Letter to the Editor Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 21, 2024; 30(39): 4308-4312
Published online Oct 21, 2024. doi: 10.3748/wjg.v30.i39.4308
Novel intervention for alcohol-associated liver disease
Fei-Qiong Gao, Department of Endocrinology, The Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310003, Zhejiang Province, China
Jia-Qi Zhu, Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
Xu-Dong Feng, Department of Clinical Laboratory, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
ORCID number: Fei-Qiong Gao (0000-0002-3572-408X); Jia-Qi Zhu (0000-0001-9472-4074); Xu-Dong Feng (0000-0002-4103-7737).
Author contributions: Gao FQ and Feng XD conceived the idea of the manuscript; Gao FQ wrote the draft; Zhu JQ reviewed the literature; Feng XD revised the manuscript; all authors approved the final manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Xu-Dong Feng, MD, PhD, Doctor, Department of Clinical Laboratory, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, No. 57 Xingning Road, Ningbo 315000, Zhejiang Province, China. xdfeng@zju.edu.cn
Received: July 20, 2024
Revised: August 27, 2024
Accepted: September 19, 2024
Published online: October 21, 2024
Processing time: 84 Days and 0.6 Hours

Abstract

A recently published article in the World Journal of Gastroenterology clarified that elafibranor, a dual peroxisome proliferator activated receptor α/δ (PPARα/δ) agonist, reduced inflammation and fibrosis in alcohol-associated liver disease (ALD). This letter aims to discuss the findings presented in that article. ALD is a global health problem, and no effective drugs has been approved by the Food and Drug Administration to cure it. Thus, finding targeted therapies is of great urgency. Herein, we focus on the pathogenesis of ALD and the role of PPARα/δ in its development. Consistent with the conclusion of the article of interest, we think that elafibranor may be a promising therapeutic option for ALD, due to the pivotal involvement of PPARα/δ in the pathogenesis of the disease. However, its treatment dose, timing, and side effects need to be further investigated in future studies.

Key Words: Alcohol-associated liver disease; Elafibranor; Peroxisome proliferator activated receptor α/δ; Therapy; Pathogenesis

Core Tip: Alcohol-associated liver disease (ALD) remains a significant global health challenge. Peroxisome proliferator-activated receptors α and δ (PPARα/δ) play a crucial role in the pathogenesis of ALD. Elafibranor, a dual PPARα/δ activator, shows promise as a potential therapeutic agent for ALD.



TO THE EDITOR

Alcohol-associated liver disease (ALD) represents a global health concern, accounting for about 47% of liver disease-related deaths worldwide[1]. ALD is caused by excessive alcohol consumption. Definitions of heavy drinking are established by the National Institute on Alcohol Abuse and Alcoholism and the Centers for Disease Control and Prevention, while criteria for low-risk drinking are formulated by the World Health Organization. As detailed in Table 1, for women, consumption of alcohol exceeding 56 g per day or 112 g per week is considered heavy drinking, while less than 20 g per day and 140 g per week is categorized as low-risk. For men, heavy drinking is defined as more than 70 g per day or 210 g per week, with low-risk thresholds set at less than 40 g per day and 280 g per week.

Table 1 Definitions of heavy drinking and low-risk drinking.
Women
Men
Alcohol intake
Drinks
Alcohol intake
Drinks
Heavy drinking (NIAAA and CDC)
Per day≥ 56 g≥ 4≥ 70 g≥ 5
Per week≥ 112 g≥ 8≥ 210 g≥ 15
Low-risk drinking (WHO)
Per day< 20 g-< 40 g-
Per week< 140 g-< 280 g-

ALD includes a spectrum of conditions ranging from simple steatosis to steatohepatitis and finally to cirrhosis. In some cases, this progression culminates in hepatocellular carcinoma. Although it has severe public health implications, there are currently no therapies approved by the Food and Drug Administration for ALD treatment[2]. Consequently, it is crucial to investigate and develop therapies targeting the underlying mechanisms of ALD. The study by Koizumi et al[3] offers valuable insights for the development of effective treatment options for ALD[3].

PATHOGENESIS OF ALD

The potential pathogenesis of ALD is diverse[4], as depicted in Figure 1. Hepatocyte death and regeneration are key factors in the progression of ALD[5]. Programmed cell death mechanisms include apoptosis, pyroptosis, necroptosis, autophagy, and ferroptosis[6,7], all of which contribute to the disease process. Oxidative and endoplasmic reticulum stress driven by ethanol metabolism can trigger these cell death pathways in ALD.

Figure 1
Figure 1 Pathogeneses of alcohol-associated liver disease. The primary mechanisms involved in the development of alcohol-associated liver disease are listed. Cell death and regeneration, inflammation, the gut-liver axis, lipid metabolism, and mitochondrial dysfunction all contribute to the development of alcohol-associated liver disease. Created with BioRender.com. TNF-α: Tumor necrosis factor-α; IL: Interleukin; TGF-β: Transforming growth factor-β.

Inflammatory response is crucial to the development of ALD. A complex interplay of immune cells and factors drive the inflammation associated with ALD. Macrophages and neutrophils significantly exacerbate ALD by producing inflammatory mediators and reactive oxygen species[8]. T cells are implicated in a profibrotic capacity[9]. And B cells promote the deposition of antibodies and the activation of the complement system, which contribute to liver damage[10]. Elevated levels of cytokines such as tumor necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, IL-22, and transforming growth factor-β have been observed in ALD patients, exerting both protective and damaging effects on the disease[11]. Chemokines like CXCL8 and CCL20 play a critical role in attracting inflammatory cells to the liver, thus amplifying hepatic inflammation and fibrosis[12].

The gut-liver axis also plays a role in ALD. Chronic alcohol consumption increases gut permeability by downregulating junction proteins[13]. And then microbes translocate into the bloodstream. Alcohol also affects intestinal flora imbalance. It leads to a decrease in microbial diversity, an increase in pathogenic bacteria such as Candida, and a reduction in beneficial bacteria. These would affect the production of virulence factors such as cytolysin, further affecting the progression of ALD[14].

Long-term drinking can seriously damage adipose tissue function, resulting in metabolic dysregulation, thereby promoting the development of ALD[15]. Alcohol stimulates lipolysis in adipose tissue, leading to an increase in circulating free fatty acids. These acids play a crucial role in the development of hepatic steatosis and further trigger inflammatory pathways[16]. Moreover, long-term drinking elevates the secretion of adipose-derived hormones such as visfatin and leptin, which enhance fibrotic and inflammatory processes within the liver[15].

Other potential pathogeneses, such as mitochondrial functionality, also play a role. High-risk drinking negatively affects mitochondrial regeneration, and escalates oxidative stress, ultimately resulting in cell death[17].

INVOLVEMENT OF PEROXISOME PROLIFERATOR-ACTIVATED RECEPTORS α AND δ IN ALD PATHOGENESIS

Peroxisome proliferator-activated receptor α (PPARα), a ligand-activated transcription factor, is widely present in organs such as the liver, heart, and adipose tissue[18]. It can regulate glucolipid metabolism, inflammation responses, and cell death. PPARα modulates β-oxidation, lipid transport, as well as bile acid metabolism[19]. These metabolism activities are vital to ALD development. Loss of PPARα is linked to severe hepatic steatosis[20]. Regarding inflammation, another critical pathogenesis of ALD, PPARα mainly acts through trans-inhibition, that is, antagonizing regulatory factors such as AP-1 and STAT, thereby inhibiting the expression of pro-inflammatory genes[21]. Moreover, PPARα has been reported to induce autophagy[22], which also influences ALD progression. In addition, PPARα activation can reduce the levels of 4-hydroxynonenal, a lipid peroxide that contributes to ALD by inhibiting the activation of NF-κB[23].

Similar to PPARα, PPARδ is expressed and functions in multiple tissues. In the liver, it mitigates liver fibrosis by interfering the transformation of profibrogenic myofibroblasts[24]. Notably, PPARδ expression is found to be lower in patients with severe hepatic steatosis[25], suggesting its potential role in protecting against steatosis. And intestinal PPARδ can enhance mucosal defense capabilities and help prevent dysbiosis[26].

Elafibranor, bezafibrate, and pemafibrate are all PPAR agonists[27]. Both bezafibrate and pemafibrate are PPARα agonists used to treat hypertriglyceridemia. However, pemafibrate is a more selective PPARα agonist, offering greater specificity and fewer side effects compared to bezafibrate. Elafibranor, a dual agonist of PPARα/δ, exhibits superior efficacy in regulating glucose and lipid metabolism, as well as in reducing inflammation and fibrosis. Consequently, elafibranor shows greater potential in the treatment of alcoholic fatty liver disease. Although still an experimental drug[28], elafibranor’s promising effects justify further investigation.

ALCOHOL CESSATION THERAPIES

It is well established that abstaining from alcohol can significantly benefit patients with ALD. However, these patients often struggle to quit drinking on their own. Alcohol cessation medications, such as disulfiram, naltrexone, and acamprosate, can be helpful in supporting their efforts. These medications, however, are primarily approved for use in alcohol use disorder (AUD)[29], and ALD patients do not always meet the criteria for AUD. Additionally, these medications can cause adverse effects like liver and kidney function damage. And unlike the PPAR activator elafibranor, these medications do not have the capacity to reverse the inflammation and fibrosis that have already developed in the liver.

FUTURE OF ELAFIBRANOR

Elafibranor is a dual agonist of PPARα and PPARδ. It has been found to have therapeutic effects on primary biliary cholangitis and non-alcoholic fatty liver disease[30,31], which share some pathogenic mechanisms with ALD. However, its efficacy in treating ALD has not yet been clarified. Given the beneficial roles of both PPARα and PPARδ in ALD, and the ability of elafibranor to activate these receptors, it is conceivable that elafibranor could significantly ameliorate ALD. Thus, investigating elafibranor’s impact on ALD presents an interesting area of research. The study conducted by Koizumi et al[3] has provided evidence that elafibranor effectively reduces liver steatosis, inflammation, and fibrosis in ALD.

CONCLUSION

ALD is a common disease with a poor prognosis. Finding targeted therapies for the disease can improve patient outcomes and their life quality. According to the research conducted by Koizumi et al[3], elafibranor represents a promising therapeutic candidate. However, further investigation into elafibranor’s application in ALD treatment is needed. This includes determining optimal therapeutic dosages and evaluating potential side effects specific to ALD.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade C

Novelty: Grade C

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Kanda T S-Editor: Li L L-Editor: Wang TQ P-Editor: Zheng XM

References
1.  Lee BP, Witkiewitz K, Mellinger J, Anania FA, Bataller R, Cotter TG, Curtis B, Dasarathy S, DeMartini KS, Diamond I, Diazgranados N, DiMartini AF, Falk DE, Fernandez AC, German MN, Kamath PS, Kidwell KM, Leggio L, Litten R, Louvet A, Lucey MR, McCaul ME, Sanyal AJ, Singal AK, Sussman NL, Terrault NA, Thursz MR, Verna EC, Radaeva S, Nagy LE, Mitchell MC. Designing clinical trials to address alcohol use and alcohol-associated liver disease: an expert panel Consensus Statement. Nat Rev Gastroenterol Hepatol. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
2.  Bataller R, Arab JP, Shah VH. Alcohol-Associated Hepatitis. N Engl J Med. 2022;387:2436-2448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 89]  [Article Influence: 44.5]  [Reference Citation Analysis (0)]
3.  Koizumi A, Kaji K, Nishimura N, Asada S, Matsuda T, Tanaka M, Yorioka N, Tsuji Y, Kitagawa K, Sato S, Namisaki T, Akahane T, Yoshiji H. Effects of elafibranor on liver fibrosis and gut barrier function in a mouse model of alcohol-associated liver disease. World J Gastroenterol. 2024;30:3428-3446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
4.  Mackowiak B, Fu Y, Maccioni L, Gao B. Alcohol-associated liver disease. J Clin Invest. 2024;134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Nagy LE, Ding WX, Cresci G, Saikia P, Shah VH. Linking Pathogenic Mechanisms of Alcoholic Liver Disease With Clinical Phenotypes. Gastroenterology. 2016;150:1756-1768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 132]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
6.  Zheng M, Kanneganti TD. The regulation of the ZBP1-NLRP3 inflammasome and its implications in pyroptosis, apoptosis, and necroptosis (PANoptosis). Immunol Rev. 2020;297:26-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 237]  [Article Influence: 59.3]  [Reference Citation Analysis (0)]
7.  Stolz A, Ernst A, Dikic I. Cargo recognition and trafficking in selective autophagy. Nat Cell Biol. 2014;16:495-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 816]  [Cited by in F6Publishing: 877]  [Article Influence: 87.7]  [Reference Citation Analysis (0)]
8.  Gao B, Ahmad MF, Nagy LE, Tsukamoto H. Inflammatory pathways in alcoholic steatohepatitis. J Hepatol. 2019;70:249-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 225]  [Article Influence: 45.0]  [Reference Citation Analysis (0)]
9.  Mehal W. Mechanisms of liver fibrosis in metabolic syndrome. eGastroenterology. 2023;1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
10.  Ahmadi AR, Song G, Gao T, Ma J, Han X, Hu M, Cameron AM, Wesson RN, Philosophe B, Ottmann S, King E, Gurakar A, Qi L, Peiffer B, Burdick J, Anders R, Zhou Z, Lu H, Feng D, Chen C, Qian J, Gao B, Zhu H, Sun Z. Discovery and characterization of cross-reactive intrahepatic antibodies in severe alcoholic hepatitis. eLife. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Wang H, Mehal W, Nagy LE, Rotman Y. Immunological mechanisms and therapeutic targets of fatty liver diseases. Cell Mol Immunol. 2021;18:73-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
12.  Gao B, Xu M. Chemokines and alcoholic hepatitis: are chemokines good therapeutic targets? Gut. 2014;63:1683-1684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
13.  Rao R. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology. 2009;50:638-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 351]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
14.  Llopis M, Cassard AM, Wrzosek L, Boschat L, Bruneau A, Ferrere G, Puchois V, Martin JC, Lepage P, Le Roy T, Lefèvre L, Langelier B, Cailleux F, González-Castro AM, Rabot S, Gaudin F, Agostini H, Prévot S, Berrebi D, Ciocan D, Jousse C, Naveau S, Gérard P, Perlemuter G. Intestinal microbiota contributes to individual susceptibility to alcoholic liver disease. Gut. 2016;65:830-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 374]  [Article Influence: 46.8]  [Reference Citation Analysis (0)]
15.  Parker R, Kim SJ, Gao B. Alcohol, adipose tissue and liver disease: mechanistic links and clinical considerations. Nat Rev Gastroenterol Hepatol. 2018;15:50-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 109]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
16.  Wei X, Shi X, Zhong W, Zhao Y, Tang Y, Sun W, Yin X, Bogdanov B, Kim S, McClain C, Zhou Z, Zhang X. Chronic alcohol exposure disturbs lipid homeostasis at the adipose tissue-liver axis in mice: analysis of triacylglycerols using high-resolution mass spectrometry in combination with in vivo metabolite deuterium labeling. PLoS One. 2013;8:e55382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 62]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
17.  Nassir F, Ibdah JA. Role of mitochondria in alcoholic liver disease. World J Gastroenterol. 2014;20:2136-2142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 89]  [Cited by in F6Publishing: 91]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
18.  Brown JD, Plutzky J. Peroxisome proliferator-activated receptors as transcriptional nodal points and therapeutic targets. Circulation. 2007;115:518-533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 258]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
19.  Rotman Y, Sanyal AJ. Current and upcoming pharmacotherapy for non-alcoholic fatty liver disease. Gut. 2017;66:180-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 268]  [Cited by in F6Publishing: 320]  [Article Influence: 45.7]  [Reference Citation Analysis (0)]
20.  Montagner A, Polizzi A, Fouché E, Ducheix S, Lippi Y, Lasserre F, Barquissau V, Régnier M, Lukowicz C, Benhamed F, Iroz A, Bertrand-Michel J, Al Saati T, Cano P, Mselli-Lakhal L, Mithieux G, Rajas F, Lagarrigue S, Pineau T, Loiseau N, Postic C, Langin D, Wahli W, Guillou H. Liver PPARα is crucial for whole-body fatty acid homeostasis and is protective against NAFLD. Gut. 2016;65:1202-1214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 491]  [Article Influence: 61.4]  [Reference Citation Analysis (0)]
21.  Lefebvre P, Chinetti G, Fruchart JC, Staels B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J Clin Invest. 2006;116:571-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 654]  [Cited by in F6Publishing: 705]  [Article Influence: 39.2]  [Reference Citation Analysis (0)]
22.  Luo R, Su LY, Li G, Yang J, Liu Q, Yang LX, Zhang DF, Zhou H, Xu M, Fan Y, Li J, Yao YG. Activation of PPARA-mediated autophagy reduces Alzheimer disease-like pathology and cognitive decline in a murine model. Autophagy. 2020;16:52-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 202]  [Article Influence: 50.5]  [Reference Citation Analysis (0)]
23.  Ding L, Wo L, Du Z, Tang L, Song Z, Dou X. Danshen protects against early-stage alcoholic liver disease in mice via inducing PPARα activation and subsequent 4-HNE degradation. PLoS One. 2017;12:e0186357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
24.  Li X, Chen Y, Wu S, He J, Lou L, Ye W, Wang J. microRNA-34a and microRNA-34c promote the activation of human hepatic stellate cells by targeting peroxisome proliferator-activated receptor γ. Mol Med Rep. 2015;11:1017-1024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
25.  Zarei M, Barroso E, Palomer X, Dai J, Rada P, Quesada-López T, Escolà-Gil JC, Cedó L, Zali MR, Molaei M, Dabiri R, Vázquez S, Pujol E, Valverde ÁM, Villarroya F, Liu Y, Wahli W, Vázquez-Carrera M. Hepatic regulation of VLDL receptor by PPARβ/δ and FGF21 modulates non-alcoholic fatty liver disease. Mol Metab. 2018;8:117-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 73]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
26.  Tomas J, Mulet C, Saffarian A, Cavin JB, Ducroc R, Regnault B, Kun Tan C, Duszka K, Burcelin R, Wahli W, Sansonetti PJ, Pédron T. High-fat diet modifies the PPAR-γ pathway leading to disruption of microbial and physiological ecosystem in murine small intestine. Proc Natl Acad Sci USA. 2016;113:E5934-E5943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 163]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
27.  Kamata S, Honda A, Kashiwagi N, Shimamura A, Yashiro S, Komori Y, Hosoda A, Akahoshi N, Ishii I. Different Coactivator Recruitment to Human PPARα/δ/γ Ligand-Binding Domains by Eight PPAR Agonists to Treat Nonalcoholic Fatty Liver Disease. Biomedicines. 2024;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
28.  Boeckmans J, Natale A, Rombaut M, Buyl K, Cami B, De Boe V, Heymans A, Rogiers V, De Kock J, Vanhaecke T, Rodrigues RM. Human hepatic in vitro models reveal distinct anti-NASH potencies of PPAR agonists. Cell Biol Toxicol. 2021;37:293-311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
29.  Kranzler HR, Soyka M. Diagnosis and Pharmacotherapy of Alcohol Use Disorder: A Review. JAMA. 2018;320:815-824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 352]  [Article Influence: 58.7]  [Reference Citation Analysis (0)]
30.  Ratziu V, Harrison SA, Francque S, Bedossa P, Lehert P, Serfaty L, Romero-Gomez M, Boursier J, Abdelmalek M, Caldwell S, Drenth J, Anstee QM, Hum D, Hanf R, Roudot A, Megnien S, Staels B, Sanyal A; GOLDEN-505 Investigator Study Group. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-α and -δ, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology. 2016;150:1147-1159.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 677]  [Cited by in F6Publishing: 739]  [Article Influence: 92.4]  [Reference Citation Analysis (0)]
31.  Kowdley KV, Bowlus CL, Levy C, Akarca US, Alvares-da-Silva MR, Andreone P, Arrese M, Corpechot C, Francque SM, Heneghan MA, Invernizzi P, Jones D, Kruger FC, Lawitz E, Mayo MJ, Shiffman ML, Swain MG, Valera JM, Vargas V, Vierling JM, Villamil A, Addy C, Dietrich J, Germain JM, Mazain S, Rafailovic D, Taddé B, Miller B, Shu J, Zein CO, Schattenberg JM; ELATIVE Study Investigators’ Group;  ELATIVE Study Investigators' Group. Efficacy and Safety of Elafibranor in Primary Biliary Cholangitis. N Engl J Med. 2024;390:795-805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]