Efficacy and safety of an orally administered DGAT2 inhibitor alone or coadministered with a liver-targeted ACC inhibitor in adults with non-alcoholic steatohepatitis (NASH): rationale and design of the phase II, dose-ranging, dose-finding, randomised, placebo-controlled MIRNA (Metabolic Interventions to Resolve NASH with fibrosis) study

Neeta B Amin, Amanda Darekar, Quentin M Anstee, Vincent Wai-Sun Wong, Frank Tacke, Manoli Vourvahis, Douglas S Lee, Michael Charlton, Naim Alkhouri, Atsushi Nakajima, Carla Yunis, Neeta B Amin, Amanda Darekar, Quentin M Anstee, Vincent Wai-Sun Wong, Frank Tacke, Manoli Vourvahis, Douglas S Lee, Michael Charlton, Naim Alkhouri, Atsushi Nakajima, Carla Yunis

Abstract

Introduction: Small molecule inhibitors of the terminal step in intrahepatic triglyceride synthesis (diacylglycerol acyltransferase 2 inhibitor (DGAT2i, PF-06865571, ervogastat)) and upstream blockade of de novo lipogenesis via acetyl-coenzyme A carboxylase inhibitor (ACCi, PF-05221304, clesacostat) showed promise in reducing hepatic steatosis in early clinical trials. This study assesses efficacy and safety of these metabolic interventions to resolve non-alcoholic steatohepatitis (NASH) with fibrosis.

Methods and analysis: This phase II, randomised, dose-ranging, dose-finding study evaluates DGAT2i 25-300 mg two times per day (BID) or 150-300 mg once a day, DGAT2i 150-300 mg BID+ACCi 5-10 mg BID coadministration or matching placebo in a planned 450 adults with biopsy-confirmed NASH and liver fibrosis stages 2-3 from approximately 220 sites in 11 countries across North America, Europe and Asia. A triage approach including double-confirmation via non-invasive markers is included prior to screening/baseline liver biopsy. On confirmation of histological diagnosis, participants enter a ≥6-week run-in period, then a 48-week double-blind, double-dummy dosing period. The primary endpoint is the proportion of participants achieving histological NASH resolution without worsening fibrosis, ≥1 stage improvement in fibrosis without worsening NASH, or both, assessed by central pathologists. Other endpoints include assessment of hepatic steatosis (imaging substudy), overall safety and tolerability, and evaluation of blood-based biomarkers and quantitative ultrasound parameters over time.

Ethics and dissemination: Metabolic Interventions to Resolve NASH with fibrosis (MIRNA) is conducted in accordance with the Declaration of Helsinki and Council for International Organisations of Medical Sciences (CIOMS) International Ethical Guidelines, International Council on Harmonisation Good Clinical Practice guidelines, applicable laws and regulations, including privacy laws. Local independent review board/ethics committees (IRB/ECs) review/approve the protocol, any amendments, informed consent and other forms. Participants provide written informed consent. Details of all IRB/ECs, as well as results, will be published in a peer-reviewed journal and publicly disclosed through ClinicalTrials.gov, EudraCT, and/or www.pfizer.com and other public registries as per applicable local laws/regulations.

Trial registration number: NCT04321031.

Keywords: clinical trials; hepatology; histopathology; magnetic resonance imaging.

Conflict of interest statement

Competing interests: NBA, AD, DSL, MV and CY are employees of, and hold stock or stock options with, Pfizer Inc. QMA has received fees for consultancy on behalf of Newcastle University, grant funding via the EU IMI2 scheme and speaker fees from Pfizer Inc in relation to the submitted work. QMA has received grant funding from AbbVie, Allergan/Tobira, AstraZeneca, Genfit SA, GlaxoSmithKline, Glympse Bio, Intercept Pharma Europe Ltd (via the EU IMI2 scheme), Novartis Pharma AG, Pfizer Inc (via the EU IMI2 scheme); speaker fees from Bristol Myers Squibb, Gilead, Kenes, Novo Nordisk, Pfizer Inc; consultancy fees on behalf of Newcastle University from 89Bio, Allergan/Tobira, Altimmune, AstraZeneca, Axcella, BGMBio, Blade, BNN Cardio, Bristol Myers Squibb, Celgene, Cirius, CymaBay, E3Bio, EcoR1, Eli Lilly & Co, Galmed, Genentech, Genfit SA, Gilead, Grunthal, HistoIndex, Imperial Innovations, Indalo, Intercept Pharma Europe Ltd., Inventiva, IQVIA, Janssen, Madrigal, MedImmune, Metacrine, NewGene, NGMBio, North Sea Therapeutics, Novartis Pharma AG, Novo Nordisk, PathAI, Pfizer Inc, Poxel, Raptor Pharma, Servier, Terns, Viking Therapeutics, outside the submitted work. VW-SW has received medical writing and article processing charges support from Pfizer Inc for the submitted work. VW-SW’s institution has received grant funding from Gilead and support for meetings attendance from AbbVie and Gilead; VW-SW has received consulting fees and payments for participation on a Data Safety Monitoring Board or Advisory Board from 3V-Bio, AbbVie, Allergan, Boehringer Ingelheim, Centre for Outcomes Research in Liver Diseases, Echosens, Gilead, Hanmi Pharmaceutical, Intercept, Inventiva, Merck, Novartis, Novo Nordisk, Perspectum Diagnostics, Pfizer Inc, ProSciento, Sagimet Biosciences, TARGET PharmaSolutions, Terns; VW-SW has received payment or honoraria for lectures, presentations, speakers bureaux, manuscript writing or educational events from Abbott, AbbVie, Bristol Myers Squibb, Echosens, Gilead; VW-SV has stock or stock options in Illuminatio Medical Technology Ltd. FT’s institution has received grants or contracts from Allergan, Bristol Myers Squibb, Galapagos, Gilead and Inventiva; FT has received consulting fees from AbbVie, Allergan, Boehringer Ingelheim, Bristol Myers Squibb, Galapagos, Gilead, Ionis, Ipsen, Inventiva, Novartis, Pfizer Inc and Roche; FT has received payment or honoraria for lectures, presentations, speakers bureaux, manuscript writing or educational events from Falk, Intercept and Gilead; he has received payment for expert testimony from Alnylam. NA has received grants or contracts from Akero, Allergan, Bristol Myers Squibb, DSM, Genentech, Genfit, Gilead, Intercept, Inventiva, Madrigal, NGMBio, North Sea Therapeutics, Novo Nordisk, Pfizer Inc, Poxel and Zydus; NA has received payment or honoraria for lectures, presentations, speakers bureaux, manuscript writing or educational events from Gilead and Intercept.MC and AN declare no competing interests. A patent rationalising the invention of administering DGAT2i+ACCi to mitigate effects of ACCi alone has been submitted.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Effects of ACC and DGAT2 inhibition on hepatic lipid metabolism. aAdaptive effects. bIn non-clinical models. ACC, acetyl-coenzyme A carboxylase; ACCi, ACC inhibitor; ACS, acyl-CoA synthetase; CoA, coenzyme A; CPT1, carnitine palmitoyl transferase 1; DAG, diacylglycerol; DGAT, diacylglycerol acyltransferase; DGAT2i, DGAT2 inhibitor; FAS, fatty acid synthase; FFA, free fatty acid; G-3-P, glycerol-3-phosphate; MUFA, monounsaturated fatty acid; PPARα, peroxisome proliferator-activated receptor α; SCD1, stearoyl-CoA desaturase 1; SREBP, sterol regulatory element-binding protein; TAG, triacylglycerol (also known as triglyceride); TG, triglyceride.
Figure 2
Figure 2
MIRNA study design. The intervals depict the maximum time between the various periods in the study. aIn addition, metformin dose reduced if dose is >1 g/day. ACCi, acetyl-CoA carboxylase inhibitor; BID, two times per day; DGAT2i, diacylglycerol acyltransferase 2 inhibitor; MIRNA, Metabolic Interventions to Resolve NASH with fibrosis; MRI-PDFF, magnetic resonance imaging-proton density fat fraction; n, target number of participants; QD, once a day.
Figure 3
Figure 3
Prospective, centralised grading and scoring of liver biopsies at screening for eligibility using the NASH-CRN definition. aConsensus review to reach agreement is not required if both pathologists agree that either the NAFLD Activity Score or fibrosis grade renders the participant ineligible. NAFLD, non-alcoholic fatty liver disease; NASH-CRN, Non-Alcoholic Steatohepatitis Clinical Research Network.

References

    1. 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;64:73–84. 10.1002/hep.28431
    1. LaBrecque DR, Abbas Z, Anania F. World gastroenterology organisation global guidelines: nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Clin Gastroenterol 2014;48:467–73. 10.1097/MCG.0000000000000116
    1. Diehl AM, Day C. Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis. N Engl J Med 2017;377:2063–72. 10.1056/NEJMra1503519
    1. Younossi Z, Anstee QM, Marietti M, et al. . Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 2018;15:11–20. 10.1038/nrgastro.2017.109
    1. Friedman SL, Neuschwander-Tetri BA, Rinella M, et al. . Mechanisms of NAFLD development and therapeutic strategies. Nat Med 2018;24:908–22. 10.1038/s41591-018-0104-9
    1. Angulo P, Kleiner DE, Dam-Larsen S, et al. . Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 2015;149:389–97.e10. 10.1053/j.gastro.2015.04.043
    1. Hagström H, Nasr P, Ekstedt M, et al. . Fibrosis stage but not NASH predicts mortality and time to development of severe liver disease in biopsy-proven NAFLD. J Hepatol 2017;67:1265–73. 10.1016/j.jhep.2017.07.027
    1. Taylor RS, Taylor RJ, Bayliss S, et al. . Association between fibrosis stage and outcomes of patients with nonalcoholic fatty liver disease: a systematic review and meta-analysis. Gastroenterology 2020;158:1611–25.e12. 10.1053/j.gastro.2020.01.043
    1. Dulai PS, Singh S, Patel J, et al. . Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta-analysis. Hepatology 2017;65:1557–65. 10.1002/hep.29085
    1. Lambert JE, Ramos-Roman MA, Browning JD, et al. . Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology 2014;146:726–35. 10.1053/j.gastro.2013.11.049
    1. Kim C-W, Addy C, Kusunoki J, et al. . Acetyl CoA carboxylase inhibition reduces hepatic steatosis but elevates plasma triglycerides in mice and humans: a bedside to bench investigation. Cell Metab 2017;26:394–406. 10.1016/j.cmet.2017.07.009
    1. Zammit VA. Hepatic triacylglycerol synthesis and secretion: DGAT2 as the link between glycaemia and triglyceridaemia. Biochem J 2013;451:1–12. 10.1042/BJ20121689
    1. Yen C-LE, Stone SJ, Koliwad S, et al. . Thematic review series: glycerolipids. DGAT enzymes and triacylglycerol biosynthesis. J Lipid Res 2008;49:2283–301. 10.1194/jlr.R800018-JLR200
    1. Kammoun HL, Chabanon H, Hainault I, et al. . GRP78 expression inhibits insulin and ER stress-induced SREBP-1c activation and reduces hepatic steatosis in mice. J Clin Invest 2009;119:1201–15. 10.1172/JCI37007
    1. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 2002;109:1125–31. 10.1172/JCI15593
    1. Choi CS, Savage DB, Kulkarni A, et al. . Suppression of diacylglycerol acyltransferase-2 (DGAT2), but not DGAT1, with antisense oligonucleotides reverses diet-induced hepatic steatosis and insulin resistance. J Biol Chem 2007;282:22678–88. 10.1074/jbc.M704213200
    1. Amin NB, Carvajal-Gonzalez S, Purkal J, et al. . Targeting diacylglycerol acyltransferase 2 for the treatment of nonalcoholic steatohepatitis. Sci Transl Med 2019;11:eaav9701. 10.1126/scitranslmed.aav9701
    1. Saxena AR, Chidsey K, Somayaji V, et al . Diacylglycerol acyltransferase 2 (DGAT2) inhibitor PF-06865571 reduces liver fat by MRI-PDFF after 2 weeks in adults with NAFLD [abstract]. Hepatology 2019;70(Suppl 1):1260A.
    1. Bates J, Vijayakumar A, Ghoshal S, et al. . Acetyl-CoA carboxylase inhibition disrupts metabolic reprogramming during hepatic stellate cell activation. J Hepatol 2020;73:896–905. 10.1016/j.jhep.2020.04.037
    1. Loomba R, Kayali Z, Noureddin M, et al. . GS-0976 reduces hepatic steatosis and fibrosis markers in patients with nonalcoholic fatty liver disease. Gastroenterology 2018;155:1463–73.e6. 10.1053/j.gastro.2018.07.027
    1. Lawitz EJ, Coste A, Poordad F, et al. . Acetyl-CoA carboxylase inhibitor GS-0976 for 12 weeks reduces hepatic de novo lipogenesis and steatosis in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2018;16:1983–91.e3. 10.1016/j.cgh.2018.04.042
    1. Bergman A, Carvajal-Gonzalez S, Tarabar S, et al. . Safety, tolerability, pharmacokinetics and pharmacodynamics of a liver-targeting ACC inhibitor (PF-05221304): a three-part randomized phase 1 study. Clin Pharmacol Drug Dev 2020;9:514–26. 10.1002/cpdd.782
    1. Calle RA, Amin NB, Carvajal-Gonzalez S, et al. . ACC inhibitor alone or co-administered with a DGAT2 inhibitor in patients with non-alcoholic fatty liver disease: two parallel, placebo-controlled, randomized phase 2a trials. Nat Med 2021;27:1836–48. 10.1038/s41591-021-01489-1
    1. Alkhouri N, Lawitz E, Noureddin M, et al. . GS-0976 (Firsocostat): an investigational liver-directed acetyl-CoA carboxylase (ACC) inhibitor for the treatment of non-alcoholic steatohepatitis (NASH). Expert Opin Investig Drugs 2020;29:135–41. 10.1080/13543784.2020.1668374
    1. Moon YA. The SCAP/SREBP pathway: a mediator of hepatic steatosis. Endocrinol Metab 2017;32:6–10. 10.3803/EnM.2017.32.1.6
    1. Goedeke L, Bates J, Vatner DF, et al. . Acetyl-CoA carboxylase inhibition reverses NAFLD and hepatic insulin resistance but promotes hypertriglyceridemia in rodents. Hepatology 2018;68:2197–211. 10.1002/hep.30097
    1. Ross TT, Crowley C, Rinaldi A, et al . Co-administration of the ACC inhibitor PF-05221304 and the DGAT2 inhibitor PF-06865571 produces greater efficacy and mitigates ACC inhibitor-mediated increases in circulating triglyceride NAFLD/NASH models [abstract]. Hepatology 2019;70(Suppl 1:1258A.
    1. Kleiner DE, Brunt EM, Wilson LA, et al. . Association of histologic disease activity with progression of nonalcoholic fatty liver disease. JAMA Netw Open 2019;2:e1912565. 10.1001/jamanetworkopen.2019.12565
    1. 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;67:328–57. 10.1002/hep.29367
    1. European Association for the Study of the Liver . EASL Clinical Practice Guidelines on non-invasive tests for evaluation of liver disease severity and prognosis - 2021 update. J Hepatol 2021;75:659–89. 10.1016/j.jhep.2021.05.025
    1. Newsome PN, Sasso M, Deeks JJ, et al. . FibroScan-AST (FAST) score for the non-invasive identification of patients with non-alcoholic steatohepatitis with significant activity and fibrosis: a prospective derivation and global validation study. Lancet Gastroenterol Hepatol 2020;5:362–73. 10.1016/S2468-1253(19)30383-8
    1. Kleiner DE, Brunt EM, Van Natta M, et al. . Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005;41:1313–21. 10.1002/hep.20701
    1. Castera L, Friedrich-Rust M, Loomba R. Noninvasive assessment of liver disease in patients with nonalcoholic fatty liver disease. Gastroenterology 2019;156:1264–81.e4. 10.1053/j.gastro.2018.12.036
    1. Noureddin N, Han MAT, Alkhouri N, et al. . Accounting for the placebo effect and optimizing outcomes in clinical trials of nonalcoholic steatohepatitis (NASH). Curr Hepatol Rep 2020;19:63–9. 10.1007/s11901-020-00505-1
    1. Glass O, Filozof C, Noureddin M, et al. . Standardisation of diet and exercise in clinical trials of NAFLD-NASH: Recommendations from the Liver Forum. J Hepatol 2020;73:680–93. 10.1016/j.jhep.2020.04.030
    1. Filozof C, Chow S-C, Dimick-Santos L, et al. . Clinical endpoints and adaptive clinical trials in precirrhotic nonalcoholic steatohepatitis: facilitating development approaches for an emerging epidemic. Hepatol Commun 2017;1:577–85. 10.1002/hep4.1079
    1. U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) . Noncirrhotic nonalcoholic steatohepatitis with liver fibrosis: developing drugs for treatment – guidance for industry, 2018. Available: [Accessed 6 Apr 2021].
    1. Cheung A, Neuschwander-Tetri BA, Kleiner DE, et al. . Defining improvement in nonalcoholic steatohepatitis for treatment trial endpoints: recommendations from the liver forum. Hepatology 2019;70:1841–55. 10.1002/hep.30672
    1. Mashhood A, Railkar R, Yokoo T, et al. . Reproducibility of hepatic fat fraction measurement by magnetic resonance imaging. J Magn Reson Imaging 2013;37:1359–70. 10.1002/jmri.23928
    1. Davison BA, Harrison SA, Cotter G, et al. . Suboptimal reliability of liver biopsy evaluation has implications for randomized clinical trials. J Hepatol 2020;73:1322–32. 10.1016/j.jhep.2020.06.025
    1. Harrison SA, Alkhouri N, Davison BA, et al. . Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase IIb study. J Hepatol 2020;72:613–26. 10.1016/j.jhep.2019.10.023
    1. Yamaguchi K, Yang L, McCall S, et al. . Inhibiting triglyceride synthesis improves hepatic steatosis but exacerbates liver damage and fibrosis in obese mice with nonalcoholic steatohepatitis. Hepatology 2007;45:1366–74. 10.1002/hep.21655
    1. Chitraju C, Mejhert N, Haas JT, et al. . Triglyceride Synthesis by DGAT1 Protects Adipocytes from Lipid-Induced ER Stress during Lipolysis. Cell Metab 2017;26:407–18.e3. 10.1016/j.cmet.2017.07.012
    1. Long MT, Zhang X, Xu H, et al. . Hepatic fibrosis associates with multiple cardiometabolic disease risk factors: the Framingham heart study. Hepatology 2021;73:548–59. 10.1002/hep.31608
    1. Viele K, Connor JT. Dose-finding trials: optimizing phase 2 data in the drug development process. JAMA 2015;314:2294–5. 10.1001/jama.2015.16702
    1. Rinella ME, Tacke F, Sanyal AJ, et al. . Report on the AASLD/EASL joint workshop on clinical trial endpoints in NAFLD. J Hepatol 2019;71:823–33. 10.1016/j.jhep.2019.04.019
    1. Committee for Medicinal Products for Human Use (CHMP) . Reflection paper on regulatory requirements for the development of medicinal products for chronic non-infectious liver diseases (PBC, PSC, NASH): EMA, 2018. Available: [Accessed 6 Apr 2021].
    1. Amin NB, Wang X, Jain SM, et al. . Dose-ranging efficacy and safety study of ertugliflozin, a sodium-glucose co-transporter 2 inhibitor, in patients with type 2 diabetes on a background of metformin. Diabetes Obes Metab 2015;17:591–8. 10.1111/dom.12460
    1. Amin NB, Aggarwal N, Pall D, et al. . Two dose-ranging studies with PF-04937319, a systemic partial activator of glucokinase, as add-on therapy to metformin in adults with type 2 diabetes. Diabetes Obes Metab 2015;17:751–9. 10.1111/dom.12474
    1. Yu XX, Murray SF, Pandey SK, et al. . Antisense oligonucleotide reduction of DGAT2 expression improves hepatic steatosis and hyperlipidemia in obese mice. Hepatology 2005;42:362–71. 10.1002/hep.20783

Source: PubMed

3
Předplatit