Randomised clinical trial: A phase 2 double-blind study of namodenoson in non-alcoholic fatty liver disease and steatohepatitis

Rifaat Safadi, Marius Braun, Adi Francis, Yael Milgrom, Muhammad Massarwa, David Hakimian, Wadi Hazou, Assaf Issachar, Zivit Harpaz, Motti Farbstein, Inbal Itzhak, Naama Lev-Cohain, Avital Bareket-Samish, Michael H Silverman, Pnina Fishman, Rifaat Safadi, Marius Braun, Adi Francis, Yael Milgrom, Muhammad Massarwa, David Hakimian, Wadi Hazou, Assaf Issachar, Zivit Harpaz, Motti Farbstein, Inbal Itzhak, Naama Lev-Cohain, Avital Bareket-Samish, Michael H Silverman, Pnina Fishman

Abstract

Background: Namodenoson, an A3 adenosine receptor (A3AR) agonist, improved liver function/pathology in non-alcoholic steatohepatitis (NASH) preclinical models.

Aim: To evaluate the efficacy and safety of namodenoson for the treatment of non-alcoholic fatty liver disease (NAFLD) with or without NASH METHODS: This phase 2 study included 60 patients with NAFLD (ALT ≥60 IU/L) who were randomised (1:1:1) to oral namodenoson 12.5 mg b.d. (n = 21), 25 mg b.d. (n = 19), or placebo (n = 20) for 12 weeks (total follow-up: 16 weeks). The main efficacy endpoint involved serum ALT after 12 weeks of treatment.

Results: Serum ALT decreased over time with namodenoson in a dose-dependent manner. The difference between change from baseline (CFB) for ALT in the namodenoson 25 mg b.d. arm vs placebo trended towards significance at 12 weeks (P = 0.066). Serum AST levels also decreased with namodenoson in a dose-dependent manner; at 12 weeks, the CFB for 25 mg b.d. vs placebo was significant (P = 0.03). At Week 12, 31.6% in the namodenoson 25 mg b.d. arm and 20.0% in the placebo arm achieved ALT normalisation (P = 0.405). At week 16, the respective rates were 36.8% and 10.0% (P = 0.038). A3AR expression levels were stable over time across study arms. Both doses of namodenoson were well tolerated with no drug-emergent severe adverse events, drug-drug interactions, hepatotoxicity, or deaths. Three adverse events were considered possibly related to study treatment: myalgia (12.5 mg b.d. arm), muscular weakness (25 mg b.d. arm), and headache (25 mg b.d. arm).

Conclusion: A3AR is a valid target; namodenoson 25 mg b.d. was safe and demonstrated efficacy signals (ClinicalTrials.gov #NCT02927314).

Keywords: clinical trial; fibrosis; liver; non-alcoholic fatty liver disease; non-alcoholic steatohepatitis.

© 2021 Can-Fite BioPharma Ltd. Alimentary Pharmacology & Therapeutics published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Patient disposition
FIGURE 2
FIGURE 2
Changes from baseline (CFB) in inflammation‐related parameters. A, CFB in ALT levels over time. B, The proportion of patients achieving normalisation of ALT levels over time. C, CFB in AST levels over time. D, CFB in adiponectin levels at Week 12. Error bars represent SE P‐values for ALT and AST CFB were derived from ad hoc analyses
FIGURE 3
FIGURE 3
Changes from baseline in liver content and fibrosis‐related parameters. A, CFB in liver fat volume at Week 12. B, The proportion of patients with CAP ≥ 331 at screening and Week 12. C, CFB in FIB‐4 score at Week 12. D, The proportion of patients with NASH (FAST > 0.67) at screening and Week 12. E, CFB in FAST scores at Week 12. Error bars represent SE. Screening data were used as baseline
FIGURE 4
FIGURE 4
CFB in body weight with namodenoson treatment over time. Error bars represent SE P‐values were derived from ad hoc analyses

References

    1. Ratziu V, Bellentani S, Cortez‐Pinto H, et al. A position statement on NAFLD/NASH based on the EASL 2009 special conference. J Hepatol. 2010;53:372‐384.
    1. Williams CD, Stengel J, Asike MI, et al. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle‐aged population utilizing ultrasound and liver biopsy: a prospective study. Gastroenterology. 2011;140:124‐131.
    1. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of non‐alcoholic fatty liver disease: Practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology. 2012;55:2005‐2023.
    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;149:389‐397.
    1. Kabbany MN, Selvakumar PKC, Watt K, et al. Prevalence of nonalcoholic steatohepatitis‐associated cirrhosis in the United States: an analysis of national health and nutrition examination survey data. Am J Gastroenterol. 2017;112:581‐587.
    1. Liu A, Galoosian A, Kaswala D, et al. Nonalcoholic fatty liver disease: epidemiology, liver transplantation trends and outcomes, and risk of recurrent disease in the graft. J Clin Transl Hepatol. 2018;6:420‐424.
    1. Vuppalanchi R, Noureddin M, Alkhouri N, et al. Therapeutic pipeline in nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol. 2021;18:373‐392.
    1. Takahashi Y, Sugimoto K, Inui H, et al. Current pharmacological therapies for nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Gastroenterol. 2015;21:3777‐3785.
    1. Guillaume M, Ratziu V. Pharmacological agents for nonalcoholic steatohepatitis. Hepatol Int. 2013;7:833‐841.
    1. Ratziu V, Goodman Z, Sanyal A. Current efforts and trends in the treatment of NASH. J Hepatol. 2015;62:S65‐75.
    1. Cave MC, Clair HB, Hardesty JE, et al. Nuclear receptors and nonalcoholic fatty liver disease. Biochim Biophys Acta. 2016;1859:1083‐1099.
    1. Ohana G, Cohen S, Rath‐Wolfson L, et al. A3 adenosine receptor agonist, CF102, protects against hepatic ischemia/reperfusion injury following partial hepatectomy. Mol Med Rep. 2016;14:4335‐4341.
    1. Cohen S, Stemmer SM, Zozulya G, et al. CF102 an A3 adenosine receptor agonist mediates anti‐tumor and anti‐inflammatory effects in the liver. J Cell Physiol. 2011;226:2438‐2447.
    1. Bar‐Yehuda S, Stemmer SM, Madi L, et al. The A3 adenosine receptor agonist CF102 induces apoptosis of hepatocellular carcinoma via de‐regulation of the Wnt and NF‐kappaB signal transduction pathways. Int J Oncol. 2008;33:287‐295.
    1. Stemmer SM, Benjaminov O, Medalia G, et al. CF102 for the treatment of hepatocellular carcinoma: a phase I/II, open‐label, dose‐escalation study. Oncologist. 2013;18:25‐26.
    1. Stemmer SM, Manojlovic NS, Marinca MV, et al. Namodenoson in advanced hepatocellular carcinoma and Child‐Pugh B cirrhosis: randomized placebo‐controlled clinical trial. Cancers. 2021;13:187.
    1. Fishman P, Cohen S, Itzhak I, et al. The A3 adenosine receptor agonist, namodenoson, ameliorates nonalcoholic steatohepatitis in mice. Int J Mol Med. 2019;44:2256‐2264.
    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;5:362‐373.
    1. Glass LM, Hunt CM, Fuchs M, et al. Comorbidities and nonalcoholic fatty liver disease: the chicken, the egg, or both? Fed Pract. 2019;36:64‐71.
    1. Cross HR, Murphy E, Black RG, et al. Overexpression of A(3) adenosine receptors decreases heart rate, preserves energetics, and protects ischemic hearts. Am J Physiol Heart Circ Physiol. 2002;283:H1562‐1568.
    1. Chen G‐J, Harvey BK, Shen H, et al. Activation of adenosine A3 receptors reduces ischemic brain injury in rodents. J Neurosci Res. 2006;84:1848‐1855.
    1. Liraglutide [Package Insert]. Novo Nordisk Inc; 2020.
    1. Bünemann M, Lee KB, Pals‐Rylaarsdam R, et al. Desensitization of G‐protein‐coupled receptors in the cardiovascular system. Annu Rev Physiol. 1999;61:169‐192.

Source: PubMed

3
Abonnere