Safety, Tolerability, and Biologic Activity of AXA1125 and AXA1957 in Subjects With Nonalcoholic Fatty Liver Disease

Stephen A Harrison, Seth J Baum, Nadege T Gunn, Ziad H Younes, Anita Kohli, Rashmee Patil, Margaret J Koziel, Harinder Chera, Jeff Zhao, Manu V Chakravarthy, Stephen A Harrison, Seth J Baum, Nadege T Gunn, Ziad H Younes, Anita Kohli, Rashmee Patil, Margaret J Koziel, Harinder Chera, Jeff Zhao, Manu V Chakravarthy

Abstract

Introduction: AXA1125 and AXA1957 are novel, orally administered endogenous metabolic modulator compositions, specifically designed to simultaneously support multiple metabolic and fibroinflammatory pathways associated with nonalcoholic fatty liver disease (NAFLD). This study assessed safety, tolerability, and biologic activity of AXA1125 and AXA1957 in NAFLD.

Methods: In this multicenter, 16-week, placebo-controlled, single-blind, randomized clinical study in subjects with NAFLD stratified by type 2 diabetes, AXA1125 24 g, AXA1957 13.5 g or 20.3 g, or placebo was administered twice daily. Key metabolism (MRI-proton density fat fraction [MRI-PDFF] and homeostasis model assessment of insulin resistance [HOMA-IR]) and fibroinflammation markers (alanine aminotransferase [ALT], corrected T1 [cT1], keratin-18 [K-18] M65, and N-terminal type III collagen propeptide [Pro-C3]) were evaluated. Safety outcomes included adverse events and standard laboratory assessments.

Results: Baseline characteristics of the 102 enrolled subjects, including 40 with type 2 diabetes, were consistent with presumed nonalcoholic steatohepatitis. AXA1125 showed consistently greater biologic activity than AXA1957 or placebo. Week 16 changes from baseline with AXA1125 vs placebo: MRI-PDFF -22.9% vs -5.7%, HOMA-IR -4.4 vs +0.7, ALT -21.9% vs -7.2%, K-18 M65 -13.6% vs +20.1%, cT1 -69.6 vs +18.3 ms (P < 0.05), and Pro-C3 -13.6% vs -3.6%. Week 16 changes from baseline with AXA1957 20.3 g: MRI-PDFF -8.1%, HOMA-IR +8.4, ALT -20.7%, K-18 M65 6.6%, cT1 -34.7 ms, and Pro-C3 -15.6%. A greater proportion of subjects treated with AXA1125 achieved clinically relevant thresholds: ≥30% MRI-PDFF, ≥17-IU/L ALT, and ≥80-ms cT1 reductions at week 16. Study products were safe and well tolerated with stable lipid and weight profiles.

Discussion: Both compositions showed multitargeted activity on relevant NAFLD pathways. AXA1125 demonstrated the greatest activity over 16 weeks, warranting continued clinical investigation in nonalcoholic steatohepatitis subjects.

Trial registration: ClinicalTrials.gov NCT04073368.

Conflict of interest statement

Guarantor of the article: Manu V. Chakravarthy, MD, PhD.

Specific author contributions: S.A.H., M.J.K., H.C., J.Z., and M.V.C.: study design and conduct of the study. S.A.H., M.K., H.C., and M.V.C.: collection, analysis, and interpretation of data. J.Z., M.K., and M.V.C.: statistical analysis of the data. M.V.C.: drafting of the manuscript. S.A.H., S.J.B., N.T.G., Z.H.Y., A.K., R.P., M.J.K., H.C., J.Z., and M.V.C.: critical revision of the manuscript for important intellectual content.

Financial support: This study was funded by Axcella Health. Medical writing support, funded by Axcella Health, was provided by Diann Glickman of Envision Pharma Group.

Potential competing interests: S.A.H.: stock shareholder: Akero, Cirius, Galectin, Genfit, HistoIndex, Madrigal, Metacrine, NGM Bio, and NorthSea; consultant: Akero, Alentis, Altimmune, Axcella, Cirius, Chronic Liver Disease Foundation, CiVi BioPharma, CymaBay, Echosens, Fibronostics, Forsite Labs, Fortress Biotech, Galectin, Genfit, Gilead, HighTide, HistoIndex, Hepion, Intercept, Madrigal, Medpace, Metacrine, NGM Bio, NorthSea, Novartis, Novo Nordisk, Perspectum, Poxel, Prometic, Ridgeline Therapeutics, Sagimet, Terns, and Viking; grant/research support: Akero, Axcella, Bristol Myers Squibb, Cirius, CiVi Biopharma, Conatus, CymaBay, Enyo, Galectin, Galmed, Genentech, Genfit, Gilead, Hepion, HighTide, Immuron, Intercept, Madrigal, Metacrine, NGM Bio, NorthSea, Novartis, Novo Nordisk, Pfizer, Sagimet, Second Genome, Tobira/Allergan, and Viking. S.J.B.: consultant: Akcea, Amgen, Esperion, GLG Group, Guidepoint Global, Madrigal, Novartis, Novo Nordisk, Regeneron, and Sanofi; speaking and teaching: Amgen, Boehringer Ingelheim, Eli Lilly, and Esperion; advisory committee or review panel: Akcea, Alexion, Altimmune, Amgen, AstraZeneca, Esperion, Madrigal, Novartis, Regeneron, and Sanofi. N.T.G.: grant/research support: Axcella, Bristol Myers Squibb, CymaBay, Genentech, Genfit, Gilead, HighTide, Madrigal, NGM Bio, NorthSea, and Novo Nordisk; speaking and teaching: AbbVie, Dova, Gilead, Intercept, and Salix. Z.H.Y.: consultant: Gilead; grant/research support: Allergan, AXA, Bristol Myers Squibb, Conatus, Cymabay, Gilead, HighTide, Intercept, Madrigal, NGM Bio, Novartis, and Novo Nordisk; speaking and teaching: Intercept. A.K.: consultant: Gilead, Intercept, and Novartis; grant/research support: Gilead. R.P.: speaking and teaching: Intercept. M.K., H.C., J.Z., and M.V.C.: current employees of Axcella Health and own stock options in the company.

Clinical trial registry website and trial number: US National Library of Medicine (Ethics: The protocol was approved by a central institutional review board (IntegReview) before study initiation. All subjects provided written informed consent.

Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of The American College of Gastroenterology.

Figures

Figure 1.
Figure 1.
Study design (a) and subject disposition (b). aCalorie-matched placebo control; breasons for exclusion are presented in Supplementary Table (see Supplemental Digital Content 2, http://links.lww.com/AJG/C100); c10 subjects were randomized but not dosed and thus were not part of the safety population; and d3 subjects who received AXA1957 high dose in error on day 1 (randomized to AXA1957 low dose) were included in the AXA1957 high-dose safety analysis, considered part of the AXA1957 low-dose completer population, and excluded from both arms of the per-protocol analysis. b.i.d., 2 times a day; BMx, biomarkers; D, day; cT1, corrected T1; MRI-PDFF, MRI-proton density fat fraction; OGTT, oral glucose tolerance test; T2D, type 2 diabetes; W, week.
Figure 2.
Figure 2.
Change from baseline in liver fat content (MRI-PDFF) (a and b) and insulin resistance (HOMA-IR) (c) in the overall safety population. *P < 0.05 vs AXA1957 high dose. b.i.d., 2 times a day; D, day; HOMA-IR, homeostatic model assessment of insulin resistance; isocal, isocaloric; isoN, isonitrogenous; MRI-PDFF, MRI-proton density fat fraction; W, week.
Figure 3.
Figure 3.
Change from baseline in ALT (a and b) and K-18 M65 (c) in the overall safety population. *P < 0.05 vs placebo. ALT, alanine aminotransferase; K-18 M65, keratin-18 measured using M65 enzyme-linked immunosorbent assay.
Figure 4.
Figure 4.
Change from baseline in biomarkers of fibroinflammation (a, b, and c) in the overall safety population. *P < 0.05 vs placebo. b.i.d., 2 times a day; cT1, corrected T1; D, day; isocal, isocaloric; isoN, isonitrogenous; Pro-C3, N-terminal type III collagen propeptide; W, week.
Figure 5.
Figure 5.
Proportion of subjects with clinically relevant thresholds of biologic activity (a, b, and c) in the overall safety population. ALT, alanine aminotransferase; cT1, corrected T1; MRI-PDFF, MRI-proton density fat fraction.

References

    1. Do A, Lim JK. Epidemiology of nonalcoholic fatty liver disease: A primer. Clin Liver Dis (Hoboken) 2016;7:106–8.
    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.
    1. Younossi ZM, Golabi P, de Avila L, et al. . The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J Hepatol 2019;71:793–801.
    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.
    1. Friedman SL, Neuschwander-Tetri BA, Rinella M, et al. . Mechanisms of NAFLD development and therapeutic strategies. Nat Med 2018;24:908–22.
    1. Hamill MJ, Afeyan R, Chakravarthy MV, et al. . Endogenous metabolic modulators: Emerging therapeutic potential of amino acids. iScience 2020;23:101628.
    1. Muto Y, Sato S, Watanabe A, et al. . Effects of oral branched-chain amino acid granules on event-free survival in patients with liver cirrhosis. Clin Gastroenterol Hepatol 2005;3:705–13.
    1. Marchesini G, Bianchi G, Merli M, et al. . Nutritional supplementation with branched-chain amino acids in advanced cirrhosis: A double-blind, randomized trial. Gastroenterology 2003;124:1792–801.
    1. Malaguarnera M, Gargante MP, Russo C, et al. . L-carnitine supplementation to diet: A new tool in treatment of nonalcoholic steatohepatitis--a randomized and controlled clinical trial. Am J Gastroenterol 2010;105:1338–45.
    1. Mardinoglu A, Bjornson E, Zhang C, et al. . Personal model-assisted identification of NAD(+) and glutathione metabolism as intervention target in NAFLD. Mol Syst Biol 2017;13:916.
    1. Marukian S, Afeyan R, Tramontin T, et al. . AXA1125, a novel composition of amino acids reprograms the multifactorial pathophysiology in NAFLD [abstract 106]. Hepatology 2018;68(Suppl 1):67A.
    1. Lee CW, Afeyan R, Luithardt H, et al. . P02-05: AXA1125, a novel designed amino acid composition (DAAC), improves NAFLD Activity Score (NAS) and reduces fibrosis in two rodent models of nonalcoholic steatohepatitis (NASH). In: First EASL NAFLD Summit 2017. EASL: Rome, Italy, 2017.
    1. Pavlides M, Banerjee R, Tunnicliffe EM, et al. . Multiparametric magnetic resonance imaging for the assessment of non-alcoholic fatty liver disease severity. Liver Int 2017;37:1065–73.
    1. Loomba R, Neuschwander-Tetri BA, Sanyal A, et al. . Multicenter validation of association between decline in MRI-PDFF and histologic response in NASH. Hepatology 2020;72:1219–29.
    1. Loomba R, Sanyal AJ, Kowdley KV, et al. . Factors associated with histologic response in adult patients with nonalcoholic steatohepatitis. Gastroenterology 2019;156:88–95.e5.
    1. Dennis A, Kelly M, Fernandes C, et al. . Correlations between MRI biomarkers PDFF and cT1 with histopathological features of non-alcoholic steatohepatitis. Front Endocrinol (Lausanne) 2020;11:575843.
    1. Feldstein AE, Wieckowska A, Lopez AR, et al. . Cytokeratin‐18 fragment levels as noninvasive biomarkers for nonalcoholic steatohepatitis: A multicenter validation study. Hepatology 2009;50:1072–8.
    1. Banerjee R, Pavlides M, Tunnicliffe EM, et al. . Multiparametric magnetic resonance for the non-invasive diagnosis of liver disease. J Hepatol 2014;60:69–77.
    1. Luo Y, Oseini A, Gagnon R, et al. . An evaluation of the collagen fragments related to fibrogenesis and fibrolysis in nonalcoholic steatohepatitis. Sci Rep 2018;8:12414.
    1. Harrison SA, Rossi SJ, Paredes AH, et al. . NGM282 improves liver fibrosis and histology in 12 weeks in patients with nonalcoholic steatohepatitis. Hepatology 2020;71:1198–212.
    1. Chakravarthy M, Neuschwander-Tetri BA. The metabolic basis of nonalcoholic steatohepatitis. Endocrinol Diabetes Metab 2020;3:e00112.
    1. Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: The central role of nontriglyceride fatty acid metabolites. Hepatology 2010;52:774–88.
    1. Daou N, Viader A, Cokol M, et al. . A novel, multitargeted endogenous metabolic modulator composition impacts metabolism, inflammation, and fibrosis in nonalcoholic steatohepatitis-relevant primary human cell models. Sci Rep 2021;11(1):11861.
    1. Chakravarthy M, Harrison SA, Confer S, et al. . Mechanistic insights into the multimodal effects of AXA1125 in T2D subjects with NAFLD (abstract 2134). Hepatology 2019;70:1264A.
    1. Honda T, Ishigami M, Luo F, et al. . Branched-chain amino acids alleviate hepatic steatosis and liver injury in choline-deficient high-fat diet induced NASH mice. Metabolism 2017;69:177–87.
    1. Miyake T, Abe M, Furukawa S, et al. . Long-term branched-chain amino acid supplementation improves glucose tolerance in patients with nonalcoholic steatohepatitis-related cirrhosis. Intern Med 2012;51:2151–5.
    1. Badoud F, Lam KP, DiBattista A, et al. . Serum and adipose tissue amino acid homeostasis in the metabolically healthy obese. J Proteome Res 2014;13:3455–66.
    1. Zhang F, Zhao S, Yan W, et al. . Branched chain amino acids cause liver injury in obese/diabetic mice by promoting adipocyte lipolysis and inhibiting hepatic autophagy. EBioMedicine 2016;13:157–67.
    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.
    1. Ekstedt M, Hagstrom H, Nasr P, et al. . Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 2015;61:1547–54.
    1. Bril F, Leeming DJ, Karsdal MA, et al. . Use of plasma fragments of propeptides of type III, V, and VI procollagen for the detection of liver fibrosis in type 2 diabetes. Diabetes Care 2019;42:1348–51.
    1. Varasteh S, Braber S, Kraneveld AD, et al. . l-Arginine supplementation prevents intestinal epithelial barrier breakdown under heat stress conditions by promoting nitric oxide synthesis. Nutr Res 2018;57:45–55.
    1. Rao R, Samak G. Role of glutamine in protection of intestinal epithelial tight junctions. J Epithel Biol Pharmacol 2012;5:47–54.
    1. Sellmann C, Baumann A, Brandt A, et al. . Oral supplementation of glutamine attenuates the progression of nonalcoholic ateatohepatitis in C57BL/6J mice. J Nutr 2017;147:2041–9.
    1. Ezerina D, Takano Y, Hanaoka K, et al. . N-acetyl cysteine functions as a fast-acting antioxidant by triggering intracellular H2S and sulfane sulfur production. Cell Chem Biol 2018;25:447–59.
    1. de Andrade KQ, Moura FA, dos Santos JM, et al. . Oxidative stress and inflammation in hepatic diseases: Therapeutic possibilities of N-acetylcysteine. Int J Mol Sci 2015;16:30269–308.
    1. Khoshbaten M, Aliasgarzadeh A, Masnadi K, et al. . N-acetylcysteine improves liver function in patients with non-alcoholic fatty liver disease. Hepat Mon 2010;10:12–6.
    1. Borsheim E, Bui QU, Tissier S, et al. . Amino acid supplementation decreases plasma and liver triacylglycerols in elderly. Nutrition 2009;25:281–8.
    1. Coker RH, Deutz NE, Schutzler S, et al. . Nutritional supplementation with essential amino acids and phytosterols may reduce risk for metabolic syndrome and cardiovascular disease in overweight individuals with mild hyperlipidemia. J Endocrinol Diabetes Obes 2015;3:1069.
    1. Hurt RT, Ebbert JO, Schroeder DR, et al. . L-arginine for the treatment of centrally obese subjects: A pilot study. J Diet Suppl 2014;11:40–52.
    1. McClure EA, Baker NL, Gipson CD, et al. . An open-label pilot trial of N-acetylcysteine and varenicline in adult cigarette smokers. Am J Drug Alcohol Abuse 2015;41:52–6.
    1. Scarna A, Gijsman HJ, McTavish SF, et al. . Effects of a branched-chain amino acid drink in mania. Br J Psychiatry 2003;182:210–3.
    1. Ballestri S, Nascimbeni F, Romagnoli D, et al. . The independent predictors of non-alcoholic steatohepatitis and its individual histological features. Hep Res 2016;46:1074–87.

Source: PubMed

3
Iratkozz fel