281
Views
10
CrossRef citations to date
0
Altmetric
Review

Experimental and Investigational Targeted Therapies for the Management of Fibrosis in NASH: An Update

, & ORCID Icon
Pages 329-338 | Published online: 18 Mar 2021

References

  • Younossi Z, Tacke F, Arrese M, et al. Global perspectives on nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology. 2019;69(6):2672–2682. doi:10.1002/hep.30251
  • Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214(2):199–210. doi:10.1002/path.2277
  • Roehlen N, Crouchet E, Baumert TF. Liver fibrosis: mechanistic concepts and therapeutic perspectives. Cells. 2020;9(4):875. doi:10.3390/cells9040875
  • Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 2017;14(7):397–411. doi:10.1038/nrgastro.2017.38
  • Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2020.
  • Lemoinne S, Friedman SL. New and emerging anti-fibrotic therapeutics entering or already in clinical trials in chronic liver diseases. Curr Opin Pharmacol. 2019;49:60–70. doi:10.1016/j.coph.2019.09.006
  • Mohagheghi S, Geramizadeh B, Nikeghbalian S, et al. Intricate role of yes-associated protein1 in human liver cirrhosis: TGF-beta1 still is a giant player. IUBMB Life. 2019;71(10):1453–1464. doi:10.1002/iub.2052
  • Dewidar B, Meyer C, Dooley S, Meindl-Beinker AN. TGF-beta in hepatic stellate cell activation and liver fibrogenesis-updated 2019. Cells. 2019;8(11):1419. doi:10.3390/cells8111419
  • Hellerbrand C, Stefanovic B, Giordano F, Burchardt ER, Brenner DA. The role of TGFbeta1 in initiating hepatic stellate cell activation in vivo. J Hepatol. 1999;30(1):77–87. doi:10.1016/S0168-8278(99)80010-5
  • Arthur MJ, Fibrogenesis II. Metalloproteinases and their inhibitors in liver fibrosis. Am J Physiol Gastrointest Liver Physiol. 2000;279(2):G245–249. doi:10.1152/ajpgi.2000.279.2.G245
  • Campbell JS, Hughes SD, Gilbertson DG, et al. Platelet-derived growth factor C induces liver fibrosis, steatosis, and hepatocellular carcinoma. Proc Natl Acad Sci U S A. 2005;102(9):3389–3394. doi:10.1073/pnas.0409722102
  • Yang L, Kwon J, Popov Y, et al. Vascular endothelial growth factor promotes fibrosis resolution and repair in mice. Gastroenterology. 2014;146(5):1339–1350e1331. doi:10.1053/j.gastro.2014.01.061
  • Noureddin M, Anstee QM, Loomba R. Review article: emerging anti-fibrotic therapies in the treatment of non-alcoholic steatohepatitis. Aliment Pharmacol Ther. 2016;43(11):1109–1123. doi:10.1111/apt.13620
  • Duran A, Hernandez ED, Reina-Campos M, et al. p62/SQSTM1 by binding to vitamin d receptor inhibits hepatic stellate cell activity, fibrosis, and liver cancer. Cancer Cell. 2016;30(4):595–609. doi:10.1016/j.ccell.2016.09.004
  • An P, Wei LL, Zhao S, et al. Hepatocyte mitochondria-derived danger signals directly activate hepatic stellate cells and drive progression of liver fibrosis. Nat Commun. 2020;11(1):2362. doi:10.1038/s41467-020-16092-0
  • Ioannou GN. The role of cholesterol in the pathogenesis of NASH. Trends Endocrinol Metab. 2016;27(2):84–95. doi:10.1016/j.tem.2015.11.008
  • Davison BA, Harrison SA, Cotter G, et al. Suboptimal reliability of liver biopsy evaluation has implications for randomized clinical trials. J Hepatol. 2020.
  • Vali Y, Lee J, Boursier J, et al. Enhanced liver fibrosis test for the non-invasive diagnosis of fibrosis in patients with NAFLD: a systematic review and meta-analysis. J Hepatol. 2020;73(2):252–262. doi:10.1016/j.jhep.2020.03.036
  • Boursier J, Guillaume M, Leroy V, et al. New sequential combinations of noninvasive fibrosis tests provide an accurate diagnosis of advanced fibrosis in NAFLD. J Hepatol. 2019;71(2):389–396. doi:10.1016/j.jhep.2019.04.020
  • Sterling RK, Lissen E, Clumeck N. Development of a simple noninvasive index to predict significant fibrosis in patients with HIV/HCV coinfection. Hepatology. 2006;43(6):1317–1325. doi:10.1002/hep.21178
  • Boursier J, Vergniol J, Guillet A, et al. Diagnostic accuracy and prognostic significance of blood fibrosis tests and liver stiffness measurement by FibroScan in non-alcoholic fatty liver disease. J Hepatol. 2016;65(3):570–578. doi:10.1016/j.jhep.2016.04.023
  • de Ledinghen V, Vergniol J. Transient elastography for the diagnosis of liver fibrosis. Expert Rev Med Devices. 2010;7(6):811–823. doi:10.1586/erd.10.46
  • de Ledinghen V, Vergniol J, Foucher J, El-Hajbi F, Merrouche W, Rigalleau V. Feasibility of liver transient elastography with FibroScan using a new probe for obese patients. Liver Int. 2010;30(7):1043–1048. doi:10.1111/j.1478-3231.2010.02258.x
  • Lee SJ, Kim SU. Noninvasive monitoring of hepatic steatosis: controlled attenuation parameter and magnetic resonance imaging-proton density fat fraction in patients with nonalcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol. 2019;13(6):523–530. doi:10.1080/17474124.2019.1608820
  • Smith AD, Porter KK, Elkassem AA, Sanyal R, Lockhart ME. Current imaging techniques for noninvasive staging of hepatic fibrosis. AJR Am J Roentgenol. 2019;1–13.
  • Jayakumar S, Middleton MS, Lawitz EJ, et al. Longitudinal correlations between MRE, MRI-PDFF, and liver histology in patients with non-alcoholic steatohepatitis: analysis of data from a Phase II trial of selonsertib. J Hepatol. 2019;70(1):133–141. doi:10.1016/j.jhep.2018.09.024
  • Han MAT, Altayar O, Hamdeh S, et al. Rates of and factors associated with placebo response in trials of pharmacotherapies for nonalcoholic steatohepatitis: systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2019;17(4):616–629e626. doi:10.1016/j.cgh.2018.06.011
  • Lin SC, Heba E, Bettencourt R, et al. Assessment of treatment response in non-alcoholic steatohepatitis using advanced magnetic resonance imaging. Aliment Pharmacol Ther. 2017;45(6):844–854. doi:10.1111/apt.13951
  • Roeb E. Matrix metalloproteinases and liver fibrosis (translational aspects). Matrix Biol. 2018;68–69:463–473. doi:10.1016/j.matbio.2017.12.012
  • Roeb E, Geier A. Nonalcoholic steatohepatitis (NASH) – current treatment recommendations and future developments. Z Gastroenterol. 2019;57(4):508–517. doi:10.1055/a-0784-8827
  • Rizzo G, Renga B, Mencarelli A, Pellicciari R, Fiorucci S. Role of FXR in regulating bile acid homeostasis and relevance for human diseases. Curr Drug Targets Immune Endocr Metabol Disord. 2005;5(3):289–303. doi:10.2174/1568008054863781
  • Venetsanaki V, Karabouta Z, Polyzos SA. Farnesoid X nuclear receptor agonists for the treatment of nonalcoholic steatohepatitis. Eur J Pharmacol. 2019;863:172661. doi:10.1016/j.ejphar.2019.172661
  • Neuschwander-Tetri BA, Loomba R, Sanyal AJ, et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015;385(9972):956–965. doi:10.1016/S0140-6736(14)61933-4
  • Younossi ZM, Ratziu V, Loomba R, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019;394(10215):2184–2196. doi:10.1016/S0140-6736(19)33041-7
  • Eaton JE, Gores GJ. Long-term outcomes with obeticholic acid in primary biliary cholangitis: reassuring, but still an itch we need to scratch. Lancet Gastroenterol Hepatol. 2019;4(6):417–418. doi:10.1016/S2468-1253(19)30084-6
  • Pedrosa M, Seyedkazemi S, Francque S, et al. A randomized, double-blind, multicenter, phase 2b study to evaluate the safety and efficacy of a combination of tropifexor and cenicriviroc in patients with nonalcoholic steatohepatitis and liver fibrosis: study design of the TANDEM trial. Contemp Clin Trials. 2020;88:105889. doi:10.1016/j.cct.2019.105889
  • Loomba R, Noureddin M, Kowdley KV, et al. Combination therapies including cilofexor and firsocostat for bridging fibrosis and cirrhosis due to NASH. Hepatology. 2020;73:S116–S117. doi:10.1016/S0168-8278(20)30753-4
  • Polyzos SA, Kang ES, Boutari C, Rhee EJ, Mantzoros CS. Current and emerging pharmacological options for the treatment of nonalcoholic steatohepatitis. Metabolism. 2020;111S:154203. doi:10.1016/j.metabol.2020.154203
  • Harrison SA, Bashir MR, Guy CD, et al. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2019;394(10213):2012–2024. doi:10.1016/S0140-6736(19)32517-6
  • Zhou J, Waskowicz LR, Lim A, et al. A liver-specific thyromimetic, VK2809, decreases hepatosteatosis in glycogen storage disease type Ia. Thyroid. 2019;29(8):1158–1167. doi:10.1089/thy.2019.0007
  • Mondal S, Mugesh G. Novel thyroid hormone analogues, enzyme inhibitors and mimetics, and their action. Mol Cell Endocrinol. 2017;458:91–104. doi:10.1016/j.mce.2017.04.006
  • Yilmaz Y, Eren F, Yonal O, et al. Increased serum FGF21 levels in patients with nonalcoholic fatty liver disease. Eur J Clin Invest. 2010;40(10):887–892. doi:10.1111/j.1365-2362.2010.02338.x
  • Gong Q, Hu Z, Zhang F, et al. Fibroblast growth factor 21 improves hepatic insulin sensitivity by inhibiting mammalian target of rapamycin complex 1 in mice. Hepatology. 2016;64(2):425–438. doi:10.1002/hep.28523
  • Degirolamo C, Sabba C, Moschetta A. Therapeutic potential of the endocrine fibroblast growth factors FGF19, FGF21 and FGF23. Nat Rev Drug Discov. 2016;15(1):51–69.
  • Sanyal A, Charles ED, Neuschwander-Tetri BA, et al. Pegbelfermin (BMS-986036), a 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. doi:10.1016/S0140-6736(18)31785-9
  • 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. 2020.
  • Baruch A, Wong C, Chinn LW, et al. Antibody-mediated activation of the FGFR1/Klothobeta complex corrects metabolic dysfunction and alters food preference in obese humans. Proc Natl Acad Sci U S A. 2020;117(46):28992–29000. doi:10.1073/pnas.2012073117
  • Zarei M, Aguilar-Recarte D, Palomer X, Vazquez-Carrera M. Revealing the role of peroxisome proliferator-activated receptor beta/delta in nonalcoholic fatty liver disease. Metabolism. 2021;114:154342. doi:10.1016/j.metabol.2020.154342
  • Fougerat A, Montagner A, Loiseau N, Guillou H, Wahli W. Peroxisome proliferator-activated receptors and their novel ligands as candidates for the treatment of non-alcoholic fatty liver disease. Cells. 2020;9(7):1638. doi:10.3390/cells9071638
  • Choudhary NS, Kumar N, Duseja A. Peroxisome proliferator-activated receptors and their agonists in nonalcoholic fatty liver disease. J Clin Exp Hepatol. 2019;9(6):731–739. doi:10.1016/j.jceh.2019.06.004
  • Kumar DP, Caffrey R, Marioneaux J, et al. The PPAR alpha/gamma agonist saroglitazar improves insulin resistance and steatohepatitis in a diet induced animal model of nonalcoholic fatty liver disease. Sci Rep. 2020;10(1):9330. doi:10.1038/s41598-020-66458-z
  • Boeckmans J, Natale A, Rombaut M, et al. Anti-NASH drug development hitches a lift on PPAR agonism. Cells. 2019;9(1):37. doi:10.3390/cells9010037
  • Jones D, Boudes PF, Swain MG, et al. Seladelpar (MBX-8025), a selective PPAR-delta agonist, in patients with primary biliary cholangitis with an inadequate response to ursodeoxycholic acid: a double-blind, randomised, placebo-controlled, phase 2, proof-of-concept study. Lancet Gastroenterol Hepatol. 2017;2(10):716–726. doi:10.1016/S2468-1253(17)30246-7
  • Haczeyni F, Wang H, Barn V, et al. The selective peroxisome proliferator-activated receptor-delta agonist seladelpar reverses nonalcoholic steatohepatitis pathology by abrogating lipotoxicity in diabetic obese mice. Hepatol Commun. 2017;1(7):663–674. doi:10.1002/hep4.1072
  • 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):1147–1159e1145. doi:10.1053/j.gastro.2016.01.038
  • Newsome PN, Buchholtz K, Cusi K, et al. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med. 2020. doi:10.1056/NEJMoa2028395
  • Liang Z, Li T, Jiang S, et al. AMPK: a novel target for treating hepatic fibrosis. Oncotarget. 2017;8(37):62780–62792. doi:10.18632/oncotarget.19376
  • Colca JR, McDonald WG, McCommis KS, Finck BN. Treating fatty liver disease by modulating mitochondrial pyruvate metabolism. Hepatol Commun. 2017;1(3):193–197. doi:10.1002/hep4.1036
  • van den Hoek AM, Pieterman EJ, van der Hoorn JW, et al. Icosabutate exerts beneficial effects upon insulin sensitivity, hepatic inflammation, lipotoxicity, and fibrosis in mice. Hepatol Commun. 2020;4(2):193–207. doi:10.1002/hep4.1453
  • Stokman G, van den Hoek AM, Denker Thorbekk D, et al. Dual targeting of hepatic fibrosis and atherogenesis by icosabutate, an engineered eicosapentaenoic acid derivative. Liver Int. 2020;40(11):2860–2876. doi:10.1111/liv.14643
  • Iruarrizaga-Lejarreta M, Varela-Rey M, Fernandez-Ramos D, et al. Role of aramchol in steatohepatitis and fibrosis in mice. Hepatol Commun. 2017;1(9):911–927. doi:10.1002/hep4.1107
  • Ratziu V, de Guevara L, Safadi R, et al. One-year results of the global phase 2b randomized placebo controlled ARREST trial of aramchol, a stearoyl CoA desaturase modulator in NASH patients. Hepatology. 2018;68:LB–5.
  • Safadi R, Konikoff FM, Mahamid M, et al. The fatty acid-bile acid conjugate aramchol reduces liver fat content in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2014;12(12):2085–2091e2081. doi:10.1016/j.cgh.2014.04.038
  • Weston CJ, Shepherd EL, Claridge LC, et al. Vascular adhesion protein-1 promotes liver inflammation and drives hepatic fibrosis. J Clin Invest. 2015;125(2):501–520. doi:10.1172/JCI73722
  • Tricot T, De Boeck J, Verfaillie C. Alternative cell sources for liver parenchyma repopulation: where do we stand? Cells. 2020;9(3):566. doi:10.3390/cells9030566
  • Lefere S, Devisscher L, Tacke F. Targeting CCR2/5 in the treatment of nonalcoholic steatohepatitis (NASH) and fibrosis: opportunities and challenges. Expert Opin Investig Drugs. 2020;29(2):89–92. doi:10.1080/13543784.2020.1718106
  • Younossi ZM, Stepanova M, Lawitz E, et al. Improvement of hepatic fibrosis and patient-reported outcomes in non-alcoholic steatohepatitis treated with selonsertib. Liver Int. 2018;38(10):1849–1859. doi:10.1111/liv.13706