Scolaris Content Display Scolaris Content Display

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

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

Behera 2018 {published data only}

Behera MK, Shukla SK, Dixit VK, Nath P, Abhilash VB, Asati PK, et al. Effect of vitamin D supplementation on sustained virological response in genotype 1/4 chronic hepatitis C treatment-naïve patients from India. Indian Journal of Medical Research 2018;148(2):200-6. CENTRAL

Boonyagard 2016 {published data only}

Boonyagard S, Techatuvanan K. Impact of vitamin D replacement on liver enzymes in non-alcoholic fatty liver disease patients. Hepatology (Baltimore, Md.) 2016;64(S1):542. CENTRAL

Dabbaghmanesh 2018 {published data only}

Dabbaghmanesh MH, Danafar F, Eshraghian A, Omrani GR. Vitamin D supplementation for the treatment of non-alcoholic fatty liver disease: a randomized double blind placebo controlled trial. Diabetes and Metabolic Syndrome 2018;12(4):513-7. 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

Geier 2018 {published data only}

Geier A, Eichinger M, Stirnimann G, Semela D, Tay F, Seifert B, et al. Treatment of non-alcoholic steatohepatitis patients with vitamin D: a double-blinded, randomized, placebo-controlled pilot study. Scandinavian Journal of Gastroenterology 2018;53(9):1114-20. CENTRAL

Hosseini 2018 {published data only}

Hosseini SM, Aliashrafi S, Ebrahimi-Mameghani M. The effect of a single intramuscular injection of cholecalciferol on the serum levels of vitamin D, adiponectin, insulin resistance, and liver function in women with non-alcoholic fatty liver disease (NAFLD): a randomized, controlled clinical trial. Iranian Red Crescent Medical Journal 2018;20(10):1-12. CENTRAL

Hussain 2019 {published data only}

Hussain M, Iqbal J, Malik SA, Waheed A, Shabnum S, Akhtar L, et al. Effect of vitamin D supplementation on various parameters in non-alcoholic fatty liver disease patients. Pakistan Journal of Pharmaceutical Sciences 2019;32(3 Special):1343-8. CENTRAL

Jeong 2019 {published data only}

Jeong JY, Jun DW, Park SJ, Sohn JH, Kim SG, Lee SW, et al. Effects of vitamin D supplements in patients with chronic hepatitis C: a randomized, multi-center, open label study. Korean Journal of Internal Medicine 2019;35(5):1074-83. CENTRAL

Jha 2017 {published data only}

Jha AK, Jha SK, Kumar A, Dayal VM, Jha SK. Effect of replenishment of vitamin D on survival in patients with decompensated liver cirrhosis: a prospective study. World Journal of Gastrointestinal Pathophysiology 2017;8(3):133-41. CENTRAL

Komolmit 2017a {published data only}

Komolmit P, Charoensuk K, Thanapirom K, Suksawatamnuay S, Thaimai P, Chirathaworn C, et al. Correction of vitamin D deficiency facilitated suppression of IP-10 and DPP IV levels in patients with chronic hepatitis C: a randomised double-blinded, placebo-control trial. PLOS ONE 2017;12(4):e0174608. CENTRAL

Komolmit 2017b {published data only}

Komolmit P, Kimtrakool S, Suksawatamnuay S, Thanapirom K, Chattrasophon K, Thaimai P, et al. Vitamin D supplementation improves serum markers associated with hepatic fibrogenesis in chronic hepatitis C patients: a randomized, double-blind, placebo-controlled study. Scientific Reports 2017;7(1):8905. 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
Shidfar F, Mousavi SN, Lorvand Amiri H, Agah S, Hoseini S, Hajimiresmail SJ. Reduction of some atherogenic indices in patients with non-alcoholic fatty liver by vitamin D and calcium co-supplementation: a double blind randomized controlled clinical trial. Iranian Journal of Pharmaceutical Research 2019;18(1):496-505. 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 (Baltimore, Md.) 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):278. CENTRAL

Sakpal 2017 {published data only}

Sakpal M, Satsangi S, Mehta M, Duseja A, Bhadada S, Das A, et al. Vitamin D supplementation in patients with nonalcoholic fatty liver disease: a randomized controlled trial. Journal of Gastroenterology and Hepatology Open 2017;1(2):62-7. 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
Sharifi N, Amani R, Hajiani E, Cheraghian B. Women may respond different from men to vitamin D supplementation regarding cardiometabolic biomarkers. Experimental Biology & Medicine (Maywood) 2016;241(8):830-8. 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

Taghvaei 2018 {published data only}

Taghvaei T, Akha O, Mouodi M, Fakheri HT, Kashi Z, Maleki I, et al. Effects of vitamin D supplementation on patients with non-alcoholic fatty liver disease (NAFLD). Acta Medica Mediterranea 2018;34:415–22. 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 response rate: a clinical trial. Razavi International Journal of Medicine 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

Chen 2015 {published data only}

Chen EQ, Bai L, Zhou TY, Fe M, Zhang DM, Tang H. Sustained suppression of viral replication in improving vitamin D serum concentrations in patients with chronic hepatitis B. Scientific Reports 2015;5:15441. CENTRAL

Dasarathy 2017 {published data only}

Dasarathy J, Varghese R, Feldman A, Khiyami A, McCullough AJ, Dasarathy S. Patients with nonalcoholic fatty liver disease have a low response rate to vitamin D supplementation. Journal of Nutrition 2017;147(10):1938-46. 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

Hasanain 2018 {published data only}

Hasanain AF, Zayed AA, Mahdy RE, Nafee AM. Cholecalciferol for prophylaxis against antituberculosis therapy-induced liver disorders among naïve patients with pulmonary tuberculosis: a randomized, comparative study. International Journal of Mycobacteriology 2017;6:149-55. 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, Jørgensen PS, Lauridsen AL, Ott P, Glerup H, Dahlerup JF. The effect of a single oral megadose of vitamin D provided as either ergocalciferol (D 2) or cholecalciferol (D 3) in alcoholic liver cirrhosis. European Journal of Gastroenterology & Hepatology 2012;24(2):172-8. CENTRAL

Naderpoor 2018 {published data only}

Naderpoor N, Mousa A, De Courten M, Scragg R, De Courten B. The relationship between 25-hydroxyvitamin D concentration and liver enzymes in overweight or obese adults: cross-sectional and interventional outcomes. Journal of Steroid Biochemistry and Molecular Biology 2018;177:193-9. CENTRAL

Omori‐Mizuno 2015 {published data only}

Omori-Mizuno Y, Nakayama N, Inao M, Funyu J, Asabe S, Tomita K, et al. Randomized study comparing vitamin D3 and 1α-hydroxyvitamin D3 in combination with pegylated interferon/ribavirin therapy for chronic hepatitis C. Journal of Gastroenterology and Hepatology 2015;30(9):1384-90. 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

Park 2017 {published data only}

Park D, Kwon H, Oh SW, Joh HK, Hwang SS, Park JH, et al. Is vitamin D an independent risk factor of nonalcoholic fatty liver disease?: a cross-sectional study of the healthy population. Journal of Korean Medical Science 2017;32(1):95-101. 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

Tavakoli 2019 {published data only}

Tavakoli H, Rostami H, Avan A, Bagherniya M, Ferns GA, Khayyatzadeh SS. High dose vitamin D supplementation is associated with an improvement in serum markers of liver function. Biofactors 2019;45(3):335-42. 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

Zhou 2019 {published data only}

Zhou Q, Li L, Chen Y, Zhang J, Zhong L, Peng Z, et al. Vitamin D supplementation could reduce the risk of acute cellular rejection and infection in vitamin D deficient liver allograft recipients. International Immunopharmacology 2019;75:105811. CENTRAL

IRCT2016020326342N1 {published data only}

IRCT2016020326342N1. Effectiveness of vitamin D supplementation on severity of cirrhosis based on CHILD and MELD scores in patients with decompensate cirrhosis. en.irct.ir/trial/21847 (first received 28 February 2016). CENTRAL

NCT02779465 {published data only}

NCT02779465. Oral vitamin D treatment for the prevention of hepatocellular carcinoma (VDHCC). clinicaltrials.gov/ct2/show/NCT02779465 (first received 20 May 2016). CENTRAL

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 2018

Atef SH. Vitamin D assays in clinical laboratory: past, present and future challenges. Journal of Steroid Biochemistry and Molecular Biology 2018;175:136-7.

Autier 2014

Autier P, Boniol M, Mullie P. Vitamin D status and ill health: a systematic review. 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.

Balshem 2011

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

Barbarawi 2019

Barbarawi M, Kheiri B, Zayed Y, Barbarawi O, Dhillon H, Swaid B, et al. Vitamin D supplementation and cardiovascular disease risks in more than 83 000 individuals in 21 randomized clinical trials: a meta-analysis. JAMA Cardiology 2019;4(8):765-76.

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.

Bischoff‐Ferrari 2020

Bischoff-Ferrari HA, Vellas B, Rizzoli R, Kressig RW, Da Silva JAP, Blauth M, et al. Effect of vitamin D supplementation, omega-3 fatty acid supplementation, or a strength-training exercise program on clinical outcomes in older adults: the DO-HEALTH randomized clinical trial. JAMA 2020;324(18):1855-68.

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. Art. No: CD007470. [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. Art. No: CD007469. [DOI: 10.1002/14651858.CD007469.pub2]

Boesen 2021

Boesen K. Methodological Limitations of Psychiatric Drug Trials with a Particular Focus on Extended-Release Methylphenidate (Metodebegraensninger i Psykiatriske Laegemiddelforsøg Med et Saerligt Fokus på Methylphenidat Depotpraeparater) [PhD thesis]. University of Copenhagen, 2020.

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.

Bolland 2018

Bolland MJ, Grey A, Avenell A. Effects of vitamin D supplementation on musculoskeletal health: a systematic review, meta-analysis, and trial sequential analysis. Lancet Diabetes & Endocrinology 2018;6(11):847-58.

Borenstein 2009

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

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.

Castellini 2018

Castellini G, Bruschettini M, Gianola S, Gluud C, Moja L. Assessing imprecision in Cochrane systematic reviews: a comparison of GRADE and Trial Sequential Analysis. Systematic Reviews 2018;7(1):110.

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.

Deeks 2021

Deeks JJ, Higgins JPT, Altman DG, editor(s). Chapter 10: Analysing data and undertaking meta-analyses. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook.

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 (Baltimore, Md.) 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.

Gartlehner 2019

Gartlehner G, Nussbaumer-Streit B, Wagner G, Patel S, Swinson-Evans T, Dobrescu A, et al. Increased risks for random errors are common in outcomes graded as high certainty of evidence. Journal of Clinical Epidemiology 2019;106:50-9.

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).

GRADEpro GDT [Computer program]

McMaster University (developed by Evidence Prime)GRADEpro GDT. Version accessed 4 November 2020. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015. Available at gradepro.org.

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 3rd, 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.

Guo 2020

Guo XF, Wang C, Yang T, Li S, Li KL, Li D. Vitamin D and non-alcoholic fatty liver disease: a meta-analysis of randomized controlled trials. Food and Function 2020;11(9):7389-99.

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.

Guyatt 2011a

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction - GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383-94. [PMID: 21195583]

Guyatt 2011b

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

Guyatt 2011c

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

Guyatt 2011d

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

Guyatt 2011e

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

Guyatt 2011f

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

Guyatt 2011g

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

Guyatt 2011h

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

Guyatt 2013a

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

Guyatt 2013b

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

Guyatt 2013c

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

Guyatt 2013d

Guyatt G, Andrews J, Oxman AD, Alderson P, Dahm P, Falck-Ytter Y, et al. GRADE guidelines: 15. Going from evidence to recommendations: the significance and presentation of recommendations. Journal of Clinical Epidemiology 2013;66(7):719-25.

Guyatt 2017

Guyatt GH, Ebrahim S, Alonso-Coello P, Johnston BC, Mathioudakis AG, Briel M, et al. GRADE guidelines 17: assessing the risk of bias associated with missing participant outcome data in a body of evidence. Journal of Clinical Epidemiology 2017;87:14-22. [PMID: 28529188]

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.

Hariri 2019

Hariri M, Zohdi S. Effect of vitamin D on non-alcoholic fatty liver disease: a systematic review of randomized controlled clinical trials. International Journal of Preventive Medicine 2019;10(1):14.

Harvey 2012

Harvey NC, Cooper C. Vitamin D: some perspective please. BMJ (Clinical Research Ed.) 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 (Clinical Research Ed.) 2003;327:557-60.

Higgins 2011

Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from training.cochrane.org/handbook/archive/v5.1.

Higgins 2021

Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 6.2 (updated February 2021). Cochrane, 2021. Available from training.cochrane.org/handbook.

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 (Clinical Research Ed.) 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.

Hu 2019

Hu Y-C, Wang W-W, Jiang W-Y, Li C-Q, Guo J-C, Xun Y-H. Low vitamin D levels are associated with high viral loads in patients with chronic hepatitis B: a systematic review and meta-analysis. BMC Gastroenterology 2019;19(1):84.

ICH‐GCP 1997

International Conference on Harmonisation Expert Working Group. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. ICH Harmonised Tripartite Guideline. Guideline for Good Clinical Practice CFR & ICH Guidelines. Vol. 1. Philadelphia: Barnett International/PAREXEL, 1997.

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 2014a

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. Art. No: CD012143. [DOI: 10.1002/14651858.CD012143.pub3]

Jaruvongvanich 2017

Jaruvongvanich V, Ahuja W, Sanguankeo A, Wijarnpreecha K, Upala S. Vitamin D and histologic severity of nonalcoholic fatty liver disease: a systematic review and meta-analysis. Digestive and Liver Disease 2017;49(6):618-22.

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.

Keum 2019

Keum N, Lee DH, Greenwood DC, Manson JE, Giovannucci E. Vitamin D supplementation and total cancer incidence and mortality: a meta-analysis of randomized controlled trials. Annals of Oncology 2019;30(5):733-43.

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.

Kim 2018

Kim H-B, Myung S-K, Lee Y-J, Park B-J. Efficacy of vitamin D supplementation in combination with conventional antiviral therapy in patients with chronic hepatitis C infection: a meta-analysis of randomised controlled trials. Journal of Human Nutrition and Dietetics 2018;31(2):168-77.

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 (Baltimore, Md.) 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. Art. No: MR000033. [DOI: 10.1002/14651858.MR000033.pub3]

Luong 2012

Luong 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.

Manson 2019

Manson JE, Cook NR, Lee IM, Christen W, Bassuk SS, Mora S, et al. Vitamin D supplements and prevention of cancer and cardiovascular disease. New England Journal of Medicine 2019;380(1):33-44.

Mansour‐Ghanaei 2019

Mansour-Ghanaei F, Pourmasoumi M, Hadi A, Ramezani-Jolfaie N, Joukar F. The efficacy of vitamin D supplementation against nonalcoholic fatty liver disease: a meta-analysis. Journal of Dietary Supplements 2019;17(4):467-85.

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.

Mosannen 2017

Mosannen Mozaffari H, Abbasi HA, Goshayeshi G, Esmaeelzadeh A, Bahari A, Mokhtarifar A, et al. Effect of vitamin D supplementation on chronic liver disease: systematic literature review. Reviews in Clinical Medicine 2017;4(1):14-9.

Mustafa 2013

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

Nordic Trial Alliance 2015

Nordic Trial Alliance Working Group on Transparency and Registration. Transparency and registration in clinical research in the Nordic countries. nta.nordforsk.org/projects/nta_transparency_report.pdf (accessed 6 November 2020).

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.

Peng 2020

Peng L, Li L, Wang P, Chong W, Li Y, Zha X, et al. Association between vitamin D supplementation and mortality in critically ill patients: a systematic review and meta-analysis of randomized clinical trials. PLOS ONE 2020;15(12):e0243768.

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 (Baltimore, Md.) 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.

Review Manager 2020 [Computer program]

Nordic Cochrane Centre, The Cochrane CollaborationReview Manager (RevMan). Version 5.4. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2020.

SACN 2016

Public Health England. SACN vitamin D and health report. 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.

Savović 2018a

Savović J, Akl EA, Hróbjartsson A. Financial conflicts of interest in clinical research. Intensive Care Medicine 2018;44(10):1767-9.

Savović 2018b

Savović J, Turner RM, Mawdsley D, Jones HE, Beynon R, Higgins JPT, et al. Association between risk-of-bias assessments and results of randomized trials in Cochrane reviews: the ROBES meta-epidemiologic study. American Journal of Epidemiology 2018;187(5):1113–22.

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.

Schroll 2013

Schroll JB, Bero L, Gøtzsche PC. Searching for unpublished data for Cochrane reviews: cross sectional study. BMJ (Clinical Research Ed.) 2013;346:f2231.

Schulz 1995

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

Schünemann 2013

Schünemann H, Brożek J, Guyatt G, Oxman A, editor(s). Handbook for grading the quality of evidence and the strength of recommendations using the GRADE approach (updated October 2013). GRADE Working Group, 2013. Available from gdt.guidelinedevelopment.org/app/handbook/handbook.html.

Scragg 2017

Scragg R, Stewart AW, Waayer D, Lawes CMM, Toop L, Sluyter J, et al. Effect of monthly high-dose vitamin D supplementation on cardiovascular disease in the Vitamin D Assessment Study: a randomized clinical trial. JAMA Cardiology 2017;2(6):608-16.

Scragg 2018

Scragg R, Khaw KT, Toop L, Sluyter J, Lawes CMM, Waayer D, et al. Monthly high-dose vitamin D supplementation and cancer risk: a post hoc analysis of the Vitamin D Assessment randomized clinical trial. JAMA Oncology 2018;4(11):e182178.

Sharifi 2019

Sharifi N, Amani R. Vitamin D supplementation and non-alcoholic fatty liver disease: a critical and systematic review of clinical trials. Critical Reviews in Food Science and Nutrition 2019;59(4):693-703.

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.

Sigma Stat 2003 [Computer program]

Systat Software Inc.Sigma Stat 3.0. Systat Software Inc., 2003.

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.

StataCorp 2005 [Computer program]

College Station, TX: StataCorp LPIntercooled Stata 8.2 for Windows. College Station, TX: StataCorp LP, 2005.

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 (Clinical Research Ed.) 2015;351:h5203.

Storebø 2018

Storebø OJ, Pedersen N, Ramstad E, Kielsholm ML, Nielsen SS, Krogh HB, et al. Methylphenidate for attention deficit hyperactivity disorder (ADHD) in children and adolescents – assessment of adverse events in non-randomised studies. Cochrane Database of Systematic Reviews 2018, Issue 5. Art. No: CD012069. [DOI: 10.1002/14651858.CD012069.pub2]

Tabrizi 2017

Tabrizi R, Moosazadeh M, Lankarani KB, Akbari M, Heydari ST, Kolahdooz F, et al. The effects of vitamin D supplementation on metabolic profiles and liver function in patients with non-alcoholic fatty liver disease: a systematic review and meta-analysis of randomized controlled trials. Diabetes & Metabolic Syndrome 2017;11:S975-82.

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 2011

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.

Thorlund 2017

Thorlund K, Engstrøm J, Wetterslev J, Brok J, Imberger G, Gluud C. User manual for Trial Sequential Analysis (TSA). ctu.dk/wp-content/uploads/2020/12/tsa_manual_ENG.pdf 2017 (accessed 2 September 2020).

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 UnitTrial Sequential Analysis. Copenhagen Trial Unit, Version 0.9.5.10 Beta. Copenhagen: Copenhagen Trial Unit, 2017. ctu.dk/tsa/downloads/.

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, editors(s). Vitamin D. 2nd edition. Amsterdam: 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 (Clinical Research Ed.) 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 (Baltimore, Md.) 2016;64(1):73-84.

Zhang 2019

Zhang Y, Fang F, Tang J, Jia L, Feng Y, Xu P, et al. Association between vitamin D supplementation and mortality: systematic review and meta-analysis. BMJ (Clinical Research Ed.) 2019;366:l4673.

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. Cochrane Database of Systematic Reviews 2015, Issue 3. Art. No: CD011564. [DOI: 10.1002/14651858.CD011564]

Bjelakovic 2017

Bjelakovic G, Nikolova D, Bjelakovic M, Gluud C. Vitamin D supplementation for chronic liver diseases in adults. Cochrane Database of Systematic Reviews 2017, Issue 11. Art. No: CD011564. [DOI: 10.1002/14651858.CD011564.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abu‐Mouch 2011

Study characteristics

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

All participants completed the trial. Vitamin D3 (Vitamidyne D, Fischer Pharmaceuticals, Israel) given by oral drops for 4 weeks before initiation of antiviral treatment and after serum levels had 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 the outcome is likely to have been 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 have been 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 in full.

Other bias

Low risk

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

Atsukawa 2016

Study characteristics

Methods

Open‐label randomised clinical trial with parallel‐group design (2 groups)

Participants

Number of participants randomised: 115 participants (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 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 μL; 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. Patients who had not received PEG IFN/ribavirin combination therapy were considered 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 administered subcutaneously at a dose of 180 μg once weekly. Ribavirin was administered orally twice daily, with doses adjusted according to body weight (600 mg daily for < 60 kg, 800 mg daily for 60 to 80 kg, and 1000 mg daily for > 80 kg). Simeprevir was administered 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 participants 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 of 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

Study authors did not report any deaths. "No patient complained of vitamin D‐related symptoms or developed signs of a vitamin D‐related adverse reaction".

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 is likely to have been influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding, and outcome measurement is likely to have been 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 was likely to induce bias

Selective reporting (reporting bias)

Unclear risk

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

Other bias

Low risk

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

Barchetta 2016

Study characteristics

Methods

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

Participants

65 participants (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, glycated haemoglobin, 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).

Study authors did not report any deaths. Authors were not contacted, as information on our outcomes of interest was found in the publication: “As per the safety profile, no major adverse events occurred during the study”.

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 were 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 it is 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 was likely to introduce bias on the results

Selective reporting (reporting bias)

Low risk

Study authors reported all predefined outcomes in full.

Other bias

Low risk

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

Behera 2018

Study characteristics

Methods

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

Participants

Country: India

60 participants (40% women), mean age 41 years

Inclusion criteria: age 18 to 65 years, chronic HCV genotype 1/4, infection with detectable HCV RNA for 6 months, and no previous treatment for hepatitis C

Exclusion criteria: advanced cirrhosis (Child‐Pugh B or C), presence of HCC, HIV and hepatitis B co‐infection, autoimmune liver disease, Wilson disease, haemochromatosis, α1‐antitrypsin deficiency, concomitant use of medications known to affect serum vitamin D metabolism, and patients with active intravenous drug addiction

Interventions

Intervention: PEG‐IFN alfa‐2a 180 μg per week, and RBV (1000 mg/day for participants weighing < 75 kg, 1200 mg/day for participants weighing > 75 kg) and vitamin D3 2000 IU/day (n = 28)

Control: PEG‐IFN alfa‐2a 180 μg per week, and RBV (1000 mg/day for participants weighing < 75 kg, 1200 mg/day for participants weighing > 75 kg) (n = 32)

For 48 weeks

Outcomes

Primary outcomes: rapid, early and sustained viral response

Stated aim of study

To assess the effect of vitamin D supplementation on treatment outcome in patients with genotype 1/4 chronic hepatitis C (CHC) infection

Notes

All participants completed the trial.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The computer‐generated block randomization schedule was prepared using random number generator to create a list of random numbers. Stat Trek programme (https://stattrek.com) was used to derive the randomization list."

Allocation concealment (selection bias)

Unclear risk

Study authors did not describe the method used to conceal the allocation, so the intervention allocations may have been foreseen before, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding, and the outcome is likely to have been 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 have been 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

The study was registered in Clinical Trials Registry‐India (CTRI/2015/07/005992).

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias, such as competing interest bias.

Boonyagard 2016

Study characteristics

Methods

Randomised placebo‐controlled clinical trial with parallel‐group design (2 groups)

Participants

Country: Thailand

Number of participants randomised: 60 participants

Inclusion criteria: NAFLD patients who have ALT elevation with vitamin D insufficiency

Exclusion criteria: none stated

Interventions

Intervention: vitamin D (n = 30)

Control: placebo (n = 30)

For 20 weeks

Outcomes

Primary outcomes: serum ALT, inflammatory markers, and homeostasis model assessment and Fibroscan

Secondary outcomes: none stated

Stated aim of study

To demonstrate the effect of vitamin D replacement on liver enzymes and inflammatory markers in NAFLD patients

Notes

Results were presented as an abstract. We were unable to find the address of the authors to contact them for the missing information. Study authors did not report any deaths. No information provided about adverse events.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Study authors did not specify the method of sequence generation.

Allocation concealment (selection bias)

Unclear risk

Study authors did not describe the method used to conceal the allocation, so the 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

Unclear risk

Information was insufficient to assess whether missing data in combination with the method used to handle missing data was likely to induce bias on the results.

Selective reporting (reporting bias)

Unclear risk

The protocol was not available.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias, such as competing interest bias.

Dabbaghmanesh 2018

Study characteristics

Methods

Randomised double‐blind placebo‐controlled clinical trial with parallel‐group design (3 groups)

Participants

Country: Iran

Number of participants randomised: 106 participants (59% women), mean age 45 years

Inclusion criteria: men and women aged between 20 and 75 years with presence of hepatic steatosis diagnosed by ultrasound

Exclusion criteria: liver cirrhosis, with positive results for hepatitis B virus surface antigen or hepatitis C virus antibody, patients with alcohol consumption (> 10 g/day), patients with autoimmune hepatitis or other causes of chronic liver diseases such as Wilson’s disease and haemochromatosis, known cancer, nephrolithiasis, nephrocalcinosis, chronic renal failure, hypercalcaemia, hypercalciuria, pregnancy, lactation, hypersensitivity to vitamin D3, patients receiving oestrogen, tamoxifen, methotrexate, amiodarone, or tetracycline, and receiving vitamin D and calcium supplementations in previous 6 months

Interventions

Intervention group 1: vitamin D3 (cholecalciferol) 50,000 IU pearl per week (n = 35)

Intervention group 2: calcitriol 0.25 mg (1,25 dihydroxycholecalciferol) pearl per day (n = 35)

Control: placebo (n = 36)

For 12 weeks

Outcomes

Primary outcomes: reduction of serum ALT, AST, GGT from baseline to 12 weeks

Secondary outcomes: improvement of metabolic component of participants including fasting plasma glucose, LDL, HDL, triglyceride, and total cholesterol

Stated aim of study

To investigate the role of vitamin D therapy in the amelioration of hepatic steatosis in people with NAFLD

Notes

Study authors did not report any deaths. No information provided about adverse events.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Block randomisation with a block size of 5 was used through a computer‐based procedure.

Allocation concealment (selection bias)

Unclear risk

Study authors did not describe the method used to conceal the allocation, so the intervention allocations may have been foreseen before, or during, enrolment.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Blinding of participants and key study personnel ensured, and it is unlikely that the blinding could have been broken. Quote: "Treatments and placebo were provided in identical packages and were given to the participants by an educated person who was blinded to the drug and patients."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and it is 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

The protocol was not available.

Other bias

Low risk

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

Esmat 2015

Study characteristics

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 (PegIntron, 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 the 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

Study authors did not report any deaths. “None of the missed patients had stopped the treatment due to adverse events”. 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 have been 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 have been 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 was likely to induce bias

Selective reporting (reporting bias)

Unclear risk

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

Other bias

Low risk

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

Foroughi 2016

Study characteristics

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 should 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 the effect of vitamin D supplementation on inflammation, liver function, and liver steatosis in people with NAFLD

Notes

All participants completed the trial. Clinical trial registered at Iranian Registry of Clinical Trials (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 in full.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias, such as competing interest bias.

Geier 2018

Study characteristics

Methods

Randomised double‐blind placebo‐controlled clinical trial with parallel‐group design (3 groups)

Participants

Country: Switzerland

Number of participants randomised: 20 participants, mean age 45 years

Inclusion criteria: increased alanine aminotransferase level (1.2‐fold ULN), histological diagnosis of NASH diagnosed according to the SAF score obtained within 18 months preceding entry and decreased 25‐OH vitamin D level (< 30 ng/L). The definition of NASH included non‐excessive alcohol consumption as fewer than 21 standard drinks on average per week in males and fewer than 14 standard drinks on average per week in females.

Exclusion criteria: cirrhosis, HCV RNA positivity, HBs antigen positivity, other liver disease including autoimmune hepatitis, hereditary haemochromatosis, alpha‐1‐antitrypsin deficiency, Wilson’s disease, drug‐induced fatty liver disease, serious diseases limiting life expectancy, pregnancy or breastfeeding, intention to become pregnant during the course of the study, or childbearing potential in women who were not using safe contraception

Interventions

Intervention: 2100 IU vitamin D3 (cholecalciferol) (n = 10)

Control: placebo (n = 10)

Orally daily for 48 weeks

Outcomes

Primary outcomes: change in ALT from baseline to end of treatment

Secondary outcomes: absolute reduction of hepatic steatosis by at least 20% or by at least 1 point in NASH, serious adverse events, safety laboratory assessments, physical examination findings and vital signs

Stated aim of study

To investigate the efficacy and safety of 48‐week treatment with vitamin D3 in NASH patients

Notes

Study authors did not report any deaths. 2 serious adverse events were recorded: circular haemorrhoidal prolapse and bronchopneumonia. The study medication (vitamin D3 2100 IU daily) and placebo were produced and provided by Antistress AG, Rapperswil‐Jona, Switzerland.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Study authors did not specify the method of sequence generation.

Allocation concealment (selection bias)

Low risk

A central and independent randomisation unit controlled the allocation. Quote: "Randomisation (stratified for the presence of diabetes, block size 10, not stratified by center) was performed by the Cantonal Pharmacy Zurich."

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 were unlikely to make treatment effects depart from plausible values.

Selective reporting (reporting bias)

Low risk

The trial was registered (NCT01571063; KEK‐ZH‐Nr. 2011‐420).

Other bias

Unclear risk

Trial may or may not have been free of other factors that could have put it at risk of bias, such as competing interest bias.

Hosseini 2018

Study characteristics

Methods

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

Participants

Country: Iran

Number of participants randomised: 82 women, aged 18 to 50 years, mean age 34 years

Inclusion criteria: BMI between 25 and 40 kg/m2 with NAFLD confirmed by single ultrasonographist, vitamin D insufficiency (serum 25‐hydroxyvitamin D < 30 ng/mL), not taking dietary supplements including calcium and vitamin D over the last 6 months

Exclusion criteria: renal, hepatic, other endocrine disorders, malignancies, pregnancy and lactation, alcohol consumption, menopause condition, receiving medications influencing vitamin D metabolism or insulin

Interventions

Intervention: vitamin D3 (cholecalciferol) 600,000 IU single intramuscular injection (n = 41)

Control: no intervention (n = 41)

All participants received pearl of vitamin E 400 IU/day for 1 month. Participants were followed up for 1 month.

Outcomes

Primary outcomes: changes in serum 25‐hydroxyvitamin D, serum adiponectin, HOMA‐IR, liver enzymes, and change in grade of NAFLD

Secondary outcomes: change in anthropometric variables

Stated aim of study

To examine the effect of single intramuscular injection of 600,000 IU of cholecalciferol on serum levels of vitamin D, adiponectin, insulin resistance, and liver function status of women with NAFLD

Notes

Study authors did not report any deaths. No information provided about adverse events.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The patients were randomly assigned in the random blocks of four subjects using a computer Random Allocation Software, version 1, with stratification by age."

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

The trial was registered in the Iranian Registry of Clinical Trials with code number of IRCT201503163320N10.

Other bias

Low risk

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

Hussain 2019

Study characteristics

Methods

Randomised double‐blind placebo‐controlled clinical trial with parallel‐group design (2 groups)

Participants

Country: Pakistan

Number of participants randomised: 109 (36% women), mean age 28 years

Inclusion criteria: randomised selection based on age, sex, BMI > 28, fatty liver on sonographic findings, moderate increase in hepatic enzymes with altered serum lipid profile

Exclusion criteria: pregnant and lactating women, smokers, type 2 diabetes, hypertension, chronic hepatitis B and C infection, alcoholic liver disease, chronic liver disease, decompensated liver disease and hepatocellular carcinoma, any history of cardiac, renal, and thyroid disorders, extremely abnormal ultrasound and liver enzymes that can indicate hereditary and autoimmune diseases of the liver

Interventions

Intervention: vitamin D3 (cholecalciferol) 50,000 IU (n = 54)

Control: placebo (n = 55)

Orally, weekly for 12 weeks

Outcomes

Primary outcomes: body weight, BMI, insulin resistance, dyslipidaemia, hepatic enzymes, CRP, and adiponectin

Stated aim of study

To investigate the effects of oral vitamin D supplementation on body weight, BMI, insulin resistance, dyslipidaemia, hepatic enzymes, CRP, and adiponectin in NAFLD patients

Notes

Study authors did not report any deaths. No information provided about adverse events.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "A computed generated number was given to each patients based upon randomization."

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

Unclear risk

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

Selective reporting (reporting bias)

Unclear risk

The trial was not registered. Unclear whether all predefined and clinically relevant and reasonably expected outcomes were reported

Other bias

Low risk

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

Jeong 2019

Study characteristics

Methods

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

Participants

Country: Republic of Korea

Number of participants randomised: 148 (49% women), aged 20 to 75 years, mean age 52 years

Inclusion criteria: age 20 to 75 years, positive in hepatitis C virus‐RNA PCR screening, normal serum calcium level before treatment, and HCV genotypes 1, 2, and 3

Exclusion criteria: HCC at enrolment or past history of HCC within last 1 year, decompensated cirrhosis (Child‐Pugh class B or C), absolute neutrophil count < 1000/mm3 or platelet count < 70,000/mm3, serum creatinine level above 1.5 times to upper normal limit, present or past history of severe psychiatric diseases, parathyroid disease, uncontrolled thyroid disease, co‐infection with other hepatitis virus or HIV, history of malignant diseases besides HCC within last 2 years, patients who were considered unfit to perform clinical trial, and pregnancy

Interventions

Intervention: PEG‐IFN‐α‐2a 180 μg weekly plus RBV daily plus vitamin D 800 IU daily (n = 77)

Control: PEG‐IFN‐α plus RBV (n = 71)

Ribavirin was given orally daily in a dose of 1000 mg (body weight < 75 kg) or 1200 mg (body weight ≥ 75 kg) daily in genotype 1, or 800 mg in genotype 2 and 3.

All participants were followed up 24 weeks after the completion of treatment.

Outcomes

Primary outcomes: rate of SVR

Secondary outcomes: change in risk factors for SVR

Stated aim of study

To assess the role of vitamin D supplementation in response to PEG‐IFN‐α plus RBV treatment in naive patients with chronic hepatitis C

Notes

Study authors did not report any deaths. “Serious adverse events occurred in 10 patients, but all recovered.”

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Study authors did not specify the method of sequence generation.

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

High risk

24 participants (33.8%) in the control group and 19 participants (24.7%) in the vitamin D group dropped out during the treatment period.

Selective reporting (reporting bias)

High risk

The study was registered at ClinicalTrials.gov (NCT01439776). No data about the early and rapid virological response

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias, such as competing interest bias.

Jha 2017

Study characteristics

Methods

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

Participants

Country: India

Number of participants randomised: 101 (24% women), aged 18 to 70 years, mean age 45 years

Inclusion criteria: decompensated cirrhosis of liver, Child Turcott Pugh (CTP) score ≥ 10, age between 18 years to 70 years

Exclusion criteria: septicaemia, infection with HIV, episodes of variceal bleeding within 6 weeks, hepatocellular carcinoma or any malignancy, hepatorenal syndrome at the time of enrolment, significant cardiac and respiratory disease, pregnancy, patients being taken up for transplant, and refusal to participate in the study

Interventions

Intervention: vitamin D (cholecalciferol 300,000 IU) intramuscularly, single dose plus 800 IU and calcium 1000 mg daily (n = 51)

Control: no intervention (n = 50)

For 6 months

Outcomes

Primary outcomes: all‐cause mortality

Secondary outcomes: clinical parameters, vitamin D level

Stated aim of study

To assess vitamin D levels in a cohort of patients with decompensated liver cirrhosis and the effect of vitamin D level replenishment on all‐cause mortality in patients with vitamin D deficient decompensated cirrhosis

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Study authors did not specify the method of sequence generation.

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 the outcome is likely to have been 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 have been 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

The protocol was not available.

Other bias

Low risk

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

Komolmit 2017a

Study characteristics

Methods

Randomised double‐blind, placebo‐controlled clinical trial with parallel‐group design (2 groups)

Participants

Country: Thailand

Number of participants randomised: 80 (46% women), aged 18 to 70 years, mean age 52 years

Inclusion criteria: patients with CHC

Exclusion criteria: none stated

Interventions

Intervention: vitamin D2 (ergocalciferol 60,000 to 100,000 IU (depending on vitamin D status)) (n = 40)

Control: placebo (n = 40)

Orally, weekly for 6 weeks

The dose of vitamin D2 was based on the ranges of vitamin D deficiency, as follows: mild deficiency (20 to ≤ 30 ng/mL) 60,000 IU/week; moderate deficiency (10 to ≤ 20 ng/mL) 80,000 IU/week; and severe deficiency (≤ 10 ng/mL) 100,000 IU/week. Each vitamin D2 capsule contained 20,000 units. The total dosage was divided into 2 separate doses given on Monday and Friday.

Outcomes

Primary outcomes: T‐helper 1/2 cytokines, IP‐10 and DPP‐IV levels

Secondary outcomes: vitamin D level

Stated aim of study

To assess the changes in serum levels T‐helper cells associated cytokines, IP‐10 and DPP‐IV, without influences driven by interferon treatment, after a short‐term period for correction of vitamin D deficiency in chronic hepatitis C patients

Notes

All participants completed the trial. The trial was conducted between April 2012 and April 2013.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomization sequence was stratified with a 1:1 allocation using random block sizes of 4 based on computer generated method based (www.randomisation.com)."

Allocation concealment (selection bias)

Low risk

Quote: "The randomization was performed by a research assistant without involvement in clinical trial. Details of the allocated group were given in sequentially numbered, opaque, sealed envelopes. After patient enrollment, the research assistant will open the envelope and inform stratified groups (A or B) to the investigators."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "The vitamin D2 (Ergocalciferol) and placebo were prepared by a pharmacist in a capsule form and identical in appearance. They were prepacked in a bottle for six‐week supplement and consecutively numbered for each CHC patient according to the randomised results."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and it is 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. All participants completed the trial.

Selective reporting (reporting bias)

Low risk

The trial was registered in the Thai Clinical Trials Registry, which was based on World Health Organization criteria under registration number TCTR20160429001.

Other bias

Unclear risk

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

Komolmit 2017b

Study characteristics

Methods

Randomised double‐blind, placebo‐controlled clinical trial with parallel‐group design (2 groups)

Participants

Country: Thailand

Number of participants randomised: 58 (38% women), aged 18 to 70 years, mean age 50 years

Inclusion criteria: patients with CHC, naive cases or non‐responder cases of CHC without decompensated cirrhosis, and serum 25‐hydroxyvitamin D levels less than 30 ng/mL

Exclusion criteria: decompensated cirrhosis, HIV infection, autoimmune diseases, active infections from other pathogens, a history of steroid or immunosuppressive therapy, or a history of interferon treatment within 12 months

Interventions

Intervention: vitamin D2 (ergocalciferol 20,000 IU orally, weekly) (n = 29)

Control: placebo (n = 29)

For 6 weeks

The dose of vitamin D2 was based on the ranges of vitamin D deficiency, as follows: mild deficiency (20 to ≤ 30 ng/mL) 60,000 IU/week; moderate deficiency (10 to ≤ 20 ng/mL) 80,000 IU/week; and severe deficiency (≤ 10 ng/mL) 100,000 IU/week. Each vitamin D2 capsule contained 20,000 units. The total dosage was divided into 2 separate doses given on Monday and Friday.

Outcomes

Primary outcomes: serum fibrotic markers

Secondary outcomes: vitamin D level

Stated aim of study

To assess the dynamic changes in serum fibrogenic cytokines/enzymes in CHC patients with vitamin D deficiency after short‐term supplementation with vitamin D

Notes

All participants completed the trial. The trial was conducted between February and December 2014.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The enrolled patients were randomized into two groups with a 1:1 allocation using a random block size of 4, which was generated by computer software based on www.randomisation.com."

Allocation concealment (selection bias)

Low risk

Quote: "The randomization was performed by a research assistant who was not involved in the clinical trial. Upon enrollment, the patients were stratified into two groups (A and B) with the details of the drugs placed in sealed envelopes."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Vitamin D2 and a placebo were prepared in identical capsules of the same weight by the Department of Pharmacy at King Chulalongkorn Memorial Hospital. All investigators and patients were blinded to the type of medication used until the end of the study."

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and it is 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)

Low risk

The clinical trial was retrospectively registered with the Thai Clinical Trials Registry, based on World Health Organization criteria on 2 November 2016 (TCTR20161103003).

Other bias

Unclear risk

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

Lorvand Amiri 2016

Study characteristics

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 cholecalciferol (Jalinus Arya Co, Iran) + calcium carbonate placebo (25 mg/day as lactose; Jalinus Arya Co, Iran) (n = 40)

Intervention 2: vitamin D 25 μg/day as cholecalciferol (Jalinus Arya Co, Iran) + calcium (500 mg/day as calcium carbonate; Jalinus Arya Co, Iran) (n = 40)

Control: placebo of calcitriol + placebo of calcium (25 mg/day as lactose; Jalinus Arya Co, Iran) (n = 40)

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 the 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

Study authors did not report any deaths. No information provided about adverse events. Clinical trial registered at Iranian Registry of Clinical Trials (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 permit judgement of 'low risk' or 'high risk'

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 in full.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias, such as competing interest bias.

Mobarhan 1984

Study characteristics

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 is likely to have been 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 have been 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.

Other bias

Low risk

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

Nimer 2012

Study characteristics

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 the influence of vitamin D supplementation on SVR in the treatment of people with chronic HCV with HCV genotype 2‐3

Notes

All participants completed the trial. 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 are likely to have been influenced by lack of blinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding, and outcome measurements are likely to have been 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

Other bias

Low risk

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

Pilz 2016

Study characteristics

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/min/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 normalised 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 the 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

“No patient died during the study and there was no excess of adverse events in the vitamin D group.”

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 is unlikely that blinding could have been broken.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and it is 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.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could put it at risk of bias, such as small‐trial bias.

Sakpal 2017

Study characteristics

Methods

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

Participants

Country: India

81 participants (32% women), aged > 12 years, mean age 38 years with NAFLD

Inclusion criteria: patients with NAFLD aged > 12 years, non‐alcoholic individuals defined as either total abstainers or individuals who consumed less than 20 g of alcohol per day, ultrasound showing features of steatosis, with or without raised ALT (> 40 IU/L), and negative viral markers (hepatitis B surface antigen and anti‐hepatitis C virus), patients with raised ALT who had negative autoimmune markers (antinuclear antibody, anti‐smooth muscle antibody, anti‐liver kidney microsomal antibody, and antimitochondrial antibody) and normal ceruloplasmin/negative Kayser–Fleischer ring with normal iron workup (serum iron, total iron‐binding capacity, ferritin, and transferrin saturation)

Exclusion criteria: pregnant females, patients with a history of drug intake likely to cause NAFLD, patients with jejunoileal bypass or extensive small bowel resection or total parenteral nutrition at the time of liver biopsy, and those with clinical, laboratory, and imaging features of cirrhosis of liver and patients with renal, hepatic, respiratory, or congestive cardiac failure

Interventions

Intervention: vitamin D single intramuscular injection 600,000 IU with lifestyle modifications (n = 51)

Control: lifestyle modifications (n = 30)

Participants were treated and followed up for 6 months.

Lifestyle modifications in both groups included moderate‐to‐vigorous exercise in the form of brisk walking, jogging, swimming, cycling, etc. for 45 to 60 min at least 5 days per week in all participants, and calorie reduction (1000 to 1200 kcal/day for overweight women and 1200 to 1600 kcal/day for overweight men and heavier or more active women) in overweight and obese subjects.

Outcomes

Primary outcomes: insulin resistance and serum ALT

Secondary outcomes: serum levels of adiponectin and tumour necrosis factor‐α

Stated aim of study

To evaluate the effect of vitamin D supplementation in patients with NAFLD

Notes

All participants completed the trial.

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 is likely to have been 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 have been influenced by lack of blinding

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

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

Selective reporting (reporting bias)

Unclear risk

Trial protocol was not available.

Other bias

Unclear risk

Trial may or may not have been free of other factors that could have put it at risk of bias, such as competing interest bias.

Sharifi 2014

Study characteristics

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 units/L for women and 30 units/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 the effect of vitamin D supplementation on serum liver enzymes, insulin resistance, oxidative stress, and inflammatory biomarkers in people with NAFLD

Notes

Study authors did not report any deaths. “Participants did not report any adverse or side effects such as hypercalcemia.”

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. Another person, who was not involved in the trial and not aware of random sequences, assigned the participants 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 is unlikely that the blinding could have been broken.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Blinding of outcome assessment ensured, and it is 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 was unclear whether all predefined and clinically relevant and reasonably expected outcomes were reported.

Other bias

Low risk

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

Shiomi 1999a

Study characteristics

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

Notes

All participants completed the trial. 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 is likely to have been influenced by lack of blinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of outcome assessment, and outcome measurement is likely to have been 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 were reported in full.

Other bias

Unclear risk

Trial may or may not have been free of other components that could put it at risk of bias, such as competing interest bias.

Shiomi 1999b

Study characteristics

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

Notes

All participants completed the trial. 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 is likely to have been influenced by lack of blinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of outcome assessment, and outcome measurement is likely to have been 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 in full.

Other bias

Unclear risk

Trial may or may not have been free of other components that could put it at risk of bias, such as competing interest bias.

Taghvaei 2018

Study characteristics

Methods

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

Participants

40 participants (50% women), aged between 30 to 70 years, mean age 42 years with NAFLD

Inclusion criteria: age above 18 years, increased levels of alanine aminotransferase level (more than 30 in women and 40 in men), a diagnosis of NAFLD and ruling out other causes of increased liver enzymes, and vitamin D level less than 30 ng/mL

Exclusion criteria: the presence of liver cirrhosis, pregnancy and lactation, alcohol consumption, drug abuse, administration of vitamins in the last 6 months and during the study, weight loss more than 5% over a year before entering the study, diabetes mellitus, weight‐lowering medications, hypercalcaemia, chronic kidney disease, end‐stage heart and lung disease, history of hyperlipidaemia which required treatment, history of medical therapies with impact on liver enzymes such as acetaminophen, statins, azathioprine, antibiotics such as sulfonamide and penicillin, amiodarone, methotrexate, anticonvulsants, isoniazid, steroids, and herbal medicines

Interventions

Intervention: vitamin D3 50,000 IU weekly plus lifestyle modification (n = 20)

Control: lifestyle modification (n = 20)

For 12 weeks. Participants were followed up for 6 months.

Outcomes

Primary outcomes: biochemical indices (ALT, AST, alkaline phosphatase, fasting blood sugar, triglycerides, cholesterol, LDL, creatinine, calcium, vitamin D) and level of liver steatosis on the basis of CAP score and fibrosis

Secondary outcomes: none stated

Stated aim of study

"Investigating the effects of vitamin D on NAFLD."

Notes

All participants completed the trial.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Random allocation of the patients in each group was performed using a random number table."

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

Quote: "Subjects and all study staffs were blinded to treatment group assignments."

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

The research was also registered in Iranian Registry of Clinical Trials (2015102624725n1).

Other bias

Unclear risk

Trial may or may not have been free of other components that could put it at risk of bias, such as competing interest bias.

Vosoghinia 2016

Study characteristics

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: absolute neutrophil count above 1500/mm3, platelet count above 90,000/mm3, and 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), a history of diarrhoea or malabsorption syndromes such as coeliac and chronic pancreatitis, or those with renal or parathyroid diseases

Interventions

Intervention: PEG‐IFN‐α‐2a (180 μg) + oral ribavirin (Rebetol, MSD), at dosage determined based on participant’s weight and genotype, was administered for 48 weeks in participants 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), at dosage determined based on participant’s weight and genotype

PEG‐IFN‐α‐2a was administered for 48 weeks in participants 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 PEG‐IFN/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 the outcome is likely to have been 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 have been 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

Registered under Iranian Registry of Clinical Trials Identifier no. IRCT201408312709N29. All predefined outcomes reported in full.

Other bias

Unclear risk

Trial may or may not have been free of other components that could put it at risk of bias, such as competing interest bias.

Xing 2013

Study characteristics

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 the effects of calcitriol on acute cellular rejection rate of liver transplant recipients

Notes

All participants completed the trial. Study sponsored by grants from Shanghai Nature Science Fund project and 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

Other bias

Low risk

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

Yokoyama 2014

Study characteristics

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

No serious side effects were observed.

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 is likely to have been influenced by lack of blinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

No blinding of outcome assessment, and outcome measurement is likely to have been 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

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
CAP: controlled attenuation parameter
CHC: chronic hepatitis C
CRP: C‐reactive protein
DPP‐IV: dipeptidyl peptidase IV
EVR: early virological response
GGT: gamma‐glutamyl transferase
HBs: hepatitis B surface
HBV: hepatitis B virus
HCC: hepatocellular carcinoma
HCV: hepatitis C virus
HDL: high‐density lipoprotein
HDV: hepatitis D virus
HOMA‐IR: homeostatic model assessment for insulin resistance
IFN: interferon
IP‐10: interferon gamma‐induced protein 10
IU: international unit
kcal: kilocalorie
LDL: low‐density lipoprotein
MRI: magnetic resonance imaging
n: number of participants
NAFLD: non‐alcoholic fatty liver disease
NASH: non‐alcoholic steatohepatitis
PCR: polymerase chain reaction
PEG: pegylated
RBV: ribavirin
RNA: ribonucleic acid
RVR: rapid viral response
SAF: steatosis‐activity‐fibrosis
SVR: sustained virological response
ULN: upper limit of normal
US: ultrasound
VD: vitamin D
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

Chen 2015

Not a randomised trial

Dasarathy 2017

Not a randomised trial

Fernández Fernández 2016

Not a randomised trial

Floreani 2007

Not a randomised trial

Hasanain 2018

Did not fulfil inclusion criteria. This trial included antituberculosis therapy‑induced liver disorders amongst naive patients with pulmonary tuberculosis.

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

Naderpoor 2018

Did not fulfil inclusion criteria. This trial included overweight or obese adults without chronic liver diseases.

Omori‐Mizuno 2015

Not a randomised trial

Papapostoli 2016

Not a randomised trial

Park 2017

Not a randomised trial

Rode 2010

Not a randomised trial

Stokes 2016

Not a randomised trial

Tavakoli 2019

Did not fulfil inclusion criteria. This trial included adolescent girls.

Terrier 2015

Not a randomised trial

Zhou 2019

Not a randomised trial

Characteristics of ongoing studies [ordered by study ID]

IRCT2016020326342N1

Study name

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

Study name

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, co‐infection with hepatitis C or D virus or HIV, other serious concurrent illness (e.g. alcoholism, uncontrolled diabetes, 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

1.1 1.1 All‐cause mortality Show forest plot

27

1979

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.51, 1.45]

Analysis 1.1

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 1: 1.1 All‐cause mortality

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 1: 1.1 All‐cause mortality

1.1.1 Non‐alcoholic fatty liver disease

11

803

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.1.2 Chronic hepatitis C

10

836

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 3.13]

1.1.3 Liver cirrhosis

5

265

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.53, 1.55]

1.1.4 Liver transplant recipients

1

75

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.2 1.1 All‐cause mortality according to vested interest Show forest plot

27

1979

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.51, 1.45]

Analysis 1.2

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 2: 1.1 All‐cause mortality according to vested interest

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 2: 1.1 All‐cause mortality according to vested interest

1.2.1 Trials with vested interest

2

38

Risk Ratio (M‐H, Random, 95% CI)

2.69 [0.15, 48.64]

1.2.2 Trials without vested interest

25

1941

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.48, 1.41]

1.3 All‐cause mortality according to vitamin D status at entry Show forest plot

27

1979

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.51, 1.45]

Analysis 1.3

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 3: All‐cause mortality according to vitamin D status at entry

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 3: All‐cause mortality according to vitamin D status at entry

1.3.1 Normal vitamin D status

8

549

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 3.13]

1.3.2 Low vitamin D status

19

1430

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.53, 1.55]

1.4 All‐cause mortality according to form of vitamin D Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 4: All‐cause mortality according to form of vitamin D

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 4: All‐cause mortality according to form of vitamin D

1.4.1 Vitamin D 3

20

1578

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.48, 1.41]

1.4.2 Vitamin D 2

3

150

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.15, 61.74]

1.4.3 1,25‐dihydroxyvitamin D

4

291

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.4.4 25‐hydroxyvitamin D

1

12

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.15, 61.74]

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

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 5: All‐cause mortality (best‐worst‐case and worst‐best‐case scenarios)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 5: All‐cause mortality (best‐worst‐case and worst‐best‐case scenarios)

1.5.1 Best‐worst‐case scenario

24

1737

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.06, 0.30]

1.5.2 Worst‐best‐case scenario

24

1737

Risk Ratio (M‐H, Random, 95% CI)

7.95 [3.55, 17.77]

1.6 Liver‐related mortality Show forest plot

1

18

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.08, 34.66]

Analysis 1.6

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 6: Liver‐related mortality

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 6: Liver‐related mortality

1.7 Serious adverse events Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 7: Serious adverse events

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 7: Serious adverse events

1.7.1 Hypercalcaemia

1

76

Risk Ratio (M‐H, Random, 95% CI)

5.00 [0.25, 100.80]

1.7.2 Myocardial infarction

2

86

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.08, 6.81]

1.7.3 Thyroiditis

1

68

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.91]

1.7.4 Circular haemorrhoidal prolapse

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.7.5 Bronchopneumonia

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.8 Liver‐related morbidity Show forest plot

0

0

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

Analysis 1.8

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 8: Liver‐related morbidity

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 8: Liver‐related morbidity

1.9 Health‐related quality of life Show forest plot

0

0

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

Analysis 1.9

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 9: Health‐related quality of life

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 9: Health‐related quality of life

1.10 Non‐serious adverse events Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.10

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 10: Non‐serious adverse events

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 10: Non‐serious adverse events

1.10.1 Glossitis

1

65

Risk Ratio (M‐H, Random, 95% CI)

3.70 [0.16, 87.58]

1.10.2 Depression

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.3 Lower back pain

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.4 Abdominal bloating

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.5 Cold

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.6 Constipation

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.7 Sore throat

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.8 Sour taste in mouth

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.9 Contused lacerated wound

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.10 Multiple white matter lesions

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.11 Gastro‐oesophageal reflux

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.12 Abdominal menstrual cramps

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.13 Tubular colon adenoma

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.14 Gastric motility disturbance

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.15 Irritable bowel syndrome

1

20

Risk Ratio (M‐H, Random, 95% CI)

5.00 [0.27, 92.62]

1.10.16 Knee pain

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.17 Severe allergy

1

109

Risk Ratio (M‐H, Random, 95% CI)

5.09 [0.25, 103.64]

1.11 Failure of rapid virological response Show forest plot

3

247

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.60, 0.95]

Analysis 1.11

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 11: Failure of rapid virological response

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 11: Failure of rapid virological response

1.12 Failure of early virological response Show forest plot

4

315

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.11, 1.00]

Analysis 1.12

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 12: Failure of early virological response

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 12: Failure of early virological response

1.13 Failure of sustained virological response Show forest plot

7

630

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.42, 1.01]

Analysis 1.13

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 13: Failure of sustained virological response

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 13: Failure of sustained virological response

1.14 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 1.14

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 14: Acute cellular rejection in liver transplant recipients

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 14: Acute cellular rejection in liver transplant recipients

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

15

1078

Mean Difference (IV, Random, 95% CI)

18.49 [14.52, 22.47]

Analysis 1.15

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 15: Vitamin D status (ng/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 15: Vitamin D status (ng/mL)

1.16 Bone mineral density (g/cm) Show forest plot

1

18

Mean Difference (IV, Random, 95% CI)

0.15 [0.04, 0.26]

Analysis 1.16

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 16: Bone mineral density (g/cm)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 16: Bone mineral density (g/cm)

1.17 Aspartate aminotransferase (IU/L) Show forest plot

12

774

Mean Difference (IV, Random, 95% CI)

‐1.75 [‐5.41, 1.91]

Analysis 1.17

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 17: Aspartate aminotransferase (IU/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 17: Aspartate aminotransferase (IU/L)

1.18 Alanine aminotransferase (IU/L Show forest plot

13

855

Mean Difference (IV, Random, 95% CI)

‐2.30 [‐7.60, 3.00]

Analysis 1.18

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 18: Alanine aminotransferase (IU/L

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 18: Alanine aminotransferase (IU/L

1.19 Alkaline phosphatases (IU/L) Show forest plot

6

344

Mean Difference (IV, Random, 95% CI)

‐0.95 [‐15.10, 13.20]

Analysis 1.19

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 19: Alkaline phosphatases (IU/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 19: Alkaline phosphatases (IU/L)

1.20 Gamma‐glutamyl transpeptidase (IU/L) Show forest plot

4

227

Mean Difference (IV, Random, 95% CI)

‐2.69 [‐5.26, ‐0.11]

Analysis 1.20

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 20: Gamma‐glutamyl transpeptidase (IU/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 20: Gamma‐glutamyl transpeptidase (IU/L)

1.21 Bilirubin (mg/dL) Show forest plot

3

74

Mean Difference (IV, Random, 95% CI)

0.32 [0.00, 0.63]

Analysis 1.21

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 21: Bilirubin (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 21: Bilirubin (mg/dL)

1.22 Triglyceride (mg/dL) Show forest plot

5

460

Mean Difference (IV, Random, 95% CI)

11.27 [‐10.99, 33.53]

Analysis 1.22

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 22: Triglyceride (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 22: Triglyceride (mg/dL)

1.23 Cholesterol (mg/dL) Show forest plot

4

400

Mean Difference (IV, Random, 95% CI)

3.51 [‐2.83, 9.85]

Analysis 1.23

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 23: Cholesterol (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 23: Cholesterol (mg/dL)

1.24 LDL cholesterol (mg/dL) Show forest plot

4

400

Mean Difference (IV, Random, 95% CI)

‐0.97 [‐8.70, 6.76]

Analysis 1.24

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 24: LDL cholesterol (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 24: LDL cholesterol (mg/dL)

1.25 Albumin (g/L) Show forest plot

3

74

Mean Difference (IV, Random, 95% CI)

‐1.18 [‐2.96, 0.59]

Analysis 1.25

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 25: Albumin (g/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 25: Albumin (g/L)

1.26 HDL cholesterol (mg/dL) Show forest plot

4

400

Mean Difference (IV, Random, 95% CI)

1.14 [‐0.64, 2.92]

Analysis 1.26

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 26: HDL cholesterol (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 26: HDL cholesterol (mg/dL)

1.27 Calcium (mg/dL) Show forest plot

7

423

Mean Difference (IV, Random, 95% CI)

0.04 [‐0.12, 0.19]

Analysis 1.27

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 27: Calcium (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 27: Calcium (mg/dL)

1.28 Glucose (mg/dL) Show forest plot

6

469

Mean Difference (IV, Random, 95% CI)

1.44 [‐5.05, 7.94]

Analysis 1.28

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 28: Glucose (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 28: Glucose (mg/dL)

1.29 Phosphorus (mg/dL) Show forest plot

4

307

Mean Difference (IV, Random, 95% CI)

0.17 [‐0.16, 0.50]

Analysis 1.29

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 29: Phosphorus (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 29: Phosphorus (mg/dL)

1.30 Adiponectin (µg/mL) Show forest plot

4

276

Mean Difference (IV, Random, 95% CI)

1.02 [‐0.27, 2.30]

Analysis 1.30

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 30: Adiponectin (µg/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 30: Adiponectin (µg/mL)

1.31 Insulin (mIU/mL) Show forest plot

6

428

Mean Difference (IV, Random, 95% CI)

0.03 [‐1.15, 1.21]

Analysis 1.31

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 31: Insulin (mIU/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 31: Insulin (mIU/mL)

1.32 Parathyroid hormone (pg/mL) Show forest plot

2

118

Mean Difference (IV, Random, 95% CI)

‐15.18 [‐38.54, 8.18]

Analysis 1.32

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 32: Parathyroid hormone (pg/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 32: Parathyroid hormone (pg/mL)

1.33 C‐reactive protein (mg/L) Show forest plot

4

254

Mean Difference (IV, Random, 95% CI)

‐0.50 [‐0.93, ‐0.07]

Analysis 1.33

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 33: C‐reactive protein (mg/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 33: C‐reactive protein (mg/L)

Study flow diagram.

Figuras y tablas -
Figure 1

Study flow diagram.

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

Figuras y tablas -
Figure 2

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

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

Figuras y tablas -
Figure 3

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

All‐cause mortality.Trial Sequential Analysis was performed based on a mortality in the control group of 2%, a relative risk reduction of 20% in the experimental intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). There was no diversity. The required information size was 63,116 participants. The cumulative Z‐curve (blue line) did not cross the trial sequential monitoring boundary for benefit or harm after the 27th trial. The trial sequential monitoring boundaries were ignored due to little information (3.14%). The blue line represents the cumulative Z‐score of the meta‐analysis. The green dotted lines represent the conventional statistical boundaries.

Figuras y tablas -
Figure 4

All‐cause mortality.

Trial Sequential Analysis was performed based on a mortality in the control group of 2%, a relative risk reduction of 20% in the experimental intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). There was no diversity. The required information size was 63,116 participants. The cumulative Z‐curve (blue line) did not cross the trial sequential monitoring boundary for benefit or harm after the 27th trial. The trial sequential monitoring boundaries were ignored due to little information (3.14%). The blue line represents the cumulative Z‐score of the meta‐analysis. The green dotted lines represent the conventional statistical boundaries.

Rapid virological response.Trial Sequential Analysis was performed based on a failure of rapid virological response in the control group of 53%, a relative risk reduction (RRR) of 20% in the intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). There was no diversity. The required information size was 1269 participants. The cumulative Z‐curve (blue line) crossed the conventional monitoring boundary for benefit but did not cross the trial sequential monitoring boundary for benefit (red down‐sloping line). The blue line represents the cumulative Z‐score of the meta‐analysis. The green dotted lines represent the conventional statistical boundaries. The red inward‐sloping lines represent the trial sequential monitoring boundaries.

Figuras y tablas -
Figure 5

Rapid virological response.

Trial Sequential Analysis was performed based on a failure of rapid virological response in the control group of 53%, a relative risk reduction (RRR) of 20% in the intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). There was no diversity. The required information size was 1269 participants. The cumulative Z‐curve (blue line) crossed the conventional monitoring boundary for benefit but did not cross the trial sequential monitoring boundary for benefit (red down‐sloping line). The blue line represents the cumulative Z‐score of the meta‐analysis. The green dotted lines represent the conventional statistical boundaries. The red inward‐sloping lines represent the trial sequential monitoring boundaries.

Early virological response.Trial Sequential Analysis was performed based on failure of early virological response in the control group of 34%, a relative risk reduction of 20% in the intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). The diversity was 88%. The required information size was 21,306 participants. The cumulative Z‐curve (blue line) crossed the conventional monitoring boundary for benefit. The trial sequential monitoring boundary was ignored due to little information (1.48%). The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries.

Figuras y tablas -
Figure 6

Early virological response.

Trial Sequential Analysis was performed based on failure of early virological response in the control group of 34%, a relative risk reduction of 20% in the intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). The diversity was 88%. The required information size was 21,306 participants. The cumulative Z‐curve (blue line) crossed the conventional monitoring boundary for benefit. The trial sequential monitoring boundary was ignored due to little information (1.48%). The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries.

Sustained virological response.Trial Sequential Analysis was performed based on failure of sustained virological response in the control group of 48%, a relative risk reduction (RRR) of 20% in the intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). Diversity was 80%. The required information size was 7570 participants. The cumulative Z‐curve (blue line) crossed the conventional monitoring boundary for benefit. However, it did not cross any of the monitoring boundaries for benefit, harm, or futility. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries. The red inward‐sloping lines represent the trial sequential monitoring boundaries for benefit and harm.

Figuras y tablas -
Figure 7

Sustained virological response.

Trial Sequential Analysis was performed based on failure of sustained virological response in the control group of 48%, a relative risk reduction (RRR) of 20% in the intervention group, a type I error of 1.25%, and a type II error of 10% (90% power). Diversity was 80%. The required information size was 7570 participants. The cumulative Z‐curve (blue line) crossed the conventional monitoring boundary for benefit. However, it did not cross any of the monitoring boundaries for benefit, harm, or futility. The blue line represents the cumulative Z‐score of the meta‐analysis. The green lines represent the conventional statistical boundaries. The red inward‐sloping lines represent the trial sequential monitoring boundaries for benefit and harm.

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 1: 1.1 All‐cause mortality

Figuras y tablas -
Analysis 1.1

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 1: 1.1 All‐cause mortality

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 2: 1.1 All‐cause mortality according to vested interest

Figuras y tablas -
Analysis 1.2

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 2: 1.1 All‐cause mortality according to vested interest

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 3: All‐cause mortality according to vitamin D status at entry

Figuras y tablas -
Analysis 1.3

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 3: All‐cause mortality according to vitamin D status at entry

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 4: All‐cause mortality according to form of vitamin D

Figuras y tablas -
Analysis 1.4

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 4: All‐cause mortality according to form of vitamin D

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 5: All‐cause mortality (best‐worst‐case and worst‐best‐case scenarios)

Figuras y tablas -
Analysis 1.5

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 5: All‐cause mortality (best‐worst‐case and worst‐best‐case scenarios)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 6: Liver‐related mortality

Figuras y tablas -
Analysis 1.6

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 6: Liver‐related mortality

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 7: Serious adverse events

Figuras y tablas -
Analysis 1.7

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 7: Serious adverse events

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 8: Liver‐related morbidity

Figuras y tablas -
Analysis 1.8

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 8: Liver‐related morbidity

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 9: Health‐related quality of life

Figuras y tablas -
Analysis 1.9

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 9: Health‐related quality of life

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 10: Non‐serious adverse events

Figuras y tablas -
Analysis 1.10

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 10: Non‐serious adverse events

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 11: Failure of rapid virological response

Figuras y tablas -
Analysis 1.11

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 11: Failure of rapid virological response

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 12: Failure of early virological response

Figuras y tablas -
Analysis 1.12

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 12: Failure of early virological response

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 13: Failure of sustained virological response

Figuras y tablas -
Analysis 1.13

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 13: Failure of sustained virological response

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 14: Acute cellular rejection in liver transplant recipients

Figuras y tablas -
Analysis 1.14

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 14: Acute cellular rejection in liver transplant recipients

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 15: Vitamin D status (ng/mL)

Figuras y tablas -
Analysis 1.15

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 15: Vitamin D status (ng/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 16: Bone mineral density (g/cm)

Figuras y tablas -
Analysis 1.16

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 16: Bone mineral density (g/cm)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 17: Aspartate aminotransferase (IU/L)

Figuras y tablas -
Analysis 1.17

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 17: Aspartate aminotransferase (IU/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 18: Alanine aminotransferase (IU/L

Figuras y tablas -
Analysis 1.18

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 18: Alanine aminotransferase (IU/L

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 19: Alkaline phosphatases (IU/L)

Figuras y tablas -
Analysis 1.19

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 19: Alkaline phosphatases (IU/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 20: Gamma‐glutamyl transpeptidase (IU/L)

Figuras y tablas -
Analysis 1.20

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 20: Gamma‐glutamyl transpeptidase (IU/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 21: Bilirubin (mg/dL)

Figuras y tablas -
Analysis 1.21

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 21: Bilirubin (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 22: Triglyceride (mg/dL)

Figuras y tablas -
Analysis 1.22

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 22: Triglyceride (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 23: Cholesterol (mg/dL)

Figuras y tablas -
Analysis 1.23

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 23: Cholesterol (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 24: LDL cholesterol (mg/dL)

Figuras y tablas -
Analysis 1.24

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 24: LDL cholesterol (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 25: Albumin (g/L)

Figuras y tablas -
Analysis 1.25

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 25: Albumin (g/L)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 26: HDL cholesterol (mg/dL)

Figuras y tablas -
Analysis 1.26

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 26: HDL cholesterol (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 27: Calcium (mg/dL)

Figuras y tablas -
Analysis 1.27

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 27: Calcium (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 28: Glucose (mg/dL)

Figuras y tablas -
Analysis 1.28

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 28: Glucose (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 29: Phosphorus (mg/dL)

Figuras y tablas -
Analysis 1.29

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 29: Phosphorus (mg/dL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 30: Adiponectin (µg/mL)

Figuras y tablas -
Analysis 1.30

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 30: Adiponectin (µg/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 31: Insulin (mIU/mL)

Figuras y tablas -
Analysis 1.31

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 31: Insulin (mIU/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 32: Parathyroid hormone (pg/mL)

Figuras y tablas -
Analysis 1.32

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 32: Parathyroid hormone (pg/mL)

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 33: C‐reactive protein (mg/L)

Figuras y tablas -
Analysis 1.33

Comparison 1: Vitamin D versus placebo or no intervention, Outcome 33: C‐reactive protein (mg/L)

Summary of findings 1. Vitamin D compared with placebo or no intervention for chronic liver diseases in adults

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

Patient or population: people with chronic liver diseases
Setting: in‐ and outpatients
Intervention: vitamin D
Comparison: placebo or no intervention

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(trials)

Certainty of the evidence
(GRADE)

Comments

Risk with placebo or no intervention

Risk with vitamin D

All‐cause mortality

Follow‐up: mean 7 months (1 to 18 months)

Study population

RR 0.86
(0.51 to 1.45)

1979
(27 RCTs)

⊕⊝⊝⊝
very low 1

21 per 1000

18 per 1000
(11 to 30)

Liver‐related mortality

Follow‐up: 12 months

Study population

RR 1.62
(0.08 to 34.66)

18
(1 RCT)

⊕⊝⊝⊝
very low 2

No information was available to calculate absolute effects.

Serious adverse events

Follow‐up: mean 10.5 months (6 to 12 months)

Study population

⊕⊝⊝⊝
very low 3

Several serious adverse events were reported: hypercalcaemia (RR 5.00, 95% CI 0.25 to 100.8; 1 trial; 76 participants); myocardial infarction (RR 0.75, 95% CI 0.08 to 6.81; 2 trials; 86 participants); thyroiditis (RR 0.33, 95% CI 0.01 to 7.91; 1 trial; 68 participants); circular haemorrhoidal prolapse (RR 3.00, 95% CI 0.14 to 65.9; 1 trial; 20 participants); bronchopneumonia (RR 0.33, 95% CI 0.02 to 7.32; 1 trial; 20 participants).

Liver‐related morbidity

Study population

(0 RCTs)

Health‐related quality of life

Study population

(0 RCTs)

Non‐serious adverse events

Follow‐up: mean 7 months (3 to 12 months)

Study population

⊕⊝⊝⊝
very low 3

1 trial reported 1 single non‐serious adverse event, and another trial reported 16 single non‐serious adverse events, for a total of 17 types of non‐serious adverse events.

Failure of sustained virological response

Follow‐up: mean 16 months (6 to 18 months)

Study population

RR 0.65
(0.42 to 1.01)

630
(7 RCTs)

⊕⊝⊝⊝
very low4

484 per 1000

315 per 1000
(203 to 489)

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

CI: confidence interval; RCT: randomised clinical trial; RR: risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: We are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1Downgraded because of risk of bias (1 level) (all trials were at high risk of bias); and imprecision (2 levels) (few events, and the optimal information size of 63,116 participants (based on a proportion of 2% in the control group, a relative risk reduction of 20%, an alpha of 1.25%, and a beta of 10%) was not met; wide CI which included both benefits and harms).
2Downgraded because of risk of bias (1 level) (the trial was at high risk of bias); and imprecision (2 levels) (very few events, and wide CI which included both benefits and harms).
3Downgraded because of risk of bias (1 level) (all trials were at high risk of bias); and imprecision (2 levels) (very few events, and wide CI which included both benefits and harms).
4Downgraded because of risk of bias (1 level) (all trials were at high risk of bias); imprecision (2 levels) (the optimal information size of 7570 participants (based on a proportion of 48% in the control group, a relative risk reduction of 20%, an alpha of 1.25%, and a beta of 10%) was not met); inconsistency (1 level) (considerable heterogeneity); and indirectness (3 levels)(sustained virological response is a surrogate outcome).

Figuras y tablas -
Summary of findings 1. Vitamin D compared with 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 group

2

High

NI

72

44

47

Atsukawa 2016

No

Parallel group

2

High

NI

115

50

64

Barchetta 2016

Yes

Parallel group

2

High

PL

65

35

59

Behera 2018

Yes

Parallel group

2

High

NI

60

40

41

Boonyagard 2016

No

Parallel group

2

High

PL

60

Dabbaghmanesh 2018

Yes

Parallel group

2

High

PL

106

59

45

Esmat 2015

No

Parallel group

2

High

NI

101

25

40

Foroughi 2016

Yes

Parallel group

2

High

PL

60

52

48

Geier 2018

Yes

Parallel group

2

High

PL

20

44

Hosseini 2018

Yes

Parallel group

2

High

NI

82

100

34

Hussain 2019

No

Parallel group

2

High

PL

109

36

28

Jeong 2019

Yes

Parallel group

2

High

NI

148

49

52

Jha 2017

No

Parallel group

2

High

NI

101

24

45

Komolmit 2017a

Yes

Parallel group

2

High

PL

80

46

52

Komolmit 2017b

Yes

Parallel group

2

High

PL

58

38

50

Lorvand Amiri 2016

Yes

Parallel group

3

High

PL

120

38

41

Mobarhan 1984

No

Parallel group

3

High

NI

18

0

61

Nimer 2012

No

Parallel group

2

High

NI

50

58

47

Pilz 2016

Yes

Parallel group

2

High

PL

36

25

61

Sakpal 2017

No

Parallel group

2

High

NI

81

32

38

Sharifi 2014

No

Parallel group

2

High

PL

60

51

60

Shiomi 1999a

No

Parallel group

2

High

NI

76

66

61

Shiomi 1999b

No

Parallel group

2

High

NI

34

100

56

Taghvaei 2018

Yes

Parallel group

2

High

NI

40

50

42

Vosoghinia 2016

Yes

Parallel group

2

High

NI

68

13

42

Xing 2013

No

Parallel group

3

High

PL

75

17

48

Yokoyama 2014

No

Parallel group

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

Behera 2018

Chronic hepatitis C genotype 1, 4

Sustained virological response

No

India

Boonyagard 2016

NAFLD

Biochemical indices, HOMA, FibroScan measurement

No information

Thailand

Dabbaghmanesh 2018

NAFLD

Biochemical indices

No

Iran

Esmat 2015

Chronic hepatitis C genotype 4

Sustained virological response

No information

Egypt

Foroughi 2016

NAFLD

Liver steatosis, liver function

No

Iran

Geier 2018

NAFLD (NASH)

Liver steatosis, liver function

Yes

Switzerland

Hosseini 2018

NAFLD

Serum 25‐hydroxyvitamin D, adiponectin, HOMA‐IR, liver enzymes, and change in grade of NAFLD

No

Iran

Hussain 2019

NAFLD

Body weight, BMI, insulin resistance, dyslipidaemia, hepatic enzymes, CRP, and adiponectin

No information

Pakistan

Jeong 2019

Chronic hepatitis C genotype 1, 2, 3

Sustained virological response

No information

Republic of Korea

Jha 2017

Liver cirrhosis

Mortality

No information

India

Komolmit 2017a

Chronic hepatitis C

Serum levels of T‐helper cells associated cytokines

No

Thailand

Komolmit 2017b

Chronic hepatitis C

Serum fibrotic markers

No

Thailand

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

Sakpal 2017

NAFLD

Insulin resistance and serum ALT

No

India

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

Taghvaei 2018

NAFLD

Biochemical indices, liver steatosis

No information

Iran

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

ALT: alanine aminotransferase
BMI: body mass index
CRP: C‐reactive protein
HOMA‐IR: homeostatic model assessment for insulin resistance
NAFLD: non‐alcoholic fatty liver disease
NASH: non‐alcoholic steatohepatitis

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

Study ID

Vitamin

Calcium
(mg)

Route

Regimen

Treatment
(weeks)

Follow‐up
(weeks)

Co‐intervention

D3
(IU)

D2
(IU)

25(OH)D
(IU)

1,25(OH)2D
(µg)

Abu‐Mouch 2011

2000

Orally

Daily

48

72

PEG‐IFN, RBV

Atsukawa 2016

2000

Orally

Daily

16

24

PEG‐IFN, RBV, SP

Barchetta 2016

2000

Orally

Daily

24

24

Behera 2018

2000

Orally

Daily

48

48

PEG‐IFN, RBV

Boonyagard 2016

Orally

Daily

20

20

Dabbaghmanesh 2018

50,000

0.25

Orally

Weekly and daily

12

12

Esmat 2015

2143

Orally

Weekly

48

72

PEG‐IFN, RBV

Foroughi 2016

7143

Orally

Weekly

10

10

Geier 2018

2100

Orally

Daily

48

48

Hosseini 2018

600,000

Intramuscularly

Single dose

Single dose

4

Vitamin E 400 IU/day

Hussain 2019

50,000

Orally

Weekly

12

12

Jeong 2019

800

Orally

Daily

24, 48

48, 72

PEG‐IFN, RBV

Jha 2017

300,000; 800

1000

Intramuscularly and orally

Single dose; daily

24

24

Komolmit 2017a

60,000; 80,000; 100,000

Orally

Weekly

6

6

Komolmit 2017b

60,000; 80,000; 100,000

Orally

Weekly

6

6

Lorvand Amiri 2016

1000

500

Orally

Daily

10

12

Mobarhan 1984

17,857

2400

Orally

Daily

52

52

Nimer 2012

2000

Orally

Daily

24

48

PEG‐IFN, RBV

Pilz 2016

2800

Orally

Daily

8

8

Sakpal 2017

600,000

Intramuscularly

Single dose

Single dose

24

Sharifi 2014

3571

Orally

Twice a week

16

16

Shiomi 1999a

1

Orally

Daily

52

52

Shiomi 1999b

1

Orally

Daily

52

52

Taghvaei 2018

50,000

Orally

Weekly

12

72

Lifestyle modification

Vosoghinia 2016

1600

Orally

Daily

12

12

PEG‐IFN, RBV

Xing 2013

0.25

1000

Orally

Daily

4

4

Yokoyama 2014

1000

Orally

Daily

16

24

PEG‐IFN, RBV

1,25(OH)2D: calcitriol
25(OH)D: calcidiol
IU: international unit
PEG‐IFN: 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

1.1 1.1 All‐cause mortality Show forest plot

27

1979

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.51, 1.45]

1.1.1 Non‐alcoholic fatty liver disease

11

803

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.1.2 Chronic hepatitis C

10

836

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 3.13]

1.1.3 Liver cirrhosis

5

265

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.53, 1.55]

1.1.4 Liver transplant recipients

1

75

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.2 1.1 All‐cause mortality according to vested interest Show forest plot

27

1979

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.51, 1.45]

1.2.1 Trials with vested interest

2

38

Risk Ratio (M‐H, Random, 95% CI)

2.69 [0.15, 48.64]

1.2.2 Trials without vested interest

25

1941

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.48, 1.41]

1.3 All‐cause mortality according to vitamin D status at entry Show forest plot

27

1979

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.51, 1.45]

1.3.1 Normal vitamin D status

8

549

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 3.13]

1.3.2 Low vitamin D status

19

1430

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.53, 1.55]

1.4 All‐cause mortality according to form of vitamin D Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.4.1 Vitamin D 3

20

1578

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.48, 1.41]

1.4.2 Vitamin D 2

3

150

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.15, 61.74]

1.4.3 1,25‐dihydroxyvitamin D

4

291

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.4.4 25‐hydroxyvitamin D

1

12

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.15, 61.74]

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

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.5.1 Best‐worst‐case scenario

24

1737

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.06, 0.30]

1.5.2 Worst‐best‐case scenario

24

1737

Risk Ratio (M‐H, Random, 95% CI)

7.95 [3.55, 17.77]

1.6 Liver‐related mortality Show forest plot

1

18

Risk Ratio (M‐H, Random, 95% CI)

1.62 [0.08, 34.66]

1.7 Serious adverse events Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.7.1 Hypercalcaemia

1

76

Risk Ratio (M‐H, Random, 95% CI)

5.00 [0.25, 100.80]

1.7.2 Myocardial infarction

2

86

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.08, 6.81]

1.7.3 Thyroiditis

1

68

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.91]

1.7.4 Circular haemorrhoidal prolapse

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.7.5 Bronchopneumonia

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.8 Liver‐related morbidity Show forest plot

0

0

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.9 Health‐related quality of life Show forest plot

0

0

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.10 Non‐serious adverse events Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.10.1 Glossitis

1

65

Risk Ratio (M‐H, Random, 95% CI)

3.70 [0.16, 87.58]

1.10.2 Depression

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.3 Lower back pain

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.4 Abdominal bloating

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.5 Cold

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.6 Constipation

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.7 Sore throat

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.8 Sour taste in mouth

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.9 Contused lacerated wound

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.10 Multiple white matter lesions

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.02, 7.32]

1.10.11 Gastro‐oesophageal reflux

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.12 Abdominal menstrual cramps

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.13 Tubular colon adenoma

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.14 Gastric motility disturbance

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.15 Irritable bowel syndrome

1

20

Risk Ratio (M‐H, Random, 95% CI)

5.00 [0.27, 92.62]

1.10.16 Knee pain

1

20

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.14, 65.90]

1.10.17 Severe allergy

1

109

Risk Ratio (M‐H, Random, 95% CI)

5.09 [0.25, 103.64]

1.11 Failure of rapid virological response Show forest plot

3

247

Risk Ratio (M‐H, Random, 95% CI)

0.75 [0.60, 0.95]

1.12 Failure of early virological response Show forest plot

4

315

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.11, 1.00]

1.13 Failure of sustained virological response Show forest plot

7

630

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.42, 1.01]

1.14 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]

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

15

1078

Mean Difference (IV, Random, 95% CI)

18.49 [14.52, 22.47]

1.16 Bone mineral density (g/cm) Show forest plot

1

18

Mean Difference (IV, Random, 95% CI)

0.15 [0.04, 0.26]

1.17 Aspartate aminotransferase (IU/L) Show forest plot

12

774

Mean Difference (IV, Random, 95% CI)

‐1.75 [‐5.41, 1.91]

1.18 Alanine aminotransferase (IU/L Show forest plot

13

855

Mean Difference (IV, Random, 95% CI)

‐2.30 [‐7.60, 3.00]

1.19 Alkaline phosphatases (IU/L) Show forest plot

6

344

Mean Difference (IV, Random, 95% CI)

‐0.95 [‐15.10, 13.20]

1.20 Gamma‐glutamyl transpeptidase (IU/L) Show forest plot

4

227

Mean Difference (IV, Random, 95% CI)

‐2.69 [‐5.26, ‐0.11]

1.21 Bilirubin (mg/dL) Show forest plot

3

74

Mean Difference (IV, Random, 95% CI)

0.32 [0.00, 0.63]

1.22 Triglyceride (mg/dL) Show forest plot

5

460

Mean Difference (IV, Random, 95% CI)

11.27 [‐10.99, 33.53]

1.23 Cholesterol (mg/dL) Show forest plot

4

400

Mean Difference (IV, Random, 95% CI)

3.51 [‐2.83, 9.85]

1.24 LDL cholesterol (mg/dL) Show forest plot

4

400

Mean Difference (IV, Random, 95% CI)

‐0.97 [‐8.70, 6.76]

1.25 Albumin (g/L) Show forest plot

3

74

Mean Difference (IV, Random, 95% CI)

‐1.18 [‐2.96, 0.59]

1.26 HDL cholesterol (mg/dL) Show forest plot

4

400

Mean Difference (IV, Random, 95% CI)

1.14 [‐0.64, 2.92]

1.27 Calcium (mg/dL) Show forest plot

7

423

Mean Difference (IV, Random, 95% CI)

0.04 [‐0.12, 0.19]

1.28 Glucose (mg/dL) Show forest plot

6

469

Mean Difference (IV, Random, 95% CI)

1.44 [‐5.05, 7.94]

1.29 Phosphorus (mg/dL) Show forest plot

4

307

Mean Difference (IV, Random, 95% CI)

0.17 [‐0.16, 0.50]

1.30 Adiponectin (µg/mL) Show forest plot

4

276

Mean Difference (IV, Random, 95% CI)

1.02 [‐0.27, 2.30]

1.31 Insulin (mIU/mL) Show forest plot

6

428

Mean Difference (IV, Random, 95% CI)

0.03 [‐1.15, 1.21]

1.32 Parathyroid hormone (pg/mL) Show forest plot

2

118

Mean Difference (IV, Random, 95% CI)

‐15.18 [‐38.54, 8.18]

1.33 C‐reactive protein (mg/L) Show forest plot

4

254

Mean Difference (IV, Random, 95% CI)

‐0.50 [‐0.93, ‐0.07]

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