887
Views
11
CrossRef citations to date
0
Altmetric
Review

Anti-diabetic drugs and NASH: from current options to promising perspectives

, , & ORCID Icon
Pages 813-825 | Received 27 Apr 2021, Accepted 01 Jul 2021, Published online: 12 Jul 2021

References

  • Younossi ZM, Koenig AB, Abdelatif D, et al. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73–84.
  • Younossi ZM, Stepanova M, Younossi Y, et al. Epidemiology of chronic liver diseases in the USA in the past three decades. Gut. 2020;69(3):564–568.
  • Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018;67(1):328–357.
  • EASL-EASD-EASO. Clinical practice guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016;64(6):1388–1402.
  • Estes C, Anstee QM, Arias-Loste M, et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016-2030. J Hepatol. 2018;69(4):896–904.
  • Paik JM, Golabi P, Younossi Y, et al. The growing burden of disability related to nonalcoholic fatty liver disease: data from the global burden of disease 2007-2017. Hepatol Commun. 2020;4(12):1769–1780.
  • Cariou B, Byrne CD, Loomba R, Sanyal AJ. Nonalcoholic fatty liver disease as a metabolic disease in humans: a literature review. Diabetes Obes Metab 2021; Jan 19.
  • Wargny M, Smati S, Pichelin M, et al. Fatty liver index is a strong predictor of changes in glycemic status in people with prediabetes: the IT-DIAB study. PLoS One. 2019;14(8):e0221524.
  • Mantovani A, Byrne CD, Bonora E, et al. Nonalcoholic fatty liver disease and risk of incident type 2 diabetes: a meta-analysis. Diabetes Care. 2018;41(2):372–382.
  • Ciardullo S, Monti T, Perseghin G. High prevalence of advanced liver fibrosis assessed by transient elastography among U.S. adults with type 2 diabetes. Diabetes Care. 2021;44(2):519–525.
  • Bril F, Cusi K. Management of nonalcoholic fatty liver disease in patients with type 2 diabetes: a call to action. Diabetes Care. 2017;40(3):419–430.
  • Sumida Y, Yoneda M, Ogawa Y, et al. Current and new pharmacotherapy options for non-alcoholic steatohepatitis. Expert Opin Pharmacother. 2020;21(8):953–967.
  • Buse JB, Wexler DJ, Tsapas A, et al. 2019 update to: management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia. 2020;63(2):221–228.
  • He L. Metformin and systemic metabolism. Trends Pharmacol Sci. 2020;41(11):868–881.
  • Tiikkainen M, Häkkinen AM, Korsheninnikova E, et al. Effects of rosiglitazone and metformin on liver fat content, hepatic insulin resistance, insulin clearance, and gene expression in adipose tissue in patients with type 2 diabetes. Diabetes. 2004;53(8):2169–2176.
  • Cariou B, Byrne CD, Loomba R, et al. Nonalcoholic fatty liver disease as a metabolic disease in humans: a literature review. Diabetes Obes Metab. 2021 Jan 19;23(5):1069–1083.
  • Said A, Akhter A. Meta-analysis of randomized controlled trials of pharmacologic agents in non-alcoholic steatohepatitis. Ann Hepatol. 2017;16(4):538–547.
  • Vilar-Gomez E, Vuppalanchi R, Desai AP, et al. Long-term metformin use may improve clinical outcomes in diabetic patients with non-alcoholic steatohepatitis and bridging fibrosis or compensated cirrhosis. Aliment Pharmacol Ther. 2019;50(3):317–328.
  • Vilar-Gomez E, Calzadilla-Bertot L, Wong VW, et al. Type 2 diabetes and metformin use associate with outcomes of patients with nonalcoholic steatohepatitis-related, Child-Pugh A cirrhosis. Clin Gastroenterol Hepatol. 2021;19(1):136–145.
  • Gross B, Pawlak M, Lefebvre P, et al. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat Rev Endocrinol. 2017;13(1):36–49.
  • Cariou B, Charbonnel B, Thiazolidinediones SB. PPARgamma agonists: time for a reassessment. Trends Endocrinol Metab. 2012;23(5):205–215.
  • Musso G, Cassader M, Paschetta E, et al. Thiazolidinediones and advanced liver fibrosis in nonalcoholic steatohepatitis: a meta-analysis. JAMA Intern Med. 2017;177(5):633–640.
  • DeFronzo RA, Tripathy D, Schwenke DC, et al. Pioglitazone for diabetes prevention in impaired glucose tolerance. N Engl J Med. 2011;364(12):1104–1115.
  • Dormandy JA, Charbonnel B, Eckland DJA, et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomized controlled trial. Lancet. 2005;366(9493):1279–1289.
  • Basaranoglu M, Acbay O, Sonsuz A. A controlled trial of gemfibrozil in the treatment of patients with nonalcoholic steatohepatitis. J Hepatol. 1999;31(2):384.
  • Fernández-Miranda C, Pérez-Carreras M, Colina F, et al. A pilot trial of fenofibrate for the treatment of non-alcoholic fatty liver disease. Dig Liver Dis. 2008;40(3):200–205.
  • Sasaki Y, Asahiyama M, Tanaka T, et al. Pemafibrate, a selective PPARalpha modulator, prevents non-alcoholic steatohepatitis development without reducing the hepatic triglyceride content. Sci Rep. 2020;10(1):7818.
  • Araki E, Yamashita S, Arai H, et al. Effects of pemafibrate, a novel selective PPARalpha modulator, on lipid and glucose metabolism in patients with type 2 diabetes and hypertriglyceridemia: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care. 2018;41(3):538–546.
  • Pradhan AD, Paynter NP, Everett BM, et al. Rationale and design of the pemafibrate to reduce cardiovascular outcomes by reducing triglycerides in patients with diabetes (PROMINENT) study. Am Heart J. 2018;206:80–93.
  • Staels B, Rubenstrunk A, Noel B, et al. Hepatoprotective effects of the dual peroxisome proliferator-activated receptor alpha/delta agonist GFT505, in rodent models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology. 2013;58(6):1941–1952. .
  • Cariou B, Hanf R, Lambert-Porcheron S, et al. Dual peroxisome proliferator-activated receptor α/δ agonist GFT505 improves hepatic and peripheral insulin sensitivity in abdominally obese subjects. Diabetes Care. 2013;36(10):2923–2930.
  • Ratziu V, Harrison SA, Francque S, et al. Elafibranor, an agonist of the peroxisome proliferator-activated receptor alpha and -delta, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening. Gastroenterology. 2016;150(5):1147–1159.
  • Corp G. GENFIT: announces Results from Interim Analysis of RESOLVE-IT Phase 3 Trial of Elafibranor in Adults with NASH and Fibrosis. 2020; https://ir.genfit.com/news-releases/news-release-details/genfit-announces-results-interim-analysis-resolve-it-phase-3. Accessed 2021 Apr 5.
  • Therapeutics C. CymaBay therapeutics halts clinical development of seladelpar. 2019; https://www.globenewswire.com/news-release/2019/11/25/1951942/0/en/CymaBay-Therapeutics-Halts-Clinical-Development-of-Seladelpar.html. Accessed 2021 Apr 5.
  • Jain MR, Giri SR, Bhoi B, et al. Dual PPARα/γ agonist saroglitazar improves liver histopathology and biochemistry in experimental NASH models. Liver Int. 2018;38(6):1084–1094.
  • Kumar DP, Caffrey R, Marioneaux J, et al. The PPARα/γ agonist saroglitazar improves insulin resistance and steatohepatitis in a diet induced animal model of nonalcoholic fatty liver disease. Sci Rep. 2020;10(1):9330.
  • Gawrieh S, Loo N, Noureddin M. et al. Saroglitazar, a PPARα/γ/agonist, for treatment of nonalcoholic fatty liver disease: a randomized controlled double-blind phase 2 trial. Hepatology. 2021;Apr2 10/1002/hep.31843
  • Boubia B, Poupardin O, Barth M, et al. Design, Synthesis, and Evaluation of a Novel Series of Indole Sulfonamide Peroxisome Proliferator Activated Receptor (PPAR) α/γ/δ Triple Activators: discovery of Lanifibranor, a New Antifibrotic Clinical Candidate. J Med Chem. 2018;61(6):2246–2265.
  • Lefere S, Puengel T, Hundertmark J, et al. Differential effects of selective- and pan-PPAR agonists on experimental steatohepatitis and hepatic macrophages. J Hepatol. 2020;73(4):757–770.
  • Ruzehaji N, Frantz C, Ponsoye M, et al. Pan PPAR agonist IVA337 is effective in prevention and treatment of experimental skin fibrosis. Ann Rheum Dis. 2016;75(12):2175–2183.
  • Avouac J, Konstantinova I, Guignabert C, et al. Pan-PPAR agonist IVA337 is effective in experimental lung fibrosis and pulmonary hypertension. Ann Rheum Dis. 2017;76(11):1931–1940.
  • Sven MF, Pierre B, Manal FA, et al. A randomised, double-blind, placebo-controlled, multi-centre, dose-range, proof-of-concept, 24-week treatment study of lanifibranor in adult subjects with non-alcoholic steatohepatitis: design of the NATIVE study. Contemp Clin Trials. 2020;98:106170.
  • INVENTIVA. Inventiva’s lanifibranor meets the primary and key secondary endpoints in the Phase IIb NATIVE clinical trial in non-alcoholic steatohepatitis (NASH). 2020. https://www.globenewswire.com/news-release/2020/06/15/2048284/0/en/Inventiva-s-lanifibranor-meets-the-primary-and-key-secondary-endpoints-in-the-Phase-IIb-NATIVE-clinical-trial-in-non-alcoholic-steatohepatitis-NASH.html Accessed 2021 Apr 05.
  • Colca JR, McDonald WG, Kletzien RF. Mitochondrial target of thiazolidinediones. Diabetes Obes Metab. 2014;16(11):1048–1054.
  • Colca JR, McDonald WG, Adams WJ. MSDC-0620K, a metabolic modulator directed at the core pathology of non-alcoholic steatohepatitis. Expert Opin Investig Drugs. 2018;27(7):631–636.
  • Sa H, Alkhouri N, Ba D, et al. Insulin sensitizer MSDC-0620K in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase IIb study. J Hepatol. 2020;72(4):613–626.
  • Zhao P, Saltiel AR. From overnutrition to liver injury: AMP-activated protein kinase in nonalcoholic fatty liver disease. J Biol Chem. 2020;295(34):12279–12289.
  • Gluais-Dagorn P, Fouqueray P, Bolze S, et al. PXL770, a new direct AMP kinase activator, demonstrates promising effects for treatment of non-alcoholic steatohepatitis. London: [Poster] Global NASH Congress; 2018. https://www.poxelpharma.com/fr/portefeuille-produits/posters
  • Lan T, Yu Y, Zhang J, et al. Cordycepin ameliorates nonalcoholic steatohepatitis via activation of AMP-activated protein kinase signaling pathway. Hepatology. 2021 feb 12; 10.1002/hep.31749.
  • Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 2018;27(4):740–756.
  • Zelniker TA, Wiviott SD, Raz I, et al. Comparison of effects of glucagon-like peptide receptor agonists and sodium-glucose cotransporter 2 inhibitors for prevention of major adverse cardiovascular and renal outcomes in type 2 diabetes mellitus. Circulation. 2019;139(17):2022–2031.
  • Scheen AJ. Cardiovascular effects of new oral glucose-lowering agents: DPP-4 and SGLT-2 inhibitors. Cir Res. 2018;122(10):1439–1459.
  • Shirakawa J, Fujii H, Ohnuma K, et al. Diet-induced adipose tissue inflammation and liver steatosis are prevented by DPP-4 inhibition in diabetic mice. Diabetes. 2011;60(4):1246–1257.
  • Aroor AR, Habibi J, Ford DA, et al. Dipeptidyl peptidase-4 inhibition ameliorates Western diet-induced hepatic steatosis and insulin resistance through hepatic lipid remodeling and modulation of hepatic mitochondrial function. Diabetes. 2015(64):1988–2001
  • Firneisz G, Varga T, Lengyel G, et al. Serum dipeptidyl peptidase-4 activity in insulin resistant patients with non-alcoholic fatty liver disease: a novel liver disease biomarker. PLoS One. 2010;5(8):e12226.
  • Barchetta I, Ceccarelli V, Cimini FA, et al. Circulating dipeptidyl peptidase‑4 is independently associated with the presence and severity of NAFLD/NASH in individuals with and without obesity and metabolic disease. J Endocrinol Invest. 2020;44(5):979–988
  • Alam S, Ghosh J, Mustafa G, et al. Effect of sitagliptin on hepatic histological activity and fibrosis of nonalcoholic steatohepatitis patients: a 1-year randomized control trial. Hepat Med. 2018;10:23–31.
  • Cui J, Philo L, Nguyen P, et al. Sitagliptin vs. Placebo for non-alcoholic fatty liver disease: a randomized controlled trial. J. Hepatol. 2016;65(2):369–376.
  • Joy TR, McKenzie CA, Tirona RG, et al. Sitagliptin in patients with non-alcoholic steatohepatitis: a randomized placebo-controlled trial. Word J Gastroenterol. 2017;23(1):141–150.
  • Panjwani N, Mulvihill EE, Longuet C, et al. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(-/-) mice. Endocrinology. 2013;154(1):127–139.
  • Flock G, Baggio LL, Longuet C, et al. Incretin receptors for glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide are essential for the sustained metabolic activation of vildagliptin in mice. Diabetes. 2007;56(12):3006–3013.
  • Mels JE, Fu PP, Sharma S, et al. GLP-1 analogue, liraglutide, ameliorates hepatic steatosis and cardiac hypertrophy in C57Bl/6J mice fed a western diet. Am J Physiol Gastrointes Liver Physiol. 2012;302(2):G225–G235.
  • Ben-Shlomo S, Zvibel I, Shnell M, et al. Glucagon-like peptide-1 reduces hepatic lipogenesis via activation of AMP-activated protein kinase. J Hepatol. 2011;54(6):1214–1223.
  • Rakipovski G, Rolin B, Nohr J, et al. The GLP-1 analogs liraglutide and semaglutide reduce atherosclerosis in ApoE−/- and LDLr−/- mice by a mechanism that includes inflammatory pathways. JACC Basic Transl Sci. 2018;3(6):844–857.
  • Armstrong MJ, Hull D, Guo K, et al. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J Hepatol. 2016;64(2):399–408.
  • Matikainen N, Söderlund S, Björnson E, et al. Liraglutide treatment improves postprandial lipid metabolism and cardiometabolic risk factors in humans with adequately controlled type 2 diabetes: a single-centre randomized controlled study. Diabetes Obes Metab. 2019;21(1):84–94.
  • Armstrong MJ, Houlihan DD, Rowe IA, et al. Safety and efficacy of liraglutide in patients with type 2 diabetes and elevated liver enzymes: individual patient data meta-analysis of the LEAD program. Aliment Pharmacol Ther. 2013;37(2):234–242.
  • Cusi K, Sattar N, Garcia-Perez LE, et al. Dulaglutide decreases plasma aminotransferases in people with Type 2 diabetes in a pattern consistent with liver fat reduction: a post hoc analysis of the AWARD programme. Diabet Med. 2018;35(10):1434–1439.
  • Petit JM, Cercueil JP, Loffroy R, et al. Effect of liraglutide therapy on liver fat content in patients with inadequately controlled type 2 diabetes: the Lira-NAFLD study. J Clin Endocrinol Metab. 2017;102(2):40715.
  • Heuvelman VD, Van Raalte DH, Smits MM. Cardiovascular effects of glucagon-like peptide 1 receptor agonists: from mechanistic studies in humans to clinical outcomes. Cardiovasc Res. 2020;116(5):916–930.
  • Ahren B, Carr MC, Murphy K, et al. Albiglutide for the treatment of type 2 diabetes mellitus: an integrated safety analysis of the HARMONY phase 3 trials. Diabetes Res Clin Pract. 2017;126:230–239.
  • Armstrong MJ, Gaunt P, Aithal GP, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016;387(10019):679–690.
  • PN N, Buchholtz K, Cusi K, et al. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med. 2021;384(12):1113–1124.
  • Brandt SJ, Müller TD, DiMarchi RD, et al. Peptide-based multi-agonists: a new paradigm in metabolic pharmacology. J Intern Med. 2018;284(6):581–602.
  • Frias JP, Nauck MA, Van J, et al. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet. 2018;392(10160):2180–2193.
  • Hartman ML, Sanyal AJ, Loomba R, et al. Effects of novel dual GIP and GLP-1 receptor agonist tirzepatide on biomarkers of nonalcoholic steatohepatitis in patients with type 2 diabetes. Diabetes Care. 2020;43(6):1352–1355.
  • Ambery P, Parker VE, Stumvoll M, et al. MEDI0382, a GLP-1 and glucagon receptor dual agonist, in obese or overweight patients with type 2 diabetes: a randomised, controlled, double-blind, ascending dose and phase 2a study. Lancet. 2018;391(10140):2607–2618.
  • Boland ML, Laker RC, Mather K, et al. Resolution of NASH and hepatic fibrosis by the GLP-1R/GcgR dual-agonist Cotadutide via modulating mitochondrial function and lipogenesis. Nat Metab. 2020;2(5):413–431.
  • Abdelmalek MF, Choi J, Han O, et al. HM15211, a novel GLP-1/GIP/Glucagon triple-receptor co-agonist significantly reduces liver fat and body weight in obese subjects with non-alcoholic fatty liver disease: a phase 1b/2a, multicenter, randomized, placebo-controlled trial. EASL ILC meeting. 2020 ( Poster).
  • Scheen AJ. Beneficial effects of SGLT2 inhibitors on fatty liver in type 2 diabetes: a common comorbidity associated with severe complications. Diabetes Metab. 2019;45(3):213–223.
  • Wright EM, Loo DD, Hirayama BA. Biology of human sodium glucose transporters. Physiol Rev. 2011;91(2):733–794.
  • Kahl S, Gancheva S, Straßburger K, et al. Empagliflozin effectively lowers liver fat content in well-controlled type 2 diabetes: a randomized, double-blind, phase 4, placebo-controlled trial. Diabetes Care. 2020;43(2):298–305.
  • Op den Kamp YJM, de Ligt M, Dautzenberg B, et al. Effects of the SGLT2 inhibitor dapagliflozin on energy metabolism in patients with type 2 diabetes: a randomized, double-blind crossover trial. Diabetes Care. 2021;Apr15:dc202887.
  • Gastaldelli A, From CK. NASH to diabetes and from diabetes to NASH: mechanisms and treatment options. JHEP Rep. 2019;1(4):312–328.
  • Kuchay MS, Krishan S, Mishra SK, et al. Effect of empagliflozin on liver fat in patients with type 2 diabetes and nonalcoholic fatty liver disease: a randomized controlled trial (E-LIFT Trial). Diabetes Care. 2018;41(8):1801–1808.
  • Bolinder J, Ljunggren O, Kullberg J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab. 2012;97(3):1020–1031.
  • Cusi K, Bril F, Barb D, et al. Effect of canagliflozin treatment on hepatic triglyceride content and glucose metabolism in patients with type 2 diabetes. Diabetes Obes Metab. 2019;21(4):812–821.
  • Quiang S, Nakatsu Y, Seno Y, et al. Treatment with the SGLT2 inhibitor luseogliflozin improves nonalcoholic steatohepatitis in a rodent model with diabetes mellitus. Diabetol Metab Syndr. 2015; 7:104.
  • Komiya C, Tsuchiya K, Shiba K, et al. Ipragliflozin improves hepatic steatosis in obese mice and liver dysfunction in type 2 diabetic patients irrespective of body weight reduction. PLoS One. 2016;11(3):e0151511.
  • Kliewer SA, Mangelsdorf DJ. A dozen years of discovery: insights into the physiology and pharmacology of FGF21. Cell Metab. 2019;29(2):246–253.
  • Xu J, Lloyd DJ, Hale C, et al. Fibroblast growth factor 21 reverses hepatic steatosis, increases energy expenditure, and improves insulin sensitivity in diet-induced obese mice. Diabetes. 2009;58(1):250–259.
  • Tillman EJ, Rolph T. FGF21: an emerging therapeutic target for non-alcoholic steatohepatitis and related metabolic diseases. Front Endocrinol (Lausanne). 2020;11:601290.
  • Gaich G, Chien JY, Fu H, et al. The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes. Cell Metab. 2013;18(3):333–340.
  • Talukdar S, Zhou Y, Li D, et al. A long-acting FGF21 molecule, PF-05231023, decreases body weight and improves lipid profile in non-human primates and type 2 diabetic subjects. 40.
  • Charles ED, Neuschwander-Tetri BA, Frias JB, et al. Pegbelfermin (BMS-986036), PEGylated FGF21, in patients with obesity and type 2 diabetes: results from a randomized phase 2 study. Obesity (Silver Spring). 2019;27(1):41–49.
  • Sanyal A, Charles ED, Neuschwander-Tetri BA, et al. Pegbelfermin (BMS-986036), PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet. 2019;392(10165):2705–2717.
  • Therapeutics A. Biopsy Data and Safety/Tolerability for 16-Week Phase 2a BALANCED Study in NASH Patients. (2020). https://ir.akerotx.com/events/event-details/akero-therapeutics-reports-16-week-exploratory-endpoints-and-safety-endpoints Accessed 2021 Apr 12.
  • 89 bio, Inc. BIO89-100 Phase 1b/2a Topline Results [2020]. https://www.globenewswire.com/news-release/2020/09/14/2092850/0/en/89bio.Announces-Positive-Topline-Results-from-its-Phase-1b-2a-Trial-of-BIO89-100-in-NASH.html. Accessed 2021 Apr 12.
  • Kunder R, Frederickson J, Yeh FL, et al. Multiple Doses of an Anti-FGFR1/KLB Bispecific Antibody (BFKB8488A) are Associated with a Decrease in Hepatic Fat in Patients in NAFLD. AASLD: The Liver Meeting (2019). Poster LP8.
  • Talukdar S, Kharitonenkov A. FGF19 and FGF21: in NASH we trust. Mol Metab. 2021;46:101152.
  • Schumacher JD, Kong B, Wu J, et al. Direct and indirect effects of fibroblast growth factor (FGF) 15 and FGF19 on liver fibrosis development. Hepatology. 2020;71(2):670–685.
  • Harrison SA, Rinella ME, Abdelmalek MF, et al. NGM282 for treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2018;391(10126):1174–1185.
  • Harrison SA, Neff G, Guy CD, et al. Efficacy and safety of aldafermin, an engineered FGF19 analog, in a randomized, double-blind, placebo-controlled trial of patients with nonalcoholic steatohepatitis. Gastroenterology. 2021;160(1):219–231.
  • Ratziu V. Treatment of NASH with ursodeoxycholic acid: pro. Clin Res Hepatol Gastroenterol. 2012;36(Suppl 1):S41–5.
  • Ratziu V, De Ledinghen V, Oberti F, et al. A randomized controlled trial of high-dose ursodeoxycholic acid for nonalcoholic steatohepatitis. J Hepatol. 2011;54(5):1011–1019.
  • Leuschner UFH, Lindenthal B, Herrmann G, et al. High-dose ursodeoxycholic therapy for nonalcoholic steatohepatitis: a double-blind randomized, placebo-controlled trial. Hepatology. 2010;52(2):472–479.
  • Beraza N, Ofner-Ziengenfuss L, Ehedego H, et al. Nor-ursodeoxycholic acid reverses hepatocyte-specific nemo-dependent steatohepatitis. Gut. 2011;60(3):387–396.
  • Traussnigg S, Schattenberg JM, Demir M, et al. Norursodeoxycholic acid versus placebo in the treatment of non-alcoholic fatty liver disease: a double-blind, randomised, placebo-controlled, phase 2 dose-finding trial. Lancet Gastroenterol Hepatol. 2019;4(10):781–793.
  • Ratziu V, de Guevera L, Safadi R, et al. One-year results of the Global Phase 2b randomised placebo-controlled ARREST Trial of Aramchol, a stearoyl CoA desaturase modulator in NASH patients. AASLD – The Liver Meeting®, 9–13 November, 2018.
  • Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology. 2015;149(2):367–378.
  • Lassailly G, Caiazzo R, Ntandja-Wandji LC, et al. Bariatric surgery provides long-term resolution of nonalcoholic steatohepatitis and regression of fibrosis. Gastroenterology. 2020;159(4):1290–1301. .
  • Ratziu V, Friedman SL. Why do so many NASH trials fail? Gastroenterology. 2020 May 18; S0016-5085(20)30680-6 (online ahead of print). 10.1053/j.gastro.2020.05.046

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.