Randomised clinical trial: Pemafibrate, a novel selective peroxisome proliferator-activated receptor α modulator (SPPARMα), versus placebo in patients with non-alcoholic fatty liver disease

Atsushi Nakajima, Yuichiro Eguchi, Masato Yoneda, Kento Imajo, Nobuharu Tamaki, Hideki Suganami, Toshiaki Nojima, Ryohei Tanigawa, Masakazu Iizuka, Yuki Iida, Rohit Loomba, Atsushi Nakajima, Yuichiro Eguchi, Masato Yoneda, Kento Imajo, Nobuharu Tamaki, Hideki Suganami, Toshiaki Nojima, Ryohei Tanigawa, Masakazu Iizuka, Yuki Iida, Rohit Loomba

Abstract

Background: Pemafibrate is a novel, selective peroxisome proliferator-activated receptor α modulator (SPPARMα). In mice, Pemafibrate improved the histological features of non-alcoholic steatohepatitis (NASH). In patients with dyslipidaemia, it improved serum alanine aminotransferase (ALT).

Aims: To evaluate the efficacy and safety of Pemafibrate in patients with high-risk, non-alcoholic fatty liver disease (NAFLD).

Methods: This double-blind, placebo-controlled, randomised multicentre, phase 2 trial randomised 118 patients (1:1) to either 0.2 mg Pemafibrate or placebo, orally, twice daily for 72 weeks. The key inclusion criteria included liver fat content of ≥10% by magnetic resonance imaging-estimated proton density fat fraction (MRI-PDFF); liver stiffness of ≥2.5 kPa, by magnetic resonance elastography (MRE); and elevated ALT levels. The primary endpoint was the percentage change in MRI-PDFF from baseline to week 24. The secondary endpoints included MRE-based liver stiffness, ALT, serum liver fibrosis markers and lipid parameters.

Results: There was no significant difference between the groups in the primary endpoint (-5.3% vs -4.2%; treatment difference -1.0%, P = 0.85). However, MRE-based liver stiffness significantly decreased compared to placebo at week 48 (treatment difference -5.7%, P = 0.036), and was maintained at week 72 (treatment difference -6.2%, P = 0.024), with significant reduction in ALT and LDL-C. Adverse events were comparable between the treatment groups and therapy was well tolerated.

Conclusions: Pemafibrate did not decrease liver fat content but had significant reduction in MRE-based liver stiffness. Pemafibrate may be a promising therapeutic agent for NAFLD/NASH, and also be a candidate for combination therapy with agents that reduce liver fat content. ClinicalTrials.gov, number: NCT03350165.

© 2021 The Authors. Alimentary Pharmacology & Therapeutics published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Flow diagram of patient disposition
FIGURE 2
FIGURE 2
Percentage change from baseline to 72 weeks in liver fat content by MRI‐PDFF (A) and liver stiffness by MRE (B). Data are expressed as least square mean. Error bars show 95% CI. *P < 0.05 vs placebo. Representative images of MRE of a patient (C). ROIs, the areas surrounded by yellow line, were set in the right lobe of the liver in accordance with the instruction in the prespecified manual [Colour figure can be viewed at wileyonlinelibrary.com]
FIGURE 3
FIGURE 3
Measured levels of liver (A‐D) and renal function (E, F) markers over 72 weeks. Data are expressed as mean. Error bars show SD. ALT, alanine aminotransferase; AST, aspartate aminotransferase; GGT, γ‐glutamyl transpeptidase; ALP, alkaline phosphatase; eGFR, estimated glomerular filtration rate [Colour figure can be viewed at wileyonlinelibrary.com]
FIGURE 4
FIGURE 4
Measured levels of lipid markers over 72 weeks. Data are expressed as mean. Error bars show SD. TC, total cholesterol; LDL‐C, low‐density lipoprotein‐cholesterol; HDL‐C, high‐density lipoprotein‐cholesterol; TG, triglyceride [Colour figure can be viewed at wileyonlinelibrary.com]

References

    1. Chalasani N, Younossi Z, Lavine Joel E, 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(6):2005‐2023.
    1. National Guideline Centre . NICE Guideline. 2016.
    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(1):73‐84.
    1. Lonardo A, Nascimbeni F, Mantovani A, et al. Hypertension, diabetes, atherosclerosis and NASH: cause or consequence? J Hepatol. 2018;68(2):335‐352.
    1. Sumida Y, Yoneda M. Current and future pharmacological therapies for NAFLD/NASH. J Gastroenterol. 2018;53(3):362‐376.
    1. Yamashita S, Masuda D, Matsuzaka Y. Pemafibrate, a new selective PPARα modulator: drug concept and its clinical applications for dyslipidemia and metabolic diseases. Curr Atheroscler Rep. 2020;22(1):5.
    1. Francque S, Verrijken A, Caron S, et al. PPARα gene expression correlates with severity and histological treatment response in patients with non‐alcoholic steatohepatitis. J Hepatol. 2015;63(1):164‐173.
    1. Fruchart J‐C. Pemafbrate (K‐877), a novel selective peroxisome proliferator‐activated receptor alpha modulator for management of atherogenic dyslipidaemia. Cardiovasc Diabetol. 2017;16:124.
    1. Pawlak M, Lefebvre P, Staels B. Molecular mechanism of PPARα action and its impact on lipid metabolism, inflammation and fibrosis in non‐alcoholic fatty liver disease. J Hepatol. 2015;62(3):720‐733.
    1. Honda Y, Kessoku T, Ogawa Y, et al. Pemafibrate, a novel selective peroxisome proliferator‐activated receptor alpha modulator, improves the pathogenesis in a rodent model of nonalcoholic steatohepatitis. Sci Rep. 2017;14(7):42477.
    1. Dulai PS, Sirlin CB, Loomba R. MRI and MRE for non‐invasive quantitative assessment of hepatic steatosis and fibrosis in NAFLD and NASH: clinical trials to clinical practice. J Hepatol. 2016;65(5):1006‐1016.
    1. Hsu C, Caussy C, Imajo K, et al. Magnetic resonance vs transient elastography analysis of patients with nonalcoholic fatty liver disease: a systematic review and pooled analysis of individual participants. Clin Gastroenterol Hepatol. 2019;17(4):630‐637.e8.
    1. Patel J, Bettencourt R, Cui J, et al. Association of noninvasive quantitative decline in liver fat content on MRI with histologic response in nonalcoholic steatohepatitis. Therap Adv Gastroenterol. 2016;9(5):692‐701.
    1. Loomba R, Lawitz E, Ghalib R, et al. SAT‐489—longitudinal changes in liver stiffness by magnetic resonance elastography (MRE), liver fibrosis, and serum markers of fibrosis in a multi‐center clinical trial in nonalcoholic steatohepatitis (NASH). J Hepatol. 2017;66(1):S671.
    1. Ajmera VH, Liu A, Singh S, et al. Clinical utility of an increase in magnetic resonance elastography in predicting fibrosis progression in nonalcoholic fatty liver disease. Hepatology. 2020;71(3):849‐860.
    1. 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 randomized, double‐blind, placebo‐controlled, phase 2a trial. Lancet. 2019;392(10165):2705‐2717.
    1. Sasaki Y, Asahiyama M, Tanaka T, et al. Pemafibrate, a selective PPARα modulator, prevents non‐alcoholic steatohepatitis development without reducing the hepatic triglyceride content. Sci Rep. 2020;10(1):7818.
    1. Matsuba I, Matsuba R, Yamashita S, et al. Effects of a novel selective peroxisome proliferator‐activated receptor‐a modulator, Pemafibrate, on hepatic and peripheral glucose uptake in patients with hypertriglyceridemia and insulin resistance. J Diabetes Investig. 2018;9(6):1323‐1332.
    1. Saponaro C, Gaggini M, Carli F, et al. The subtle balance between lipolysis and lipogenesis: a critical point in metabolic homeostasis. Nutrients. 2015;7(11):9453‐9474.
    1. Imajo K, Kessoku T, Honda Y, et al. Magnetic resonance imaging more accurately classifies steatosis and fibrosis in patients with nonalcoholic fatty liver disease than transient elastography. Gastroenterology. 2016;150(3):626.e7‐637.e7.
    1. 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.
    1. Tamaki N, Higuchi M, Kurosaki M, et al. Wisteria floribunda agglutinin‐positive mac‐2 binding protein as an age‐independent fibrosis marker in nonalcoholic fatty liver disease. Sci Rep. 2019;9(1):10109.
    1. Ekstedt M, Hagström 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(5):1547‐1554.
    1. Ratziu V, Harrison SA, Francque S, et al. Elafibranor, an agonist of the peroxisome proliferator‐activated receptor‐α and ‐δ, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening. Gastroenterology. 2016;150(5):1147.e5‐1159.e5.
    1. Stephen AH, Rinella ME, Abdelmalek MF, et al. NGM282 for treatment of non‐alcoholic steatohepatitis: a multicentre, randomised, double‐blind, placebo‐controlled, phase 2 trial. Lancet. 2018;391(10126):1174‐1185.
    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(5):1463.e6‐1473.e6.
    1. Camont L, Lhomme M, Rached F, et al. Small, dense high‐density lipoprotein‐3 particles are enriched in negatively charged phospholipids: relevance to cellular cholesterol efflux, antioxidative, antithrombotic, anti‐inflammatory, and antiapoptotic functionalities. Arterioscler Thromb Vasc Biol. 2013;33(12):2715‐2723.
    1. Hennuyer N, Duplan I, Paquet C, et al. The novel selective PPARα modulator (SPPARMα) Pemafibrate improves dyslipidemia, enhances reverse cholesterol transport and decreases inflammation and atherosclerosis. Atherosclerosis. 2016;249:200‐208.
    1. Yamashita S, Arai H, Yokote K, et al. Effects of Pemafibrate (K‐877) on cholesterol efflux capacity and postprandial hyperlipidemia in patients with atherogenic dyslipidemia. J Clin Lipidol. 2018;12(5):1267.e4‐1279.e4.
    1. Mantovani A, Petracca G, Beatrice G, et al. Non‐alcoholic fatty liver disease and risk of incident chronic kidney disease: an updated meta‐analysis. Gut. 2020;gutjnl‐2020‐323082.
    1. Stine JG, Munaganuru N, Barnard A, et al. Change in MRI‐PDFF and histologic response in patients with nonalcoholic steatohepatitis: a systematic review and meta‐analysis. Clin Gastroenterol Hepatol. 2020;S1542–3565(20):31220‐31229.

Source: PubMed

3
Abonnieren