.

Foroughi 2016

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

60 participants (52% women), aged 30 to 70 years, mean age 48.5 years with NAFLD.

Inclusion criteria: NAFLD confirmed by US and normal range of ALT and AST (< 31 IU/L).

Exclusion criteria: acute illnesses, chronic kidney disease, hyperparathyroidism, hypoparathyroidism, chronic heart failure, HCV or HBV, Wilson's syndrome, history of chronic liver diseases or disorders that affect gallbladder and bile ducts, pregnancy, history of taking any drugs affecting levels of ALT (e.g. valproic acid, tamoxifen, 3‐hydroxy‐3‐methylglutaryl‐coenzyme A reductase inhibitors, metformin, angiotensin converting enzyme 1 and angiotensin‐converting enzyme‐related 1). Furthermore, participants should not have followed any special diet, and not take oral vitamin D, calcium, or multivitamin supplements.

Interventions

Intervention: vitamin D3 50,000 IU (n = 30).

Control: placebo (n = 30).

Weekly for 10 weeks.

Outcomes

Primary outcomes: inflammatory markers, liver function, lipid profile, body composition, and liver steatosis.

Secondary outcomes: none stated.

Stated aim of study

To investigate effect of vitamin D supplementation on inflammation, liver function, and liver steatosis in people with NAFLD.

Notes

Clinical trial registered at Iranian Registry of Clinical Trials (www.irct.ir) IRCT number: IRCT2013060411763N8. Funded by Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Used computer‐generated random numbers.

Allocation concealment (selection bias)

Unclear risk

Method used to conceal the allocation not described so intervention allocations may have been foreseen before, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of 'low risk' or 'high risk'.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of 'low risk' or 'high risk'.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported fully.

For‐profit bias

Low risk

Trial appeared to be free of industry sponsorship or other type of for‐profit support that could manipulate the trial design, conductance, or trial results.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias.

Lorvand Amiri 2016

Methods

Randomised clinical trial with parallel group design (3 groups).

Participants

120 participants (38% women), aged 18 to 65 years, mean age 41 years, with NAFLD.

Inclusion criteria: BMI 25 kg/m2 to 35 kg/m2, serum 25‐hydroxyvitamin D3 level < 15 ng/mL, reporting a daily calcium intake 700 mg/day to 800 mg/day, and willingness to introduce a dietary change to lose weight.

Exclusion criteria: calcium intake < 700 mg/day or > 800 mg/day (in diet or as a supplement); drugs for blood glucose or lipid control; pregnancy or having given birth in the past year or planning a pregnancy in the next 6 months; lactation; weight loss ≥ 10% of body weight within the 6 months before enrolment; participation in competitive sport; abnormal thyroid hormone concentration; intake of medications that could affect body weight or energy expenditure (or both); allergy; smoking; diagnosis of chronic diseases including inflammatory diseases; heart, liver, and renal failure; cancer; acute myocardial infarction; diabetes; stroke; or serious injuries and any other conditions that were not suitable for the trial as evaluated by the physician.

Interventions

Intervention 1: vitamin D 25 μg/day as calcitriol (Jalinus Arya Co., Iran) + calcium carbonate placebo (25 mg/day as lactose; Jalinus Arya Co, Iran) (n = 37).

Intervention 2: vitamin D 25 μg/day as calcitriol (Jalinus Arya Co., Iran) + calcium (500 mg/day as calcium carbonate; Jalinus Arya Co., Iran) (n = 37).

Control: placebo of calcitriol + placebo of calcium (25 mg/day as lactose; Jalinus Arya Co., Iran) (n = 36).

After lunch with a glass of water for 12 weeks.

Outcomes

Primary outcomes: weight loss, body fat, fasting plasma glucose, serum insulin concentrations, lipid profiles, and liver function tests.

Secondary outcomes: carbohydrate and lipid metabolism.

Stated aim of study

To compare effect of vitamin D supplementation with and without calcium on anthropometric measures and biochemical parameters in people with NAFLD during a weight‐loss programme.

Notes

Clinical trial registered at Iranian Registry of Clinical Trials (www.irct.ir) IRCT registration number: IRCT201408312709N29. Trial did not receive any specific grant from funding agencies in the public, commercial, or not‐for‐profit sectors. Additional information received through personal communication with authors on 20 January 2017.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants randomly assigned using computer‐generated random‐numbers method by project co‐ordinator.

Allocation concealment (selection bias)

Low risk

Participant allocations could not have been foreseen in advance of, or during, enrolment. Allocation controlled by a central and independent randomisation unit.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Products administered by blinded research assistant to blinded participants. Shape, colour, and packaging of placebo similar to supplements in the intervention group.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to assess whether missing data in combination with method used to handle missing data were likely to induce bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported fully.

For‐profit bias

Unclear risk

Trial may or may not have been free of for‐profit bias as no information provided on clinical trial support or sponsorship.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias.

Mobarhan 1984

Methods

Randomised clinical trial with parallel group design (3 groups).

Participants

18 men, aged 32 to 61 years, mean age 52 years, with alcoholic cirrhosis.

Inclusion criteria: men with advanced biopsy‐confirmed alcoholic cirrhosis with low levels of serum 25‐hydroxyvitamin D (< 20 ng/mL) and decreased bone density (i.e. > 1.5 standard deviations below mean of healthy Baltimore men of same ages).

Exclusion criteria: history of corticosteroid, anticonvulsant, or vitamin D intake; renal disease.

Interventions

Intervention 1: vitamin D2 50,000 IU 2 or 3 times weekly (n = 6).

Intervention 2: 25‐hydroxyvitamin D3 800 IU/day to 2000 IU/day (prepared and supplied as identical soft elastic capsules (20 or 50 μg) by Upjohn Co.) (n = 6).

Control: no intervention (n = 6).

For 1 year.

Outcomes

Outcomes reported in abstract of publication.

Primary outcomes: bone mineral density.

Secondary outcomes: none stated.

Stated aim of study

To compare the efficacy of 25‐hydroxyvitamin D3 or vitamin D2 in correcting the bone disease of people with alcoholic cirrhosis.

Notes

This study was supported by grants from Upjohn Co. and the Veterans Administration.

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 used to conceal the allocation was not described so that intervention allocations may have been foreseen in advance of, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and the outcome was likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of outcome assessment, and the outcome measurement is likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Low risk

All clinically relevant and reasonably expected outcomes were reported.

For‐profit bias

High risk

The trial is sponsored by the industry.

Other bias

Low risk

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

Nimer 2012

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

50 participants (58% women), mean age 47 years, with chronic HCV genotype 2 or 3.

Inclusion criteria: aged 18 to 65 years; chronic genotype 2 or 3 HCV infection; no previous treatment for HCV; seronegative for HBV, hepatitis A virus, and HIV infection; absolute neutrophil count > 1500/mm3; platelet count > 90,000/mm3; and normal haemoglobin level. Liver biopsies not required prior to study entrance.

Exclusion criteria: decompensated liver disease (cirrhosis with Child‐Pugh score > 9), another cause of clinically significant liver disease, or presence of hepatocellular carcinoma.

Interventions

Intervention: PEG‐IFN‐α‐2a 180 μg weekly + oral ribavirin 800 mg/day + oral vitamin D3 2000 IU/day (Vitamidyne D, Fischer Pharmaceuticals, Israel), given by oral drops (n = 20).

Control: PEG‐IFN‐α‐2a 180 μg weekly + oral ribavirin 800 mg/day (n = 30).

For 24 weeks.

Outcomes

Outcomes reported in abstract of publication.

Primary outcome: SVR defined as undetectable HCV‐RNA at 24 weeks' post‐treatment.

Secondary outcomes: treatment efficacy at weeks 4 (RVR), and 12 (EVR) during therapy, and 24 weeks after cessation of therapy (SVR).

Stated aim of study

To assess prospectively influence of vitamin D supplementation on SVR in treatment of people with chronic HCV with HCV genotype 2‐3.

Notes

Additional information received through personal communication with authors on 8 February 2017.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence generation performed using computer random number generation.

Allocation concealment (selection bias)

Low risk

Participant allocations could not have been foreseen in advance of, or during, enrolment. Allocation sequence hidden in sequentially numbered, opaque, and sealed envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and outcomes were likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding, and outcome measurements were likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Unclear risk

Unclear whether all predefined and clinically relevant and reasonably expected outcomes were reported.

For‐profit bias

Unclear risk

Trial may or may not have been free of for‐profit bias as no information on clinical trial support or sponsorship was provided.

Other bias

Low risk

Trial appeared to be free of other factors that could put it at risk of bias.

Pilz 2016

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

36 participants (25% women), aged 18 to 75 years, mean age 61 years, with liver cirrhosis.

Inclusion criteria: compensated cirrhosis, 25‐hydroxyvitamin D < 30 ng/mL, aged 18 to 75 years, and a negative pregnancy test in women of childbearing potential.

Exclusion criteria: presence of hepatocellular carcinoma, hypercalcaemia (plasma calcium concentrations > 2.65 mmol/L), pregnant or lactating women, drug intake as part of another clinical study, estimated glomerular filtration rate according to Modification of Diet in Renal Disease formula < 15 mL/minute/1.73 m2, any clinically significant acute disease requiring drug treatment, regular intake (in addition to study medication) of vitamin D > 800 IU daily during the last 4 weeks before study entry.

Interventions

Intervention: vitamin D3 2800 IU/day (Oleovit D3, Fresenius Kabi, Austria) (n = 18).

Control: placebo daily (n = 18).

For 8 weeks.

Outcomes

Primary outcome: vitamin D status.

Secondary outcomes: liver function tests (i.e. AST, ALT, gamma glutamyl transpeptidase, and alkaline phosphatase), albumin, International Normalized Ratio, bilirubin, and hyaluronic acid; and parameters of mineral metabolism (i.e. parathyroid hormone, total plasma calcium, free plasma calcium, urinary midstream calcium to creatinine ratio, and plasma phosphate).

Stated aim of study

To evaluate effects of vitamin D supplementation on 25‐hydroxyvitamin D, parameters of liver function and synthesis, and hyaluronic acid as a marker of liver fibrosis.

Notes

Study sponsored by the Medical University of Graz, Austria.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence generation performed using computer random number generation.

Allocation concealment (selection bias)

Low risk

Participant allocations could not have been foreseen in advance of, or during, enrolment. Central and independent randomisation unit controlled allocation. Investigators were unaware of allocation sequence.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured, and it was unlikely that blinding could have been broken.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and unlikely that blinding could have been broken.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported.

For‐profit bias

Low risk

Trial appeared to be free of industry sponsorship or other type of for‐profit support that could manipulate the trial design, conductance, or trial results.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias.

Sharifi 2014

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

60 (51% women), aged 18 to 70 years, mean age 42 years, with NAFLD.

Inclusion criteria: diagnosis of NAFLD by US and increased serum levels of ALT (> 19 U/L for women and 30 U/L for men).

Exclusion criteria: alcohol consumption > 20 g/day; pregnant and lactating women; hereditary haemochromatosis; Wilson's disease; α1‐antitrypsin deficiency; history of jejunoileal bypass surgery or gastroplasty; using total parenteral nutrition in the past 6 months; taking hepatotoxic drugs such as calcium channel blocker, high doses of synthetic oestrogens, methotrexate, amiodarone, and chloroquine; history of hypothyroidism, Cushing's syndrome, renal failure, and kidney stones; serum calcium levels > 10.6 mg/dL; and intake of vitamin D, vitamin E, and calcium supplements during the last 6 months.

Interventions

Intervention: vitamin D3 50,000 IU (D‐Vitin Zahravi Pharm Co., Tabriz, Iran) (n = 30).

Control: placebo (Zahravi Pharm Co.) (n = 30).

Every 14 days for 4 months.

Outcomes

Primary outcomes: changes in serum ALT and changes in insulin resistance index.

Secondary outcomes: other liver enzymes, oxidative stress, and inflammatory biomarkers.

Stated aim of study

To determine effect of vitamin D supplementation on serum liver enzymes, insulin resistance, oxidative stress, and inflammatory biomarkers in people with NAFLD.

Notes

Study financially supported by grant (No. RDC‐9105) from Vice‐Chancellor for Research Affairs of Jundishapur University of Medical Sciences and approved by the Research Institute for Infectious Diseases of the Digestive System, Jundishapur University of Medical Sciences, Ahvaz, Iran.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The study authors performed sequence generation using computer random number generation

Allocation concealment (selection bias)

Low risk

An investigator with no clinical involvement in the trial packed the supplements and placebos in numbered bottles based on the random list. The other person, who was not involved in the trial and not aware of random sequences, assigned the patients to the numbered bottles of pearls.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured, and it was unlikely that the blinding could have been broken.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and unlikely that the blinding could have been broken.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Unclear risk

It is unclear whether all predefined and clinically relevant and reasonably expected outcomes were reported.

For‐profit bias

Low risk

The trial appeared to be free of industry sponsorship or other type of for‐profit support that could manipulate the trial design, conductance, or trial results.

Other bias

Low risk

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

Shiomi 1999a

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

76 participants (66% women), aged 38 to 84 years, mean age 61 years, with cirrhosis and an underlying infection of liver (HBV and HCV).

Inclusion criteria: liver cirrhosis and an underlying infection of the liver (HBV and HCV).

Exclusion criteria: none stated.

Interventions

Intervention: calcitriol 0.5 μg twice daily (n = 38).

Control: no intervention (n = 38).

For 1 year.

Outcomes

Outcomes reported in abstract of publication.

Primary outcome: bone mineral density of the lumbar vertebrae.

Secondary outcomes: none stated.

Stated aim of study

To evaluate efficacy of calcitriol (1,25‐dihydroxyvitamin D) in treatment of bone disease associated with cirrhosis and an underlying hepatitis viral infection.

Notes

Additional information received through personal communication with the authors on 12 February 2014.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of sequence generation not specified.

Allocation concealment (selection bias)

Low risk

Allocation sequence hidden in sequentially numbered, opaque, and sealed envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and outcome was likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of outcome assessment, and outcome measurement was likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

High risk

Not all predefined outcomes reported fully.

For‐profit bias

Unclear risk

Trial may or may not have been free of for‐profit bias as no information on clinical trial support or sponsorship was provided.

Other bias

Unclear risk

Trial may or may not have been free of other components that could put it at risk of bias.

Shiomi 1999b

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

34 women, aged 36 to 72 years, mean age 56 years, with primary biliary cirrhosis.

Inclusion criteria: primary biliary cirrhosis.

Exclusion criteria: none stated.

Interventions

Intervention: calcitriol 0.5 μg twice a day (n = 17).

Control: no intervention (n = 17).

For 1 year.

Outcomes

Outcomes reported in abstract of publication.

Primary outcome: bone mineral density.

Secondary outcomes: none stated.

Stated aim of study

To evaluate efficacy of calcitriol (1,25‐dihydroxyvitamin D) in treatment of bone disease associated with primary biliary cirrhosis.

Notes

Additional information received through personal communication with authors on 12 February 2014.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of sequence generation not specified.

Allocation concealment (selection bias)

Unclear risk

Method used to conceal allocation not described so that intervention allocations may have been foreseen in advance of, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and outcome was likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of outcome assessment, and outcome measurement was likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

High risk

Not all predefined outcomes reported fully.

For‐profit bias

Unclear risk

Trial may or may not have been free of for‐profit bias as no information on clinical trial support or sponsorship was provided.

Other bias

Unclear risk

Trial may or may not have been free of other components that could put it at risk of bias.

Vosoghinia 2016

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

68 participants (13% women), mean age 42 years, with chronic HCV genotype 1,2,3,4.

Inclusion criteria: adult patients with chronic HCV infection (> 6 months) and detectable serum levels of HCV RNA (genotype 1, 2, 3 or 4) with compensated liver disease fulfilling the following criteria of an absolute neutrophil count above 1500 permm3, a platelet count above 90,000 permm3, and a normal haemoglobin level.

Exclusion criteria: co‐infection with hepatitis B virus or HIV, decompensated liver disease (Child‐Pugh classification B or C), autoimmune or metabolic liver disease, hepatocellular carcinoma, a history of anti‐HCV therapy or use of medications which alter vitamin D3 levels or metabolism (calcium, vitamin D supplementation, oestrogen, alendronate, isoniazid, anticonvulsants, and orlistat), or a history of diarrhoea or malabsorption syndromes like celiac and chronic pancreatitis or those with renal or parathyroid diseases.

Interventions

Intervention: PEG‐IFN‐α‐2a (180 μg) + oral ribavirin (Rebetol, MSD) dosage determined based on patient’s weight and genotype, was administered for 48 weeks in patients with genotypes 1 and 4 and for 24 weeks in those with genotypes 2 and 3, and vitamin D3 1600 IU/day (n = 34).

Control: PEG‐IFN‐α‐2a (180 μg) + oral ribavirin (Rebetol, MSD), dosage determined based on patient’s weight and genotype.

PEG‐IFN‐α‐2a was administered for 48 weeks in patients with genotypes 1 and 4 and for 24 weeks in those with genotypes 2 and 3 (n = 34).

Vitamin D3 was administered for 12 weeks.

Outcomes

Primary outcome: EVR defined as undetectable HCV‐RNA at 12 weeks' post‐treatment.

Stated aim of study

To assess the influence of vitamin D supplementation on viral response to PegINF/RBV therapy

Notes

The research council of Mashhad University of Medical Sciences,Mashhad, Iran financially supported this study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of sequence generation not specified.

Allocation concealment (selection bias)

Low risk

Participant allocations could not have been foreseen in advance of, or during, enrolment. Allocation sequence hidden in sequentially numbered, opaque, and sealed envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and assessment of outcomes likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding, and assessment of outcomes likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Low risk

All predefined outcomes reported fully.

For‐profit bias

Low risk

The trial appeared to be free of industry sponsorship or other type of for‐profit support that could manipulate the trial design, conductance, or trial results.

Other bias

Unclear risk

The trial may or may not have been free of other components that could put it at risk of bias.

Xing 2013

Methods

Randomised clinical trial with parallel group design (3 groups).

Participants

75 participants (17% women), aged 28 to 65 years, mean age 48 years, undergoing liver transplantation.

Inclusion criteria: primary liver transplant recipients.

Exclusion criteria: history of corticosteroid, anticonvulsant, or vitamin D intake; renal disease.

Interventions

Intervention 1: calcitriol 0.25 μg/day + calcium gluconate (n = 25).

Intervention 2: calcium gluconate (n = 25).

Control: placebo (n = 25).

For 1 month.

Outcomes

Outcomes reported in abstract of publication:

Primary outcomes: acute cellular rejection rate at 1 month' post transplant.

Secondary outcomes: none stated.

Stated aim of study

To investigate effects of calcitriol on acute cellular rejection rate of liver transplant recipients.

Notes

Study sponsored by a grant from Shanghai Nature Science Fund project and a grant from Science and Technology Department of Shanghai. Additional information received through personal communication with the authors on 13 February 2014.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of sequence generation not specified.

Allocation concealment (selection bias)

Unclear risk

Method used to conceal allocation not described so that intervention allocations may have been foreseen in advance of, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of 'low risk' or 'high risk'.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgement of 'low risk' or 'high risk'.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Unclear risk

Unclear whether all predefined and clinically relevant and reasonably expected outcomes were reported.

For‐profit bias

Low risk

Trial appeared to be free of industry sponsorship or other type of for‐profit support that could manipulate the trial design, conductance, or trial results.

Other bias

Low risk

Trial appeared to be free of other factors that could put it at risk of bias.

Yokoyama 2014

Methods

Randomised clinical trial with parallel group design (2 groups).

Participants

84 participants (49% women), aged 30 to 78 years, mean age 59 years, with HCV genotype 1b.

Inclusion criteria: aged ≥ 20 years, chronically infected with HCV genotype 1 and plasma HCV RNA concentrations ≥ 100 log IU/mL.

Exclusion criteria: decompensated cirrhosis, liver cancer, HBV or HIV infection, renal insufficiency, history of heart disease or cerebral infarction, pregnancy or breastfeeding.

Interventions

Intervention: subcutaneous injections of PEG‐IFN‐α‐2b (1.5 μg/kg body weight) once weekly, along with weight‐based oral ribavirin (600 mg/day to 1200 mg/day) + vitamin D3 1000 IU (n = 42).

Control: subcutaneous injections of PEG‐IFN‐α‐2b (1.5 μg/kg body weight) once weekly, along with weight‐based oral ribavirin (600 mg/day to 1200 mg/day) (n = 42).

For 16 weeks.

Outcomes

Primary outcome: undetectable HCV RNA at week 24.

Secondary outcomes: none stated.

Stated aim of study

To rigorously evaluate the antiviral effects of vitamin D supplementation in people with HCV genotype‐1 infection being treated with PEG‐IFN + ribavirin.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of sequence generation not specified.

Allocation concealment (selection bias)

Unclear risk

Method used to conceal the allocation not described so intervention allocations may have been foreseen before, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and outcome was likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of outcome assessment, and outcome measurement was likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing data were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Unclear risk

Unclear whether all predefined and clinically relevant and reasonably expected outcomes were reported.

For‐profit bias

Unclear risk

Trial may or may not have been free of for‐profit bias as trial did not provide any information on clinical trial support or sponsorship.

Other bias

Low risk

Trial appeared to be free of other factors that could put it at risk of bias.

ALT: alanine aminotransferase; AST: aspartate aminotransferase; BMI: body mass index; EVR: early viral response; HBV: hepatitis B virus; HCV: hepatitis C virus; HDV: hepatitis D virus; IFN: interferon; IU: international unit; MRI: magnetic resonance imaging; n: number of participants; NAFLD: non‐alcoholic fatty liver disease; PCR: polymerase chain reaction; PEG: pegylated; RNA: ribonucleic acid; RVR: rapid viral response; SVR: sustained virological response; US: ultrasound; WBC: white blood cell count.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Atsukawa 2013

Not a randomised trial.

Benetti 2008

Not a randomised trial.

Bitetto 2010

Not a randomised trial.

Fernández Fernández 2016

Not a randomised trial.

Floreani 2007

Not a randomised trial.

Kitson 2016

Not a randomised trial.

Kondo 2013

Not a randomised trial.

Ladero 2013

Not a randomised trial.

Long 1978

Not a randomised trial.

Malham 2012

Not a randomised trial.

Papapostoli 2016

Not a randomised trial.

Rode 2010

Not a randomised trial.

Stokes 2016

Not a randomised trial.

Terrier 2015

Not a randomised trial.

Characteristics of ongoing studies [ordered by study ID]

IRCT2016020326342N1

Trial name or title

Effectiveness of Vitamin D Supplementation on Severity of Cirrhosis Based on CHILD and MELD Scores in Patients with Decompensate Cirrhosis.

Methods

Randomised clinical trial using parallel group design (2 groups).

Participants

Country: Iran.

Estimated number of participants: 80.

Inclusion criteria: people with HIV, renal failure due to reasons other than liver failure, malabsorption such as chronic diarrhoea, coeliac disease, chronic pancreatitis; people undergoing corticosteroid treatment; pregnancy; and people with cirrhosis secondary to cholestasis such as primary biliary cirrhosis.

Interventions

Intervention: vitamin D3 (50,000 IU) and popular drugs using for liver cirrhosis.

Control: popular drugs using for liver cirrhosis.

Daily for 3 months.

Outcomes

Primary outcome: liver function measured by Model for End‐Stage Liver Disease score.

Secondary outcomes: liver function measured by Child‐Turcotte‐Pugh score.

Starting date

March 2016.

Contact information

Hossein Ali Abbasi, Emam Reza Hospital, Emam Reza Square, Ebne Sina Avenue, Mashhad, Iran, [email protected].

Notes

NCT02779465

Trial name or title

Study of Oral Vitamin D Treatment for the Prevention of Hepatocellular Carcinoma in Patients with Chronic Hepatitis B.

Methods

Randomised clinical trial using parallel group design (2 groups).

Participants

Country: China.

Estimated number of participants: 1500.

Inclusion criteria: age 18 to 70 years; with chronic hepatitis B and under the oral antivirus treatment; no evidence of hepatocellular carcinoma on entry imaging study; Model for End‐Stage Liver Disease score < 22; not currently participating in another intervention study; not pregnant or lactating; and willing to use effective contraception during study period; absence of any psychological, familial, sociological, or geographical condition potentially hampering compliance with the study protocol and follow‐up schedule; and ability to provide written informed consent according to national or local regulations.

Exclusion criteria: evidence of hepatocellular carcinoma within 6 months after enrolment; serum alanine aminotransferase level > 10 times the upper limit of normal, elevated serum creatinine level, diagnosis of kidney stones, diagnosis of hyperparathyroidism or other serious disturbance of calcium metabolism in past 5 years, evidence of autoimmune hepatitis, coinfection with hepatitis C or D virus or HIV, other serious concurrent illness (e.g. alcoholism, uncontrolled diabetes, or cancer), treatment with immunomodulatory agent within 6 months before screening, treatment with any investigational drug within 30 days before the study began.

Interventions

Intervention: vitamin D3 800 IU/day besides the antivirus treatment with nucleos(t)ide medicine.

Control: no intervention.

For 1 year.

Outcomes

Primary outcomes: change in serum levels of 25‐hydroxyvitamin D at baseline, and at 6 and 12 months, and change in serum levels of 25‐hydroxyvitamin D at 6 and 12 months compared to baseline.

Secondary outcomes: change in serum creatinine at baseline, and at 6 and 12 months; change in serum creatinine at 6 and 12 months compared to baseline; change in fibrosis score at baseline, and at 6 and 12 months; fibrosis score at 6 and 12 months compared to baseline; number of participants on vitamin D treatment with adverse events.

Starting date

June 2016.

Contact information

Yutian Chong, MD, Third Affiliated Hospital, Sun Yat‐Sen University, [email protected].

Notes

IU: international unit.

Data and analyses

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

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

15

1034

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.70 [0.09, 5.38]

Analysis 1.1

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 1 All‐cause mortality.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 1 All‐cause mortality.

2 All‐cause mortality ('best‐worst' case and 'worst‐best' case scenarios) Show forest plot

15

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 2 All‐cause mortality ('best‐worst' case and 'worst‐best' case scenarios).

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 2 All‐cause mortality ('best‐worst' case and 'worst‐best' case scenarios).

2.1 'Best‐worst' case scenario

15

1034

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.11 [0.05, 0.24]

2.2 'Worst‐best' case scenario

15

1034

Peto Odds Ratio (Peto, Fixed, 95% CI)

7.80 [3.67, 16.57]

3 Liver‐related mortality Show forest plot

1

18

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.08, 34.66]

Analysis 1.3

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 3 Liver‐related mortality.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 3 Liver‐related mortality.

4 Serious adverse events Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 4 Serious adverse events.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 4 Serious adverse events.

4.1 Hypercalcaemia

1

76

Risk Ratio (M‐H, Random, 95% CI)

5.0 [0.25, 100.80]

4.2 Myocardial infarction

2

86

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.08, 6.81]

4.3 Thyroiditis

1

68

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.91]

5 Non‐serious adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 5 Non‐serious adverse events.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 5 Non‐serious adverse events.

5.1 Glossitis

1

65

Risk Ratio (M‐H, Random, 95% CI)

3.70 [0.16, 87.58]

6 Failure of rapid virological response Show forest plot

2

187

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.52, 0.94]

Analysis 1.6

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 6 Failure of rapid virological response.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 6 Failure of rapid virological response.

7 Failure of early virological response Show forest plot

2

140

Risk Ratio (M‐H, Random, 95% CI)

0.10 [0.03, 0.33]

Analysis 1.7

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 7 Failure of early virological response.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 7 Failure of early virological response.

8 Failure of sustained virological response Show forest plot

5

422

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.28, 1.21]

Analysis 1.8

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 8 Failure of sustained virological response.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 8 Failure of sustained virological response.

9 Acute cellular rejection in liver transplant recipients Show forest plot

1

75

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 2.62]

Analysis 1.9

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 9 Acute cellular rejection in liver transplant recipients.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 9 Acute cellular rejection in liver transplant recipients.

10 Vitamin D status (ng/mL) Show forest plot

6

424

Mean Difference (IV, Random, 95% CI)

17.24 [12.46, 22.02]

Analysis 1.10

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 10 Vitamin D status (ng/mL).

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 10 Vitamin D status (ng/mL).

11 Biochemical indices Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.11

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 11 Biochemical indices.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 11 Biochemical indices.

11.1 Aspartate aminotransferase (IU/L)

6

313

Mean Difference (IV, Random, 95% CI)

‐1.40 [‐2.88, 0.08]

11.2 Alanine aminotransferase (IU/L)

6

313

Mean Difference (IV, Random, 95% CI)

‐0.52 [‐5.10, 4.06]

11.3 Alkaline phosphatases (IU/L)

2

96

Mean Difference (IV, Random, 95% CI)

7.39 [‐39.89, 54.67]

11.4 Gamma‐glutamyl transpeptidase (IU/L)

2

101

Mean Difference (IV, Random, 95% CI)

3.64 [0.33, 6.96]

11.5 Albumin (g/L)

2

48

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.40, 0.20]

11.6 Bilirubin (mg/dL)

2

48

Mean Difference (IV, Random, 95% CI)

0.38 [0.21, 0.55]

11.7 Triglyceride (mg/dL)

2

115

Mean Difference (IV, Random, 95% CI)

23.69 [‐13.90, 61.27]

11.8 Cholesterol (mg/dL)

1

55

Mean Difference (IV, Random, 95% CI)

2.75 [‐4.75, 10.25]

11.9 Calcium (mg/dL)

2

72

Mean Difference (IV, Random, 95% CI)

2.01 [‐0.53, 4.56]

Study flow diagram
Figuras y tablas -
Figure 1

Study flow diagram

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

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

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

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

Funnel plot of comparison: 1 Vitamin D versus placebo or no intervention, outcome: 1.1 All‐cause mortality.
Figuras y tablas -
Figure 4

Funnel plot of comparison: 1 Vitamin D versus placebo or no intervention, outcome: 1.1 All‐cause mortality.

Trial Sequential Analysis on all‐cause mortality up to 1.4‐year follow‐up in 15 vitamin D trials, based on mortality rate in the control group of 10%, a relative risk reduction of 28% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 6396 participants. The cumulative Z‐curve (blue line) did not cross the trial sequential monitoring boundary for benefit or harm (red inward sloping lines) and did not enter the trial sequential monitoring area for futility (inner‐wedge with red outward sloping lines).
Figuras y tablas -
Figure 5

Trial Sequential Analysis on all‐cause mortality up to 1.4‐year follow‐up in 15 vitamin D trials, based on mortality rate in the control group of 10%, a relative risk reduction of 28% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 6396 participants. The cumulative Z‐curve (blue line) did not cross the trial sequential monitoring boundary for benefit or harm (red inward sloping lines) and did not enter the trial sequential monitoring area for futility (inner‐wedge with red outward sloping lines).

Trial Sequential Analysis on rapid virological response in the two vitamin D trials was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 11958 participants. The cumulative Z‐curve crossed the conventional monitoring boundary for benefit. The trial sequential monitoring boundary is ignored due to little information use (1.56%).
Figuras y tablas -
Figure 6

Trial Sequential Analysis on rapid virological response in the two vitamin D trials was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 11958 participants. The cumulative Z‐curve crossed the conventional monitoring boundary for benefit. The trial sequential monitoring boundary is ignored due to little information use (1.56%).

Trial Sequential Analysis on early virological response in the two vitamin D trials was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 11958 participants. The cumulative Z‐curve crossed the conventional monitoring boundary for benefit. The trial sequential monitoring boundary is ignored due to little information use (1.17%).
Figuras y tablas -
Figure 7

Trial Sequential Analysis on early virological response in the two vitamin D trials was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 11958 participants. The cumulative Z‐curve crossed the conventional monitoring boundary for benefit. The trial sequential monitoring boundary is ignored due to little information use (1.17%).

Trial Sequential Analysis on sustained virological response in the five vitamin D trials was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 69798 participants. The trial sequential monitoring boundary is ignored due to little information use (0.45%).
Figuras y tablas -
Figure 8

Trial Sequential Analysis on sustained virological response in the five vitamin D trials was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 69798 participants. The trial sequential monitoring boundary is ignored due to little information use (0.45%).

Trial Sequential Analysis on acute cellular rejection in the one vitamin D trial was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 11958 participants. The cumulative Z‐curve did not cross the conventional monitoring boundary. The trial sequential monitoring boundary is ignored due to little information use (0.84%).
Figuras y tablas -
Figure 9

Trial Sequential Analysis on acute cellular rejection in the one vitamin D trial was performed based on a mortality rate in the control group of 5%, a relative risk reduction (RRR) of 30% in the intervention group, a type I error of 2.5%, and type II error of 10% (90% power). There was no diversity. The required information size was 11958 participants. The cumulative Z‐curve did not cross the conventional monitoring boundary. The trial sequential monitoring boundary is ignored due to little information use (0.84%).

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 1 All‐cause mortality.
Figuras y tablas -
Analysis 1.1

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 1 All‐cause mortality.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 2 All‐cause mortality ('best‐worst' case and 'worst‐best' case scenarios).
Figuras y tablas -
Analysis 1.2

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 2 All‐cause mortality ('best‐worst' case and 'worst‐best' case scenarios).

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 3 Liver‐related mortality.
Figuras y tablas -
Analysis 1.3

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 3 Liver‐related mortality.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 4 Serious adverse events.
Figuras y tablas -
Analysis 1.4

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 4 Serious adverse events.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 5 Non‐serious adverse events.
Figuras y tablas -
Analysis 1.5

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 5 Non‐serious adverse events.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 6 Failure of rapid virological response.
Figuras y tablas -
Analysis 1.6

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 6 Failure of rapid virological response.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 7 Failure of early virological response.
Figuras y tablas -
Analysis 1.7

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 7 Failure of early virological response.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 8 Failure of sustained virological response.
Figuras y tablas -
Analysis 1.8

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 8 Failure of sustained virological response.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 9 Acute cellular rejection in liver transplant recipients.
Figuras y tablas -
Analysis 1.9

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 9 Acute cellular rejection in liver transplant recipients.

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 10 Vitamin D status (ng/mL).
Figuras y tablas -
Analysis 1.10

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 10 Vitamin D status (ng/mL).

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 11 Biochemical indices.
Figuras y tablas -
Analysis 1.11

Comparison 1 Vitamin D versus placebo or no intervention, Outcome 11 Biochemical indices.

Summary of findings for the main comparison. Vitamin D compared to placebo or no intervention for chronic liver diseases in adults

Vitamin D compared to placebo or no intervention for chronic liver diseases in adults

Patient or population: adults with chronic liver diseases.
Setting: inpatients and outpatients from Austria, China, Egypt, Iran, Israel, Italy, Japan, USA.
Intervention: vitamin D
Comparison: placebo or no intervention

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(trials)

Quality of the evidence
(GRADE)

Comments

Risk with placebo or no intervention

Risk with vitamin D

All‐cause mortality at the end of follow‐up

Follow‐up: 0.1 to 1.4, mean 0.6 years

Study population

OR 0.70
(0.09 to 5.38)

1034
(15 RCTs)

⊕⊝⊝⊝
Very low1,2,3

Trial Sequential Analyses‐adjusted CI was 0.00 to 2534.

4 per 1.000

3 per 1.000
(0 to 21)

Liver‐related mortality

Follow‐up: mean 1 year

Study population

RR 1.62
(0.08 to 34.66)

18
(1 RCT)

⊕⊝⊝⊝
Very low1,3

Due to few data, we did not conduct Trial Sequential Analysis which would only have revealed larger imprecision.

0 per 1.000

0 per 1.000
(0 to 0)

Serious adverse events ‐ hypercalcaemia

Follow‐up: mean 1 year

Study population

RR 5.00
(0.25 to 100.80)

76
(1 RCT)

⊕⊝⊝⊝
Very low1,3

Due to few data, we did not conduct Trial Sequential Analysis which would only have revealed larger imprecision.

0 per 1.000

0 per 1.000
(0 to 0)

Serious adverse events ‐ myocardial infarction

Follow‐up: 0.2 to 1, mean 0.6 years

Study population

RR 0.75
(0.08 to 6.81)

86
(2 RCTs)

⊕⊝⊝⊝
Very low1,3

Due to few data, we did not conduct Trial Sequential Analysis which would only have revealed larger imprecision.

25 per 1.000

19 per 1.000
(2 to 170)

Serious adverse events ‐ thyroiditis

Follow‐up: mean 0.2 years

Study population

RR 0.33
(0.01 to 7.91)

68
(1 RCT)

⊕⊝⊝⊝
Very low1,3

Due to few data, we did not conduct Trial Sequential Analysis which would only have revealed larger imprecision.

29 per 1.000

10 per 1.000
(0 to 233)

Failure of sustained virological response

Follow‐up: 0.3 to 1.4, mean 0.9 years

Study population

RR 0.59
(0.28 to 1.21)

422
(5 RCTs)

⊕⊝⊝⊝

Very low

1,2,3,6

The trial sequential monitoring boundary is ignored due to little information use (0.6%).

465 per 1.000

275 per 1.000
(130 to 563)

Acute cellular rejection in liver transplant recipients

Follow‐up: mean 0.08 years

Study population

RR 0.33
(0.04 to 2.62)

75
(1 RCT)

⊕⊝⊝⊝

Very low

1,3,7

The trial sequential monitoring boundary is ignored due to little information use (0.84%).

120 per 1.000

40 per 1.000
(5 to 314)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: Confidence interval; RR: Risk ratio; RCT: randomised clinical trial; OR: Odds ratio.

GRADE Working Group grades of evidence

  • High quality: this research provides a very good indication of the likely effect; the likelihood that the effect will be substantially different is low.

  • Moderate quality: this research provides a good indication of the likely effect; the likelihood that the effect will be substantially different is moderate.

  • Low quality: this research provides some indication of the likely effect; however, the likelihood that it will be substantially different is high.

  • Very low quality: this research does not provide a reliable indication of the likely effect; the likelihood that the effect will be substantially different is very high.

1 Downgraded one level due to risk of bias: all trials were at high risk of bias.
2 Downgraded one level due to inconsistency of evidence: intertrial heterogeneity was significant.
3 Downgraded one level due to imprecision of evidence: Trial Sequential Analysis of vitamin D trials shows that we had insufficient information.
4 Downgraded one level due to indirectness of evidence: rapid virological response is a surrogate outcome.
5 Downgraded one level due to indirectness of evidence: early virological response is a surrogate outcome.
6 Downgraded one level due to indirectness of evidence: sustained virological response is a surrogate outcome.
7 Downgraded one level due to indirectness of evidence: acute cellular rejection is a surrogate outcome.

Figuras y tablas -
Summary of findings for the main comparison. Vitamin D compared to placebo or no intervention for chronic liver diseases in adults
Table 1. Characteristics of included trials (I)

Study ID

Protocol

Design

Groups

Bias
risk

Blinding

Participants
(n)

Women
(%)

Mean
age (years)

Abu‐Mouch 2011

Yes

Parallel

2

High

NI

72

44

47

Atsukawa 2016

No

Parallel

2

High

NI

115

50

64

Barchetta 2016

Yes

Parallel

2

High

PL

65

35

59

Esmat 2015

No

Parallel

2

High

NI

101

25

40

Foroughi 2016

Yes

Parallel

2

High

PL

60

52

48

Lorvand Amiri 2016

Yes

Parallel

3

High

PL

120

38

41

Mobarhan 1984

No

Parallel

3

High

NI

18

0

61

Nimer 2012

No

Parallel

2

High

NI

50

58

47

Pilz 2016

Yes

Parallel

2

High

PL

36

25

61

Sharifi 2014

No

Parallel

2

High

PL

60

51

60

Shiomi 1999a

No

Parallel

2

High

NI

76

66

61

Shiomi 1999b

No

Parallel

2

High

NI

34

100

56

Vosoghinia 2016

Yes

Parallel

2

High

NI

68

13

42

Xing 2013

No

Parallel

3

High

PL

75

17

48

Yokoyama 2014

No

Parallel

2

High

NI

84

49

59

n: number of participants; NI: no intervention; PL: placebo.

Figuras y tablas -
Table 1. Characteristics of included trials (I)
Table 2. Characteristics of included trials (II)

Study ID

Participants

Outcome measures

Sponsor

Country

Abu‐Mouch 2011

Chronic hepatitis C genotype 1

Sustained virological response

No information

Israel

Atsukawa 2016

Chronic hepatitis C genotype 1

Sustained virological response

No information

Japan

Barchetta 2016

NAFLD

Liver steatosis, liver function

No

Italy

Esmat 2015

Chronic hepatitis C genotype 4

Sustained virological response

No information

Egypt

Foroughi 2016

NAFLD

Liver steatosis, liver function

No

Iran

Lorvand Amiri 2016

NAFLD

Liver function, body fat

No

Iran

Mobarhan 1984

Liver cirrhosis

Bone mineral density

Yes

USA

Nimer 2012

Chronic hepatitis C genotype 2 or 3

Sustained virological response

No information

Israel

Pilz 2016

Liver cirrhosis

Vitamin D status, liver function

No

Austria

Sharifi 2014

NAFLD

Liver function, insulin resistance index

No

Iran

Shiomi 1999a

Liver cirrhosis

Bone mineral density

No information

Japan

Shiomi 1999b

Primary biliary cirrhosis

Bone mineral density

No information

Japan

Vosoghinia 2016

Chronic hepatitis C genotype 1,2,3,4

Early virological response

No

Iran

Xing 2013

Liver transplant recipients

Acute cellular rejection rate

No

China

Yokoyama 2014

Chronic hepatitis C genotype 1

Sustained virological response

No information

Japan

NAFLD: non‐alcoholic fatty liver disease.

Figuras y tablas -
Table 2. Characteristics of included trials (II)
Table 3. Characteristics of included studies (III)

Study ID

Vitamin

Calcium
(mg)

Regimen*

Treatment
(weeks)

Follow‐up
(weeks)

Cointervention

D3
(IU)

D2
(IU)

25(OH)D
(IU)

1,25(OH)2D
(µg)

Abu‐Mouch 2011

2000

Daily

48

72

PEG‐INF, RBV

Atsukawa 2016

2000

Daily

16

16

PEG‐INF, RBV, SP

Barchetta 2016

2000

Daily

24

24

Esmat 2015

2143

Weekly

48

72

PEG‐INF, RBV

Foroughi 2016

7143

Weekly

10

10

Lorvand Amiri 2016

1000

500

Daily

10

12

Mobarhan 1984

17,857

2400

Daily

52

52

Nimer 2012

2000

Daily

24

48

PEG‐INF, RBV

Pilz 2016

2800

Daily

8

8

Sharifi 2014

3571

Twice a week

16

16

Shiomi 1999a

1

Daily

52

52

Shiomi 1999b

1

Daily

52

52

Vosoghinia 2016

1600

Daily

12

12

PEG‐INF, RBV

Xing 2013

0.25

1000

Daily

4

4

Yokoyama 2014

1000

Daily

16

24

PEG‐INF, RBV

* Vitamin D was administered orally in all trials.
1,25(OH)2D: calcitriol; 25(OH)D: calcidiol; PEG‐INF: pegylated‐interferon; RBV: ribavirin; SP: simeprevir.

Figuras y tablas -
Table 3. Characteristics of included studies (III)
Comparison 1. Vitamin D versus placebo or no intervention

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

15

1034

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.70 [0.09, 5.38]

2 All‐cause mortality ('best‐worst' case and 'worst‐best' case scenarios) Show forest plot

15

Peto Odds Ratio (Peto, Fixed, 95% CI)

Subtotals only

2.1 'Best‐worst' case scenario

15

1034

Peto Odds Ratio (Peto, Fixed, 95% CI)

0.11 [0.05, 0.24]

2.2 'Worst‐best' case scenario

15

1034

Peto Odds Ratio (Peto, Fixed, 95% CI)

7.80 [3.67, 16.57]

3 Liver‐related mortality Show forest plot

1

18

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.08, 34.66]

4 Serious adverse events Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Hypercalcaemia

1

76

Risk Ratio (M‐H, Random, 95% CI)

5.0 [0.25, 100.80]

4.2 Myocardial infarction

2

86

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.08, 6.81]

4.3 Thyroiditis

1

68

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.91]

5 Non‐serious adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 Glossitis

1

65

Risk Ratio (M‐H, Random, 95% CI)

3.70 [0.16, 87.58]

6 Failure of rapid virological response Show forest plot

2

187

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.52, 0.94]

7 Failure of early virological response Show forest plot

2

140

Risk Ratio (M‐H, Random, 95% CI)

0.10 [0.03, 0.33]

8 Failure of sustained virological response Show forest plot

5

422

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.28, 1.21]

9 Acute cellular rejection in liver transplant recipients Show forest plot

1

75

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 2.62]

10 Vitamin D status (ng/mL) Show forest plot

6

424

Mean Difference (IV, Random, 95% CI)

17.24 [12.46, 22.02]

11 Biochemical indices Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

11.1 Aspartate aminotransferase (IU/L)

6

313

Mean Difference (IV, Random, 95% CI)

‐1.40 [‐2.88, 0.08]

11.2 Alanine aminotransferase (IU/L)

6

313

Mean Difference (IV, Random, 95% CI)

‐0.52 [‐5.10, 4.06]

11.3 Alkaline phosphatases (IU/L)

2

96

Mean Difference (IV, Random, 95% CI)

7.39 [‐39.89, 54.67]

11.4 Gamma‐glutamyl transpeptidase (IU/L)

2

101

Mean Difference (IV, Random, 95% CI)

3.64 [0.33, 6.96]

11.5 Albumin (g/L)

2

48

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.40, 0.20]

11.6 Bilirubin (mg/dL)

2

48

Mean Difference (IV, Random, 95% CI)

0.38 [0.21, 0.55]

11.7 Triglyceride (mg/dL)

2

115

Mean Difference (IV, Random, 95% CI)

23.69 [‐13.90, 61.27]

11.8 Cholesterol (mg/dL)

1

55

Mean Difference (IV, Random, 95% CI)

2.75 [‐4.75, 10.25]

11.9 Calcium (mg/dL)

2

72

Mean Difference (IV, Random, 95% CI)

2.01 [‐0.53, 4.56]

Figuras y tablas -
Comparison 1. Vitamin D versus placebo or no intervention