Safety, tolerability, and anti-fibrotic efficacy of the CBP/β-catenin inhibitor PRI-724 in patients with hepatitis C and B virus-induced liver cirrhosis: An investigator-initiated, open-label, non-randomised, multicentre, phase 1/2a study

Kiminori Kimura, Tatsuya Kanto, Shinji Shimoda, Kenichi Harada, Masamichi Kimura, Koji Nishikawa, Jun Imamura, Eiichi Ogawa, Masanao Saio, Yoshihiro Ikura, Takuji Okusaka, Kazuaki Inoue, Tetsuya Ishikawa, Ichiro Ieiri, Junji Kishimoto, Koji Todaka, Terumi Kamisawa, Kiminori Kimura, Tatsuya Kanto, Shinji Shimoda, Kenichi Harada, Masamichi Kimura, Koji Nishikawa, Jun Imamura, Eiichi Ogawa, Masanao Saio, Yoshihiro Ikura, Takuji Okusaka, Kazuaki Inoue, Tetsuya Ishikawa, Ichiro Ieiri, Junji Kishimoto, Koji Todaka, Terumi Kamisawa

Abstract

Background: We conducted an exploratory study to assess the safety tolerability, and anti-fibrotic effects of PRI-724, a CBP/β-catenin inhibitor, in patients with hepatitis C virus (HCV)- and hepatitis B virus (HBV)-induced cirrhosis.

Methods: This multicentre, open-label, non-randomised, non-placebo-controlled phase 1/2a trial was conducted at three hospitals in Japan. Between July 27, 2018, and July 13, 2021, we enrolled patients with HCV- and HBV-induced cirrhosis classified as Child-Pugh (CP) class A or B. In phase 1, 15 patients received intravenous infusions of PRI-724 at escalating doses of 140, 280, and 380 mg/m2/4 h twice weekly for 12 weeks. In phase 2a, 12 patients received the recommended PRI-724 dose. The primary endpoints of phases 1 and 2a were the frequency and severity of adverse events and efficacy in treating cirrhosis based on liver biopsy. This study was registered at ClinicalTrials.gov (no. NCT03620474).

Findings: Three patients from phase 1 who received the recommended PRI-724 dose were evaluated to obtain efficacy and safety data in phase 2a. Serious adverse events occurred in three patients, one of which was possibly related to PRI-724. The most common adverse events were diarrhoea and nausea. PRI-724 did not decrease hepatic fibrosis with any statistical significance, either by ordinal scoring or measurement of collagen proportionate area at 12 weeks; however, we observed statistically significant improvements in liver stiffness, Model for End-stage Liver Disease score, and serum albumin level.

Interpretation: Intravenous administration of 280 mg/m2/4 h PRI-724 over 12 weeks was preliminarily assessed to be well tolerated; however, further evaluation of anti-fibrotic effects in patients with cirrhosis is warranted.

Funding: AMED, Ohara Pharmaceutical.

Keywords: Anti-fibrotic drug; HBV; HCV; Liver cirrhosis; PRI-724.

Conflict of interest statement

There are no conflicts of interest.

Copyright © 2022 The Author(s). Published by Elsevier B.V. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile (Phase 1/2a). Consort diagram.
Figure 2
Figure 2
Pharmacokinetics of PRI-724 and C-82 after intravenous infusion of PRI-724 (Phase 1). (a) Mean concentration profiles of PRI-724 in plasma from patients with HCV and HBV liver cirrhosis during intravenous infusion of PRI-724. (b) Mean plasma concentration profiles of C-82, a PRI-724 metabolite, in the same patients.
Figure 3
Figure 3
Effects of PRI-724 treatment on liver function and liver stiffness measure (Phase 2a).Changes in (a) serum ALB, PT, and TBIL before, during, and after 12 weeks of PRI-724 administration. (b) Change in LSM from baseline to post-treatment following 12 weeks of PRI-724 administration (n = 13). The graph (right) shows the results of analysing patients with liver cirrhosis and a baseline LSM of ≥ 20 kPa. Statistical analyses were performed using paired t-tests.

References

    1. Gines P, Krag A, Abraldes JG, Sola E, Fabrellas N, Kamath PS. Liver cirrhosis. Lancet. 2021;398(10308):1359–1376.
    1. Schuppan D, Afdhal NH. Liver cirrhosis. Lancet. 2008;371(9615):838–851.
    1. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115(2):209–218.
    1. Karlsen TH, Sheron N, Zelber-Sagi S, et al. The EASL-Lancet Liver Commission: protecting the next generation of Europeans against liver disease complications and premature mortality. Lancet. 2022;399(10319):61–116.
    1. Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380(15):1450–1462.
    1. Rockey DC, Friedman SL. Fibrosis regression after eradication of hepatitis C virus: from bench to bedside. Gastroenterology. 2021;160(5) 1502-20 e1.
    1. Friedman SL, Pinzani M. Hepatic fibrosis 2022: Unmet needs and a blueprint for the future. Hepatology. 2022;75(2):473–488.
    1. Loomba R, Noureddin M, Kowdley KV, et al. Combination therapies including Cilofexor and Firsocostat for bridging fibrosis and cirrhosis attributable to NASH. Hepatology. 2021;73(2):625–643.
    1. Rinella ME, Tacke F, Sanyal AJ, Anstee QM, participants of the AEW Report on the AASLD/EASL Joint Workshop on Clinical Trial Endpoints in NAFLD. Hepatology. 2019;70(4):1424–1436.
    1. 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.
    1. 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.
    1. Nusse R, Clevers H. Wnt/beta-Catenin signaling, disease, and emerging therapeutic modalities. Cell. 2017;169(6):985–999.
    1. Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell. 2012;149(6):1192–1205.
    1. Konigshoff M, Eickelberg O. WNT signaling in lung disease: a failure or a regeneration signal? Am J Respir Cell Mol Biol. 2010;42(1):21–31.
    1. Schunk SJ, Floege J, Fliser D, Speer T. WNT-beta-catenin signalling - a versatile player in kidney injury and repair. Nat Rev Nephrol. 2021;17(3):172–184.
    1. Thompson MD, Monga SP. WNT/beta-catenin signaling in liver health and disease. Hepatology. 2007;45(5):1298–1305.
    1. Guo Y, Xiao L, Sun L, Liu F. Wnt/beta-catenin signaling: a promising new target for fibrosis diseases. Physiol Res. 2012;61(4):337–346.
    1. Hao S, He W, Li Y, et al. Targeted inhibition of beta-catenin/CBP signaling ameliorates renal interstitial fibrosis. J Am Soc Nephrol. 2011;22(9):1642–1653.
    1. Henderson WR, Jr., Chi EY, Ye X, et al. Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc Natl Acad Sci U S A. 2010;107(32):14309–14314.
    1. Tokunaga Y, Osawa Y, Ohtsuki T, et al. Selective inhibitor of Wnt/beta-catenin/CBP signaling ameliorates hepatitis C virus-induced liver fibrosis in mouse model. Sci Rep. 2017;7(1):325.
    1. Osawa Y, Oboki K, Imamura J, et al. Inhibition of cyclic adenosine monophosphate (cAMP)-response element-binding protein (CREB)-binding protein (CBP)/beta-catenin reduces liver fibrosis in mice. EBioMedicine. 2015;2(11):1751–1758.
    1. Nishikawa K, Osawa Y, Kimura K. Wnt/beta-Catenin signaling as a potential target for the treatment of liver cirrhosis using antifibrotic drugs. Int J Mol Sci. 2018;19(10)
    1. Kimura K, Ikoma A, Shibakawa M, et al. Safety, tolerability, and preliminary efficacy of the anti-fibrotic small molecule PRI-724, a CBP/beta-catenin inhibitor, in patients with hepatitis C virus-related cirrhosis: a single-center, open-label, dose escalation phase 1 trial. EBioMedicine. 2017;23:79–87.
    1. D'Ambrosio R, Aghemo A, Rumi MG, et al. A morphometric and immunohistochemical study to assess the benefit of a sustained virological response in hepatitis C virus patients with cirrhosis. Hepatology. 2012;56(2):532–543.
    1. Verna EC, Morelli G, Terrault NA, et al. DAA therapy and long-term hepatic function in advanced/decompensated cirrhosis: real-world experience from HCV-TARGET cohort. J Hepatol. 2020;73(3):540–548.
    1. Lau DT, Luxon BA, Xiao SY, Beard MR, Lemon SM. Intrahepatic gene expression profiles and alpha-smooth muscle actin patterns in hepatitis C virus induced fibrosis. Hepatology. 2005;42(2):273–281.
    1. Guido M, Rugge M, Leandro G, Fiel IM, Thung SN. Hepatic stellate cell immunodetection and cirrhotic evolution of viral hepatitis in liver allografts. Hepatology. 1997;26(2):310–314.
    1. Younossi ZM, Anstee QM, Wai-Sun Wong V, et al. The association of histologic and noninvasive tests with adverse clinical and patient-reported outcomes in patients with advanced fibrosis due to nonalcoholic steatohepatitis. Gastroenterology. 2021;160(5) 1608-19 e13.
    1. Xiao G, Zhu S, Xiao X, Yan L, Yang J, Wu G. Comparison of laboratory tests, ultrasound, or magnetic resonance elastography to detect fibrosis in patients with nonalcoholic fatty liver disease: A meta-analysis. Hepatology. 2017;66(5):1486–1501.
    1. Elsharkawy A, Abdel Alem S, Fouad R, et al. Changes in liver stiffness measurements and fibrosis scores following Sofosbuvir based treatment regimens without Interferon. J Gastroenterol Hepatol. 2017
    1. de Franchis R, Bosch J, Garcia-Tsao G, Reiberger T, Ripoll C, Baveno VIIF. Baveno VII - Renewing consensus in portal hypertension. J Hepatol. 2021
    1. Moreno C, Mueller S, Szabo G. Non-invasive diagnosis and biomarkers in alcohol-related liver disease. J Hepatol. 2019;70(2):273–283.
    1. Foster GR, Irving WL, Cheung MC, et al. Impact of direct acting antiviral therapy in patients with chronic hepatitis C and decompensated cirrhosis. J Hepatol. 2016;64(6):1224–1231.
    1. Chalasani N, Abdelmalek MF, Garcia-Tsao G, et al. Effects of Belapectin, an inhibitor of Galectin-3, in patients with nonalcoholic steatohepatitis with cirrhosis and portal hypertension. Gastroenterology. 2020;158(5) 1334-45 e5.
    1. Harrison SA, Ruane PJ, Freilich BL, et al. Efruxifermin in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a trial. Nat Med. 2021;27(7):1262–1271.

Source: PubMed

3
Se inscrever