eISSN: 2449-8238
ISSN: 2392-1099
Clinical and Experimental Hepatology
Current issue Archive Manuscripts accepted About the journal Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2022
vol. 8
 
Share:
Share:
Original paper

Pemafibrate therapy for non-alcoholic fatty liver disease is more effective in lean patients than obese patients

Satoshi Shinozaki
1, 2
,
Toshiyuki Tahara
3
,
Kouichi Miura
2
,
Alan Kawarai Lefor
4
,
Hironori Yamamoto
2

1.
Shinozaki Medical Clinic, Japan
2.
Department of Medicine, Division of Gastroenterology, Jichi Medical University, Tochigi, Japan
3.
Saiseikai Utsunomiya Hospital, Tochigi, Japan
4.
Department of Surgery, Jichi Medical University, Tochigi, Japan
Clin Exp HEPATOL 2022; 8, 4: 278-283
Online publish date: 2022/11/29
Article file
- Pemafibrate.pdf  [0.11 MB]
Get citation
 
PlumX metrics:
 

Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most common type of chronic liver disease in the world, and its incidence is currently increasing. The prevalence of NAFLD is 23.7% and 27.4% in Europe and Asia, respectively [1]. The prevalence is predicted to increase to 15-56% [2]. NAFLD includes non-alcoholic fatty liver and non-alcoholic steatohepatitis (NASH). The pathogenesis of NAFLD involves steatosis, inflammation and fibrosis, and the degree of fibrosis determines the patients’ long-term prognosis. A recent systematic review reported that progression of fibrosis in patients with NAFLD increases all-cause mortality and liver-related mortality [3]. Monitoring the fibrosis staging and development is important to predict the long-term prognosis of patients with NAFLD, and therefore the suppression of hepatic fibrosis should be the primary consideration in the treatment of patients with NAFLD.

Pemafibrate, a novel selective peroxisome proliferator-activated receptor α (PPAR-α) modulator, was made available in Japan in 2018. Pemafibrate has the potential to prevent cirrhosis and liver failure by inhibiting persistent hepatic inflammation and fibrosis due to NAFLD/NASH. We previously reported an improvement of hepatic markers of inflammation and fibrosis in both short-term and long-term studies [4, 5]. In routine clinical practice, we sometimes encounter patients with NAFLD who are refractory to pemafibrate therapy. There are no reports regarding predictors of improvement with pemafibrate therapy in patients with NAFLD, although in some studies correlation analyses were performed [4-7]. Identification of predictors for susceptibility to pemafibrate therapy is important to determine optimal treatment strategies such as dose escalation and/or the use of additional medications. The aim of this study is to identify predictors for improvement of hepatic inflammation and fibrosis after pemafibrate therapy in patients with NAFLD.

Material and methods

Study population

This dual-center retrospective study included patients with NAFLD with dyslipidemia who started treatment with pemafibrate between June 2019 and November 2021 at the Saiseikai Utsunomiya Hospital and the Shinozaki Medical Clinic. Inclusion criteria were: 1) fatty liver diagnosed by abdominal ultrasound, 2) dyslipidemia treated with pemafibrate, 3) sustained alanine aminotransferase (ALT) elevation > 30 U/l for more than three months before starting pemafibrate therapy, 4) negative hepatitis B surface antigen or anti-hepatitis C antibody, 5) normal serum immunoglobulin-G level, 6) alcohol consumption < 30 g/day in males and < 20 g/day in females [4, 5]. Exclusion criteria were: 1) severe chronic kidney disease (estimated glomerular filtration rate < 30 ml/min/1.73 m2), 2) history of previous pemafibrate use, 3) cessation of pemafibrate therapy within six months or 4) patients with diabetes mellitus. This retrospective observational study was approved by the Institutional Review Board of both centers.

Predictors of response of hepatic inflammation and fibrosis

Improvement of hepatic inflammation was evaluated by reduction of ALT level, which is a representative validated marker associated with progression of NAFLD toward hepatic fibrosis [8, 9]. The definition of response of inflammation was 50% or more decrease of ALT levels at six months compared to baseline. Improvement of hepatic fibrosis was assessed by reduction of Mac-2 binding protein glycosylation isomer (M2BPGi) reflecting the grade of hepatic fibrosis [10]. The definition of response of fibrosis was 20% or more reduction in M2BPGi levels at six months compared to the baseline.

Statistical analysis

Changes in parameters after six months of pemafibrate therapy were assessed with the Wilcoxon rank sum test. Cut-off values were determined by receiver operating characteristic curve except for the cut-off value of body mass index (BMI), which was determined by the definition of lean NAFLD (BMI < 25) [11]. To evaluate predictors of response, univariate and multivariate logistic regression analysis were used. To perform multivariate analysis, factors were selected based on clinical significance. Data for lean and non-lean NAFLD were compared using the Mann-Whitney U test. Differences were considered significant when p < 0.05. StatFlex 7.0 software (Artech Co., Ltd., Osaka, Japan) was used.

Results

Baseline characteristics and changes in parameters

One hundred two patients fulfilled the inclusion criteria, and 31 patients were excluded for the following reasons: lost to follow-up (n = 23) and diabetes (n = 8). Consequently, the remaining 71 patients were analyzed (Table 1). All patients were treated with pemafibrate 0.1 mg twice daily without dose escalations. Approximately half of the patients had been treated with a statin before starting pemafibrate, and pemafibrate was given in addition. There was no combination treatment with vitamin E or ursodeoxycholic acid. No adverse events were observed. During six months of pemafibrate therapy, significant improvements were observed in aspartate aminotransferase (AST), ALT, γ-glutamyl transpeptidase (γ-GTP), platelet count, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, triglyceride, total bilirubin, serum albumin, albumin-bilirubin score and M2BPGi (Table 2).

Table 1

Patient baseline characteristics

ParameterN = 71
Age (years), mean ± standard error50.7 ±1.6
Gender (male), n (%)48 (68)
Current smoker, n (%)5 (7)
Complications treated with medications, n (%)
Hypertension21 (30)
Gastroesophageal reflux disease17 (24)
Diabetes mellitus0 (0)
Concurrent medications, n (%)
Statins40 (56)
Ezetimibe10 (14)
Angiotensin II receptor blockers7 (10)
Eicosapentaenoic acid/docosahexaenoic acid1 (1)
Table 2

Changes in clinical parameters after six months of pemafibrate therapy

ParameterBaseline6 monthsp-value
Body mass index, mean ± SE28.3 ±0.428.5 ±0.40.160
AST (U/l)49.2 ±2.841.1 ±2.5< 0.001
ALT (U/l)78.2 ±5.552.0 ±5.0< 0.001
γ-GTP (U/l)74.5 ±7.439.6 ±3.8< 0.001
Platelet count (×104/µl)26.5 ±0.528.6 ±0.6< 0.001
Estimated GFR (ml/min/ 1.73 m2)79.9 ±2.078.0 ±2.00.066
LDL cholesterol (mg/dl)104.2 ±3.495.3 ±3.2< 0.001
HDL cholesterol (mg/dl)50.2 ±1.652.4 ±1.30.021
Triglyceride (mg/dl)179.4 ±20.7111.1 ±8.2< 0.001
Uric acid (mg/dl)6.0 ±0.16.0 ±0.10.724
Total bilirubin (mg/dl)0.91 ±0.050.75 ±0.04< 0.001
Serum albumin (g/dl)4.4 ±0.034.5 ±0.03< 0.001
ALBI score–3.00 ±0.03–3.18 ±0.03< 0.001
M2BPGi0.67 ±0.040.56 ±0.03< 0.001

[i] SD – standard error, AST – aspartate aminotransferase, ALT – alanine aminotransferase, γ-GTP – γ-glutamyl transpeptidase, GFR – glomerular filtration rate, LDL – low-density lipoprotein, HDL – high-density lipoprotein, ALBI – albumin-bilirubin, M2BPGi – Mac-2 binding protein glucosylation isomer

Predictors of improvement of hepatic inflammation

Changes in serum ALT levels, a serum marker for hepatic inflammation, were evaluated (Table 3). A greater than 50% decrease in serum ALT levels was found in 35% (25/71) of patients. In univariate analysis, lean NAFLD (BMI < 25) was identified as a significant positive predictor of response to treatment. Subsequent multivariate analysis confirmed this result.

Table 3

Predictors for reduction of serum alanine aminotransferase (ALT) level after pemafibrate therapy

FactorUnivariate analysisMultivariate analysis
Odds ratio95% confidence intervalp-valueOdds ratio95% confidence intervalp-value
Age, > 50 years1.3880.522-3.6950.5111.0350.307-3.4910.956
Male, gender0.5910.212-1.6480.314
Body mass index, < 254.6121.339-15.8830.0155.5571.421-21.7310.013
Statin use1.6300.599-4.4330.338
AST, > 50 U/l1.9950.721-5.5180.183
ALT, > 60 U/l1.7850.643-4.9560.2662.0550.584-7.2320.261
γ-GTP, > 70 U/l2.5000.883-7.0760.084
Platelet count, > 270 × 103/µl1.2220.456-3.2800.690
Estimated GFR, > 70 ml/min/1.73 m21.8100.632-5.1810.269
Uric acid, > 6 mg/dl0.4730.174-1.2870.142
LDL cholesterol, > 100 mg/dl1.2730.478-3.3860.629
HDL cholesterol, > 50 mg/dl0.7310.268-1.9940.540
Triglyceride, > 120 mg/dl1.4950.537-4.1680.441
Serum albumin, > 4.4 g/dl0.3620.130-1.0070.051
Hemoglobin A1c, 5.6%0.6040.226-1.6130.314
M2BPGi, > 0.73.4471.222-9.7190.0192.8740.833-9.9170.094

[i] ARB – angiotensin II receptor blocker, AST – aspartate aminotransferase,γγ-GTP – γ-glutamyl transpeptidase, GFR – glomerular filtration rate, LDL – low-density lipoprotein, HDL – high-density lipoprotein, M2BPGi – Mac-2 binding protein glucosylation isomer

Next, patients were categorized into three groups based on BMI (lean < 25, overweight 25-30 and obese > 30) (Fig. 1A). Compared with baseline levels, all groups showed a significant reduction in serum ALT levels after six months of pemafibrate therapy (lean p < 0.001, overweight p < 0.001 and obese p = 0.001). The reduction of serum ALT in the lean NAFLD group was significantly greater than in the obese NAFLD group (p = 0.034), consistent with a greater improvement in hepatic inflammation.

Fig. 1

Reductions in serum markers after six months of pemafibrate therapy according to body mass index (BMI): A) alanine aminotransferase (ALT), B) Mac-2 binding protein glycosylation isomer (M2BPGi). Bar: standard error. All differences among groups were evaluated, but only statistically significant results are shown

/f/fulltexts/CEH/47958/CEH-8-47958-g001_min.jpg

Predictors of improvement of hepatic fibrosis

Changes in serum M2BPGi levels, a marker of hepatic fibrosis, were assessed after treatment (Table 4). A greater than 20% reduction in serum M2BPGi levels was found in 51% (36/71) of patients. In univariate analysis, lean NAFLD (BMI < 25) and age > 50 years were identified as significant positive predictors of response to treatment. Subsequent multivariate analysis confirmed this result.

Table 4

Predictors for reduction of serum Mac-2 binding protein glucosylation isomer (M2BPGi) level after pemafibrate therapy

FactorUnivariate analysisMultivariate analysis
Odds ratio95% confidence intervalp-valueOdds ratio95% confidence intervalp-value
Age, > 50 years2.9941.140-7.8650.0265.6401.381-23.0400.015
Male, gender0.7080.260-1.9240.498
Body mass index, < 254.6931.181-18.6490.0285.4741.097-27.3310.038
Statin use1.1790.461-3.0140.731
AST, > 50 U/l0.7450.278-1.9970.557
ALT, > 60 U/l0.5830.224-1.5180.2690.4910.140-1.7310.268
γ-GTP, > 70 U/l1.6330.590-4.5210.3453.3950.837-13.7620.086
Platelet count, > 270 × 103/µl2.1820.829-5.7410.1143.2900.953-11.3550.059
Estimated GFR, > 70 ml/min/1.73 m20.8200.321-2.1570.687
Uric acid, > 6 mg/dl0.6020.235-1.5380.288
LDL cholesterol, > 100 mg/dl0.8420.332-2.1390.717
HDL cholesterol, > 50 mg/dl1.3540.524-3.5000.531
Triglyceride, > 120 mg/dl0.4470.167-1.1950.108
Serum albumin, > 4.4 g/dl0.7560.297-1.9220.556
Hemoglobin A1c, 5.6%1.6630.650-4.2510.288
M2BPGi, > 0.72.0630.754-5.6470.1580.8620.221-3.3620.831

[i] ARB – angiotensin II receptor blocker, AST – aspartate aminotransferase, ALT – alanine aminotransferase, γ-GTP – γ-glutamyl transpeptidase, GFR – glomerular filtration rate, LDL – low-density lipoprotein, HDL – high-density lipoprotein

We compared three groups based on BMI (lean BMI < 25, overweight BMI 25-30 and obese BMI > 30) (Fig. 1B). Pemafibrate therapy for six months significantly reduced serum M2BPGi levels in all groups compared to baseline (lean p < 0.001, overweight p = 0.001 and obese p = 0.046). The reduction of serum M2BPGi in the lean NAFLD group was significantly greater than in the obese NAFLD group (p = 0.022), consistent with a greater improvement in hepatic fibrosis.

Discussion

This retrospective observational study shows that pemafibrate therapy for six months improved both markers of hepatic inflammation and fibrosis regardless of BMI. Notably, treatment was more effective in patients with lean NAFLD (BMI < 25) compared with obese NAFLD (BMI > 30). The baseline ALT or M2BPGilevels were not significant predictors of response, and concurrent use of statins did not influence these responses. Age > 50 years was also a positive predictor of improvement of hepatic fibrosis. This is the first report to identify predictors of response to pemafibrate therapy in patients with NAFLD.

Lean NAFLD is more prevalent in Asia (19%) than in the United States (7%) [11]. The prevalence of NAFLD in Japan is 25.5%, and lean NAFLD comprises 20.7% of these patients [12]. Patients with lean NAFLD have a long-term prognosis similar to those with obese NAFLD despite an increased risk of severe liver disease [13]. The presence of lean NAFLD is significantly associated with a higher risk of incident cardiovascular disease compared to lean individuals without NAFLD [14]. Therefore, long-term control of hepatic inflammation and fibrosis in patients with lean NAFLD is important, as it is in those with obese NAFLD. The results of the present study show that patients with lean NAFLD have a greater response to pemafibrate therapy compared to those with obese NAFLD. Pemafibrate concentration and/or body distribution may be better in lean patients than in obese patients. Older patients are also more responsive to pemafibrate therapy. Adherence to pemafibrate may be better in older patients than in younger ones. Genetic predispositions such as variants in patatin-like phospholipase domain containing 3 (PNPLA3), sarcopenia and/or visceral adiposity may affect the response to pemafibrate therapy [12]. Visceral adiposity and insulin resistance have a central role in the development and progression of lean NAFLD compared with obese NAFLD. Pioglitazone, a PPAR-γ agonist, mainly decreases visceral adiposity rather than peripheral adiposity and improves insulin resistance [15]. Actually, a pan-PPAR agonist showed favorable results with histologic improvement in NASH, but an increase in weight was also observed [16]. For lean NAFLD, additional use of pioglitazone may enhance the improvement observed with pemafibrate.

Recent animal studies from Japan reported the effect of pemafibrate on intrahepatic triglyceride levels and lipid droplet formation [17, 18]. Pemafibrate administration alone prevents NASH development without reducing hepatic triglyceride content [18]. Pemafibrate monotherapy reduces the ballooning score and increases lipid droplet numbers [17]. In short, pemafibrate diminishes the size of lipid droplets but does not decrease the total amount of triglycerides in the liver. These results are compatible with a recent phase II trial evaluating the effect of pemafibrate on NAFLD steatosis and fibrosis [19]. The trial did not show a significant difference in steatosis evaluated by magnetic resonance imaging-estimated proton density fat fraction (MRI-PDFF) but there was a significant difference compared to placebo in fibrosis as evaluated by magnetic resonance elastography (MRE). Taken together, these results suggest that pemafibrate significantly increases small lipid droplet numbers by diminishing large droplets. Decreased lipid droplet size and hepatocyte ballooning due to pemafibrate may reduce compression of hepatic sinusoids, leading to improvement of hypoxia and activation of hepatic stellate cells [20]. The persistent improvement of hepatic ballooning and inflammation might result in amelioration of hepatic fibrosis evaluated by MRE without changing the total liver fat content as evaluated by MRI-PDFF in the phase II trial [19]. Therefore, the optimal primary endpoint of a future trial may be the percentage change in MRE-based liver stiffness or liver-biopsy-based fibrosis scoring.

In the present study, obese patients (BMI > 30) were comparatively refractory to pemafibrate therapy. Additional strategies should be considered for these patients. First, double-dose pemafibrate (0.2 mg twice daily) is a viable option for obese patients to elevate the blood concentration. Second, concurrent sodiumglucose cotransporter-2 (SGLT2) inhibitors may be recommended [21]. In the NASH mouse model, combination therapy with pemafibrate and an SGLT2 inhibitor improves both hepatic steatosis and fibrosis, resulting in prolonged survival and suppression of hepatocellular carcinoma development [17]. Therefore, the synergistic effects of pemafibrate and an SGLT2 inhibitor may be useful for obese patients with NAFLD even if the patient does not have diabetes mellitus. A new randomized controlled trial (ClinicalTrials.gov Identifier: NCT05327127) is being undertaken by Kowa Research Institute to evaluate the effects of pemafibrate and/or an SGLT2 inhibitor on NASH.

Decreased levels of AST, ALT, γ-GTP, LDL cholesterol, triglyceride and total bilirubin were observed in this study, and these changes are consistent with the results of a phase II trial investigating the effect of pemafibrate on NAFLD [19]. An elevated platelet count and decreased M2BPGi levels are also consistent with the results of the phase II trial [19]. Therefore, the results of the present study are considered to be reproducible.

The present study has acknowledged limitations. First, this is a retrospective observational study. Second, the combined use of statin, ezetimibe, and an angiotensin II receptor blocker may influence the improvement of NAFLD, but these medications had been started more than six months before starting treatment with pemafibrate [21]. Third, no histological evaluations were performed before or after the treatment. The major advantage of this study is that patients with diabetes mellitus and those who take anti-diabetic medications were excluded. The development and clinical course of NAFLD are largely influenced by concurrent diabetes mellitus, and anti-diabetes medications such as an SGLT2 inhibitor, glucagon-like pepitide-1 and pioglitazone could improve NAFLD [21].

Conclusions

Pemafibrate therapy improves markers of hepatic inflammation and fibrosis regardless of BMI. Patients with lean NAFLD (BMI < 25) have a greater response to pemafibrate therapy than patients with obese NAFLD (BMI > 30). For patients with obese NAFLD, doubledose pemafibrate and/or combined treatment with an SGLT2 inhibitor should be considered. A randomized controlled trial focusing on improvement of hepatic fibrosis is necessary to clarify the effect of pemafibrate on NAFLD/NASH.

Disclosure

The authors S.S. and K.M. have received honoraria from Kowa Pharmaceuticals. The other authors declare no conflict of interest.

References

1 

Younossi Z, Tacke F, Arrese M, et al. Global perspectives on nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology 2019; 69: 2672-2682.

2 

Estes C, Anstee QM, Arias-Loste MT, et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016-2030. J Hepatol 2018; 69: 896-904.

3 

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: 1611-1625.e1612.

4 

Shinozaki S, Tahara T, Lefor AK, et al. Pemafibrate improves hepatic inflammation, function and fibrosis in patients with non-alcoholic fatty liver disease: a one-year observational study. Clin Exp Hepatol 2021; 7: 172-177.

5 

Shinozaki S, Tahara T, Lefor AK, et al. Pemafibrate decreases markers of hepatic inflammation in patients with non-alcoholic fatty liver disease. Clin Exp Hepatol 2020; 6: 270-274.

6 

Seko Y, Yamaguchi K, Umemura A, et al. Effect of pemafibrate on fatty acid levels and liver enzymes in non-alcoholic fatty liver disease patients with dyslipidemia: A single-arm, pilot study. Hepatol Res 2020; 50: 1328-1336.

7 

Hatanaka T, Kosone T, Saito N, et al. Effect of 48-week pemafibrate on non-alcoholic fatty liver disease with hypertriglyceridemia, as evaluated by the FibroScan-aspartate aminotransferase score. JGH Open 2021; 5: 1183-1189.

8 

Ekstedt M, Franzen LE, Mathiesen UL, et al. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology 2006; 44: 865-873.

9 

Seko Y, Sumida Y, Tanaka S, et al. Serum alanine aminotransferase predicts the histological course of non-alcoholic steatohepatitis in Japanese patients. Hepatol Res 2015; 45: E53-61.

10 

Abe M, Miyake T, Kuno A, et al. Association between Wisteria floribunda agglutinin-positive Mac-2 binding protein and the fibrosis stage of non-alcoholic fatty liver disease. J Gastroenterol 2015; 50: 776-784.

11 

VanWagner LB, Armstrong MJ. Lean NAFLD: A not so benign condition? Hepatol Commun 2018; 2: 5-8.

12 

Ito T, Ishigami M, Zou B, et al. The epidemiology of NAFLD and lean NAFLD in Japan: a meta-analysis with individual and forecasting analysis, 1995-2040. Hepatol Int 2021; 15: 366-379.

13 

Hagström H, Nasr P, Ekstedt M, et al. Risk for development of severe liver disease in lean patients with nonalcoholic fatty liver disease: a long-term follow-up study. Hepatol Commun 2018; 2: 48-57.

14 

Yoshitaka H, Hamaguchi M, Kojima T, et al. Nonoverweight nonalcoholic fatty liver disease and incident cardiovascular disease: A post hoc analysis of a cohort study. Medicine (Baltimore) 2017; 96: e6712.

15 

Kuchay MS, Martínez-Montoro JI, Choudhary NS, et al. Non-alcoholic fatty liver disease in lean and non-obese individuals: current and future challenges. Biomedicines 2021; 9: 1346.

16 

Francque SM, Bedossa P, Ratziu V, et al. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. N Engl J Med 2021; 385: 1547-1558.

17 

Murakami K, Sasaki Y, Asahiyama M, et al. Selective PPARα modulator pemafibrate and sodium-glucose cotransporter 2 inhibitor tofogliflozin combination treatment improved histopathology in experimental mice model of non-alcoholic steatohepatitis. Cells 2022; 11: 720.

18 

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: 7818.

19 

Nakajima A, Eguchi Y, Yoneda M, et al. Randomised clinical trial: Pemafibrate, a novel selective peroxisome proliferator-activated receptor α modulator (SPPARMα), versus placebo in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2021; 54: 1263-1277.

20 

Scorletti E, Carr RM. A new perspective on NAFLD: Focusing on lipid droplets. J Hepatol 2022; 76: 934-945.

21 

Tokushige K, Ikejima K, Ono M, et al. Evidence-based clinical practice guidelines for nonalcoholic fatty liver disease/nonalcoholic steatohepatitis 2020. Hepatol Res 2021; 51: 1013-1025.

Copyright: © 2022 Clinical and Experimental Hepatology. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.