Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 14, 2024; 30(30): 3541-3547
Published online Aug 14, 2024. doi: 10.3748/wjg.v30.i30.3541
Glucagon-like peptide 1 agonists are potentially useful drugs for treating metabolic dysfunction-associated steatotic liver disease
Maurizio Soresi, Lydia Giannitrapani, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo 90127, Italy
ORCID number: Maurizio Soresi (0000-0001-7850-555X); Lydia Giannitrapani (0000-0003-2845-5296).
Author contributions: Soresi M and Giannitrapani L contributed to this paper; Soresi M designed the overall manuscript concept and outline; Giannitrapani L contributed to manuscript discussion and design; Soresi M and Giannitrapani L contributed to the writing, and editing of the manuscript, illustrations, literature review, and read and approved the final manuscript.
Conflict-of-interest statement: The authors declare that they have no conflicts of interest.
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: Maurizio Soresi, MD, Associate Professor, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Via del Vespro 141, Palermo 90127, Italy. maurizio.soresi@unipa.it
Received: January 26, 2024
Revised: June 26, 2024
Accepted: July 18, 2024
Published online: August 14, 2024
Processing time: 195 Days and 16.1 Hours

Abstract

In this editorial, we comment on Yin et al’s recently published Letter to the editor. In particular, we focus on the potential use of glucagon-like peptide 1 receptor agonists (GLP-1RAs) alone, but even more so in combination therapy, as one of the most promising therapies in metabolic dysfunction-associated steatotic liver disease (MASLD), the new definition of an old condition, non-alcoholic fatty liver disease, which aims to better define the spectrum of steatotic pathology. It is well known that GLP-1RAs, having shown outstanding performance in fat loss, weight loss, and improvement of insulin resistance, could play a role in protecting the liver from progressive damage. Several clinical trials have shown that, among GLP-1RAs, semaglutide is a safe, well-studied therapeutic choice for MASLD patients; however, most studies demonstrate that, while semaglutide can reduce steatosis, including steatohepatitis histological signs (in terms of inflammatory cell infiltration and hepatocyte ballooning), it does not improve fibrosis. Combinations of therapies with different but complementary mechanisms of action are considered the best way to improve efficiency and slow disease progression due to the complex pathophysiology of the disease. In particular, GLP-1RAs associated with antifibrotic drug therapy, dual glucose-dependent insulinotropic polypeptide (GIP)/GLP-1RA or GLP-1 and glucagon RAs have promoted greater improvement in hepatic steatosis, liver biochemistry, and non-invasive fibrosis tests than monotherapy. Therefore, although to date there are no definitive indications from international drug agencies, there is the hope that soon the therapeutic lines in the most advanced phase of study will be able to provide a therapy for MASLD, one that will certainly include the use of GLP-1RAs as combination therapy.

Key Words: Non-alcoholic fatty liver disease, Glucagon-like peptide 1, Semaglutide, Liver fibrosis, Therapy

Core Tip: In this editorial, we comment on Yin et al’s recently published Letter to the editor. Despite the widespread diffusion, up to now there have been no drugs capable of reliably blocking the evolution of non-alcoholic steato-hepatitis towards advanced stages of fibrosis. We agree with Yin et al that glucagon-like peptide receptor agonists monotherapy does not perform well as an antifibrotic therapy. The use of combination therapy according to disease stage and co-morbidities, will also be a challenge in the near future.



INTRODUCTION

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease[1], characterized by an accumulation of fat in the liver greater than 5%.

The highest NAFLD prevalence has been described in Latin America (44.37%), followed by the Middle East and North Africa (36.53%), South Asia (33.83%), South-East Asia (33.07%), North America (31.20%), East Asia (29.71%), Asia Pacific (28.02%) and Western Europe (25.10%)[1].

It can be considered as an “umbrella” term under which various histological and clinical liver conditions are grouped: From simple liver steatosis (NAFL) to non-alcoholic steato-hepatitis (NASH), which in addition to steatosis shows lobular inflammation and hepatocyte ballooning, with or without fibrosis; the latter, if present, can be perisinusoidal or extended and become pan lobular[2].

NASH, in turn, can regress towards steatosis alone or evolve towards liver cirrhosis, which can be complicated with hepatocellular carcinoma and portal hypertension of varying degrees[3].

Recently, a multi society Delphi consensus published a statement on the new fatty liver disease nomenclature, changing the definition of NAFLD into steatotic liver disease (SLD), an overarching term to encompass the various etiologies of steatosis[4], which in turn includes metabolic dysfunction-associated SLD (MASLD), a condition in which steatosis is associated with metabolic dysfunctions typical of metabolic syndrome (MetS), type 2 diabetes mellitus (T2DM), arterial hypertension, visceral obesity, hypertriglyceridemia, low high-density lipoprotein levels, all of which are conditions related to insulin resistance.

MASLD associated with alcohol intake (MetALD) of varying degrees, is between 20 g and 50 g/day in women and 30-60 g/day in men.

Alcohol related liver disease (ALD) is associated with > 50 g/day in women and > 60 g/day in men.

SPECIFIC ETIOLOGY OF SLD

Drug-induced liver injury includes Monogenic diseases: Lysosomal acid lipase deficiency, Wilson disease, hypobetalipoproteinemia, and inborn errors of metabolism; Miscellaneous: Celiac disease, human immunodeficiency virus, malnutrition, and hepatitis C virus (HCV).

Those with no identifiable cause (cryptogenic SLD), may be re-categorized in the future pending developments in our understanding of disease pathophysiology. There are also particular situations, such as MASLD associated with autoimmune hepatitis or HCV or HBV infections, defined as MASLD + (Tables 1 and 2).

Table 1 Steatotic liver disease, new classification.
Subcategories
Etiologies
MASLDPresence of hepatic steatosis and 1 cardiometabolic risk factor out of 5 (see Table 2) and no other discernible cause for hepatic steatosis. If additional drivers of steatosis are identified, then this is consistent with a combination etiology
MetALDMASLD and increased alcohol intake: 20-50 g/day (females); 30-60 g/day (males)
Alcohol Associated (Alcohol related liver disease-ALD)Alcohol intake > 50 g/day (females) and > 60 g/day (males)
Specific etiology SLD(1) Drug-induced liver injury; (2) Monogenic diseases: Lysosomal acid lipase deficiency, Wilson disease, hypo-betalipoproteinemia, inborn errors of metabolism; and (3) Miscellaneous: Celiac disease, HIV, malnutrition, HCV
Cryptogenic SLDThose with no identifiable cause (cryptogenic SLD) may be re-categorized in the future pending developments in our understanding of disease pathophysiology
Table 2 Cardio-metabolic risk factors.
Risk factors
Cut-off values
BMIBMI > 25 kg/m2 (23 in Asia) or waist circumference > 94 cm (males)/ > 80 cm (females) or ethnicity-adjusted
Fasting serum glucoseFasting serum glucose ≥ 5.6 mmol/L (100 mg/dL) or 2-h post-load glucose levels ≥ 7.8 mmol/L (≥ 140 mg/dL) or HbA1c ≥ 5.7% (39 mmol/L) or T2DM or treatment for T2DM
Blood pressureBlood pressure 130/85 mmHg or specific antihypertensive drug treatment
Plasma triglyceridesPlasma triglycerides ≥ 1.70 mmol/L (≥ 150 mg/dL) or lipid lowering treatment
Plasma HDL cholesterolPlasma HDL cholesterol ≤ 1.0 mmol/L (≤ 40 mg/dL) (males) and ≤ 1.3 mmol/L (≤ 50 mg/dL) (females) or lipid lowering treatment

This new classification better defines the spectrum of steatotic pathology, also allowing a better prognostic evaluation; indeed, a MASLD patient has a lower risk of mortality from all causes [hazard ratio (HR) = 1.234, 95%CI: 1.12-1.39] compared to MetALD (HR 1.69, 95%CI: 1.21-2.39) and ALD patients with MASLD (HR = 1.99, 95%CI: 1.27-3.12)[5,6]. The natural history of SLD is variable and depends on non-modifiable (age, sex, race/ethnicity, family history, genetics) and modifiable (lifestyle/diet/exercise, comorbidities, drugs, alcohol) risk factors[7,8].

Despite the widespread diffusion of SLD, up to now there have been no drugs capable of reliably blocking the evolution of NASH towards advanced stages of fibrosis and its consequent complications. The complex pathophysiological mechanisms that often interact with each other probably make it difficult to find an effective therapy, and even lifestyle and diet modifications, although important, are not effective on their own.

THERAPY

The severity of the clinical pictures that the evolution of NASH can produce, along with the complexity of its pathogenesis, have led to the identification of a series of therapeutic targets, from those used to treat diseases that contribute to its development and progression (i.e., antidiabetic agents) to drugs targeting liver inflammation and fibrosis.

The first to be studied were metabolic targets: Insulin resistance, adipose tissue dysfunction, lipid flux in the liver, and de novo lipogenesis.

Historical studies were conducted on the effects of the PPAR γ agonist pioglitazone on the NAFLD activity score (NAS) or other histological components of NASH improvements[9,10]. Then, other PPAR agonists (PPAR α agonist, pemafibrate, dual PPAR γ and PPAR α, saroglitazar, dual PPAR α and PPAR δ, elafibranor and pan-PPAR agonist, lanifibranor) were tested, with wide-ranging results from no improvement at all to improvements in NASH histology[11].

Certain other drugs lead to improvements in NASH as they improve insulin resistance and cause weight loss[12,13], while others have a direct effect on the liver[14,15].

Categories of drugs that aim to control the mechanisms of cell death and inflammation resulting from lipotoxicity, such as chemokine antagonists, anti-apoptotics or VAP1 inhibitors (NCT04897594), are currently under study or have been discontinued, such as c-Jun N-terminal kinase inhibitors (NCT04048876)[16].

Another class of drugs are antifibrotics which act directly on the fibrogenic process. Unfortunately, these drugs (i.e., simtuzumab, belapectin, emricasan) have not produced good results in clinical trials. This may be due to two reasons: Firstly, because they have been used on patients with very advanced stage fibrosis and cirrhosis, and, secondly, because antifibrotic action alone, without blocking the inflammatory condition present in NASH, may be insufficient[16].

FXR agonists, and in particular obeticholic acid (OCA), a semi-synthetic chenodeoxycholic acid analogue, have been tested in several clinical trials, with the most important being a multicenter, randomized, placebo-controlled phase 3 trial whose interim analysis showed that 25 mg OCA significantly improved fibrosis and key components of NASH disease activity among patients with NASH[17]. However, in a very recent 48-week trial of a highly potent non-bile acid FXR agonist, Tropifexor, no improvement in fibrosis or resolution of NASH relative to placebo was observed[18].

The involvement of thyroid hormone receptor beta in the regulation of lipid metabolism and insulin sensitivity has led to the use of thyroid hormone receptor beta agonists in trials in NAFLD patients. Resmetirom is the most frequently used, with promising results, and a double-blind placebo controlled randomized phase 3 study to evaluate whether Resmetirom resolves NASH and/or reduces fibrosis on liver biopsy and prevents progression to cirrhosis and/or advanced liver disease in patients with NASH and fibrosis (MAESTRO-NASH) is still ongoing.

Finally, among the newer antidiabetic drugs that work on the pathophysiological mechanisms shared by T2DM and MASLD, sodium-glucose cotransporter (SGLT2) inhibitors and glucagon-like peptide 1 receptor agonists (GLP-1RAs) have long been studied. In particular, SGLT2 inhibitors, developed for the management of T2DM due to their beneficial effects on glycemic control, have now entered the guidelines for the treatment of heart failure thanks to their action on sodium homeostasis and water retention[19]. As a consequence of their role in the treatment of MetS, to date several studies have been conducted to evaluate the efficacy of SGLT2 inhibitors in the treatment of NAFLD, with significant heterogeneity of end points and results, but most of them have been proven to be efficient in reducing liver fat content (LFC), aspartate aminotransferase/alanine aminotransferase levels, and even liver stiffness[20].

GLP-1 is an incretin synthesised by the intestine that induces the release of insulin from pancreatic beta cells in response to blood glucose levels, suppresses glucagon production, delays gastric emptying and decreases appetite, thus contributing to glycemic homeostasis[21]. GLP-1RAs have recently become one of the cornerstones of T2DM and obesity treatment.

As a consequence, considering the hypothetical role of antidiabetic drugs with weight loss against steatosis, several studies have been conducted on the use of GLP-1RAs in NASH patients. In particular, a randomized controlled trial (the LEAN trial) in 52 patients with NASH found that liraglutide reduced steatosis and hepatocyte ballooning[22].

Moreover, in a phase 2 study, semaglutide at a dosage of 0.4 mg was shown to reduce NASH when compared to placebo (59% vs 17%, P < 0.001), but had no effect on fibrosis[23]. A randomized, placebo-controlled trial that included 71 patients with compensated cirrhosis related to NASH, showed that high-dose (2.4 mg) semaglutide is safe; however, it did not improve fibrosis or increase NASH resolution rates compared to placebo[23,24]. A meta-analysis of randomized controlled trials found that liraglutide and semaglutide improved MRI-assessed steatosis, the histological signs of NASH, but confirmed no improvement in fibrosis[25].

Furthermore, recent studies have revealed that the use of GLP-1 agonists in cirrhotic diabetic subjects reduces mortality from all causes, from cardiovascular diseases and the probability of decompensated cirrhosis, both when comparing the population taking GLP-1RAs with non-using patients[26] and when compared with patients taking dipeptidylpeptidase-4 inhibitors or sulfonylureas[27].

Starting from these premises, it has been hypothesized that combination therapy with drugs having complementary mechanisms could be more beneficial (Table 3), and clinical trials have been launched to prove this. For example, Cotadutide, a dual GLP-1 and glucagon receptor agonist, has been studied in overweight or obese and T2DM subjects[15], and is now under development for NASH (NCT04019561).

Table 3 Clinical trials with glucagon-like peptide 1 receptor agonists dual therapy.
Molecule and trial
Primary aim
Type of study
Duration of therapy
State of recruitment
Main results
Cotadutide (GLP-1/glucagon receptor agonist)[28]To evaluate the effects of cotadutide on hepatic and metabolic parameters in participants with overweight/obesity and type 2 diabetesRandomized, phase 2b study54 weeksCompletedImproved glycemic control and weight loss, improvements in hepatic parameters
Cotadutide (GLP-1/glucagon receptor agonist), NCT04019561To evaluate the safety (including hepatic safety), tolerability and pharmacodynamic effects of two dosage levels of cotadutide in obese subjects with non-alcoholic fatty liver disease/non-alcoholic steatohepatitisRandomized, double-blind, placebo-controlled, phase 2 studyCompleted
Tirzepatide (GIP/ GLP-1 receptor agonist)[30]To determine the effect of tirzepatide on biomarkers of non-alcoholic steatohepatitis and fibrosis in patients with type 2 diabetesPost hoc analysis in a phase 2 trial26 weeksCompletedHigher tirzepatide doses significantly decreased non-alcoholic steatohepatitis-related biomarkers and increased adiponectin in patients with type 2 diabetes
Tirzepatide (GIP/GLP-1 receptor agonist)[31]To characterize the changes in liver fat content, volume of visceral adipose tissue, and abdominal subcutaneous adipose tissue in response to tirzepatide or insulin degludec in a subpopulation of the SURPASS-3 studyRandomized, open-label, parallel-group, phase 3 study52 weeksCompletedSignificant reduction in liver fat content and visceral adipose tissue and abdominal subcutaneous adipose tissue volumes compared with insulin degludec in this subpopulation of patients with type 2 diabetes in the SURPASS-3 study
Tirzepatide, (GIP/GLP-1 receptor agonist), NCT04166773 (SYNERGY-NASH)To determine whether tirzepatide, administered once weekly, is safe and effective as a treatment for non-alcoholic steatohepatitisRandomized, double-blind, placebo-controlled phase 2 study52 weeksCompleted
Efinopegdutide (GLP-1/glucagon receptor co-agonist)[32]To assess the effects of the GLP-1/glucagon receptor co-agonist
efinopegdutide relative to the selective GLP-1 receptor agonist semaglutide on liver fat content in patients with non-alcoholic fatty liver disease
Randomized, phase 2a, active-comparator-controlled, parallel-group, open-label study24 weeksCompletedIn patients with non-alcoholic fatty liver disease, treatment with efinopegdutide 10 mg weekly led to a significantly greater reduction in liver fat content than semaglutide 1 mg weekly

Tirzepatide, a novel dual incretin receptor agonist (twincretin) acting on both GLP-1 and GIP receptors, has demonstrated superior efficacy on HbA1c and body weight improvements in T2DM than incretins alone[28]. Tirzepatide has also been studied in a post hoc analysis in a phase 2 trial, proving its positive effect on biomarkers of NASH and liver fibrosis in T2DM individuals[29]. In a sub-study of the SURPASS-3 trial (SURPASS-3 magnetic resonance imaging study), tirzepatide promoted a significant reduction in LFC when compared with insulin degludec[30], and it is being used in the ongoing trial, “A study of Tirzepatide (LY3298176) in participants with non-alcoholic steatohepatitis (NASH) (SYNERGY-NASH)”, NCT04166773, which has been recently completed.

Finally, a very recent paper evaluated the efficacy and safety of efinopegdutide (a GLP-1/glucagon receptor co-agonist) in patients with NAFLD, demonstrating in a phase IIa randomized active-comparator-controlled parallel-group open-label study, the superiority of treatment with efinopegdutide in terms of reducing magnetic resonance imaging-estimated LFC than semaglutide in patients with NAFLD[31].

CONCLUSION

We agree with Yin et al[32] that GLP-RA monotherapy does not perform well as an antifibrotic therapy, but we also have to consider its proven efficacy in reducing LFC and controlling the risk factors of MetS. It is undeniable that combination therapy looks like the most promising strategy, with ongoing clinical trials that will give us answers regarding the possible indications in MASLD, at least in its early phases. In any case, tailoring the use of combination therapy to the individual patient, according to disease stage and co-morbidities, will also be a challenge in the near future and further exploration of the mechanisms of action of these drugs will be crucial to track future research directions.

Footnotes

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

Peer-review model: Single blind

Corresponding Author's Membership in Professional Societies: Società Italiana di Ultrasonologia in Medicina e Biologia; Società Italiana di Medicina Interna; Associazione Italiana per lo Studio del Fegato; Federazione delle Associazioni dei Dirigenti Ospedalieri Internisti.

Specialty type: Gastroenterology and hepatology

Country of origin: Italy

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Feng X S-Editor: Liu H L-Editor: Webster JR P-Editor: Yuan YY

References
1.  Younossi ZM, Golabi P, Paik JM, Henry A, Van Dongen C, Henry L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology. 2023;77:1335-1347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 529]  [Article Influence: 529.0]  [Reference Citation Analysis (1)]
2.  Singh S, Allen AM, Wang Z, Prokop LJ, Murad MH, Loomba R. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: a systematic review and meta-analysis of paired-biopsy studies. Clin Gastroenterol Hepatol. 2015;13:643-54.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 932]  [Cited by in F6Publishing: 1063]  [Article Influence: 118.1]  [Reference Citation Analysis (0)]
3.  Associazione Italiana per lo Studio del Fegato (AISF), Società Italiana di Diabetologia (SID) and Società Italiana dell'Obesità (SIO); Members of the guidelines panel;  Coordinator;  AISF Members;  SID Members;  SIO Members;  Metodologists. Non-alcoholic fatty liver disease in adults 2021: A clinical practice guideline of the Italian Association for the Study of the Liver (AISF), the Italian Society of Diabetology (SID) and the Italian Society of Obesity (SIO). Dig Liver Dis. 2022;54:170-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
4.  Kholodenko BN, Kolch W, Rukhlenko OS. Reversing pathological cell states: the road less travelled can extend the therapeutic horizon. Trends Cell Biol. 2023;33:913-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
5.  Li M, Xie W. Are there all-cause mortality differences between metabolic dysfunction-associated steatotic liver disease subtypes? J Hepatol. 2024;80:e53-e54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
6.  Ciardullo S, Carbone M, Invernizzi P, Perseghin G. Exploring the landscape of steatotic liver disease in the general US population. Liver Int. 2023;43:2425-2433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 44]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
7.  Powell EE, Wong VW, Rinella M. Non-alcoholic fatty liver disease. Lancet. 2021;397:2212-2224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 461]  [Cited by in F6Publishing: 1101]  [Article Influence: 367.0]  [Reference Citation Analysis (33)]
8.  Loomba R, Friedman SL, Shulman GI. Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell. 2021;184:2537-2564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 899]  [Cited by in F6Publishing: 811]  [Article Influence: 270.3]  [Reference Citation Analysis (36)]
9.  Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, Balas B, Gastaldelli A, Tio F, Pulcini J, Berria R, Ma JZ, Dwivedi S, Havranek R, Fincke C, DeFronzo R, Bannayan GA, Schenker S, Cusi K. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med. 2006;355:2297-2307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1307]  [Cited by in F6Publishing: 1256]  [Article Influence: 69.8]  [Reference Citation Analysis (0)]
10.  Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, Van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR; NASH CRN. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 2010;362:1675-1685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2215]  [Cited by in F6Publishing: 2243]  [Article Influence: 160.2]  [Reference Citation Analysis (2)]
11.  Sangro P, de la Torre Aláez M, Sangro B, D'Avola D. Metabolic dysfunction-associated fatty liver disease (MAFLD): an update of the recent advances in pharmacological treatment. J Physiol Biochem. 2023;79:869-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 15]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
12.  Harrison SA, Alkhouri N, Davison BA, Sanyal A, Edwards C, Colca JR, Lee BH, Loomba R, Cusi K, Kolterman O, Cotter G, Dittrich HC. Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled phase IIb study. J Hepatol. 2020;72:613-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 131]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
13.  Capitanio U, Larcher A, Montorsi F. Re: Toni K. Choueiri, Piotr Tomczak, Se Hoon Park, et al. Adjuvant Pembrolizumab after Nephrectomy in Renal-Cell Carcinoma. N Engl J Med 2021;385:683-94: Adjuvant Pembrolizumab After Nephrectomy: A Plea to Reconsider the Need for Lymph Node Dissection. Eur Urol. 2022;81:e28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
14.  Cusi K, Orsak B, Bril F, Lomonaco R, Hecht J, Ortiz-Lopez C, Tio F, Hardies J, Darland C, Musi N, Webb A, Portillo-Sanchez P. Long-Term Pioglitazone Treatment for Patients With Nonalcoholic Steatohepatitis and Prediabetes or Type 2 Diabetes Mellitus: A Randomized Trial. Ann Intern Med. 2016;165:305-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 592]  [Cited by in F6Publishing: 638]  [Article Influence: 79.8]  [Reference Citation Analysis (0)]
15.  Nahra R, Wang T, Gadde KM, Oscarsson J, Stumvoll M, Jermutus L, Hirshberg B, Ambery P. Effects of Cotadutide on Metabolic and Hepatic Parameters in Adults With Overweight or Obesity and Type 2 Diabetes: A 54-Week Randomized Phase 2b Study. Diabetes Care. 2021;44:1433-1442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 151]  [Article Influence: 50.3]  [Reference Citation Analysis (0)]
16.  Ratziu V, Francque S, Sanyal A. Breakthroughs in therapies for NASH and remaining challenges. J Hepatol. 2022;76:1263-1278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 70]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
17.  Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, Bedossa P, Geier A, Beckebaum S, Newsome PN, Sheridan D, Sheikh MY, Trotter J, Knapple W, Lawitz E, Abdelmalek MF, Kowdley KV, Montano-Loza AJ, Boursier J, Mathurin P, Bugianesi E, Mazzella G, Olveira A, Cortez-Pinto H, Graupera I, Orr D, Gluud LL, Dufour JF, Shapiro D, Campagna J, Zaru L, MacConell L, Shringarpure R, Harrison S, Sanyal AJ; REGENERATE Study Investigators. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019;394:2184-2196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 610]  [Cited by in F6Publishing: 760]  [Article Influence: 152.0]  [Reference Citation Analysis (0)]
18.  Sanyal AJ, Lopez P, Lawitz EJ, Lucas KJ, Loeffler J, Kim W, Goh GBB, Huang JF, Serra C, Andreone P, Chen YC, Hsia SH, Ratziu V, Aizenberg D, Tobita H, Sheikh AM, Vierling JM, Kim YJ, Hyogo H, Tai D, Goodman Z, Schaefer F, Carbarns IRI, Lamle S, Martic M, Naoumov NV, Brass CA. Tropifexor for nonalcoholic steatohepatitis: an adaptive, randomized, placebo-controlled phase 2a/b trial. Nat Med. 2023;29:392-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 50]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
19.  Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. 2022;145:e895-e1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 671]  [Article Influence: 335.5]  [Reference Citation Analysis (0)]
20.  Mirarchi L, Amodeo S, Citarrella R, Licata A, Soresi M, Giannitrapani L. SGLT2 Inhibitors as the Most Promising Influencers on the Outcome of Non-Alcoholic Fatty Liver Disease. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
21.  Karras SN, Koufakis T, Mustafa OG, Kotsa K. Anti-incretin effect: The other face of Janus in human glucose homeostasis. Obes Rev. 2019;20:1597-1607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
22.  Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, Hazlehurst JM, Guo K; LEAN trial team, Abouda G, Aldersley MA, Stocken D, Gough SC, Tomlinson JW, Brown RM, Hübscher SG, Newsome PN. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016;387:679-690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1100]  [Cited by in F6Publishing: 1235]  [Article Influence: 154.4]  [Reference Citation Analysis (1)]
23.  Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, Sanyal AJ, Sejling AS, Harrison SA; NN9931-4296 Investigators. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N Engl J Med. 2021;384:1113-1124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 513]  [Cited by in F6Publishing: 838]  [Article Influence: 279.3]  [Reference Citation Analysis (0)]
24.  Loomba R, Abdelmalek MF, Armstrong MJ, Jara M, Kjær MS, Krarup N, Lawitz E, Ratziu V, Sanyal AJ, Schattenberg JM, Newsome PN; NN9931-4492 investigators. Semaglutide 2·4 mg once weekly in patients with non-alcoholic steatohepatitis-related cirrhosis: a randomised, placebo-controlled phase 2 trial. Lancet Gastroenterol Hepatol. 2023;8:511-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 113]  [Article Influence: 113.0]  [Reference Citation Analysis (0)]
25.  Mantovani A, Petracca G, Beatrice G, Csermely A, Lonardo A, Targher G. Glucagon-Like Peptide-1 Receptor Agonists for Treatment of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis: An Updated Meta-Analysis of Randomized Controlled Trials. Metabolites. 2021;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 138]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
26.  Yen FS, Hou MC, Cheng-Chung Wei J, Shih YH, Hsu CY, Hsu CC, Hwu CM. Glucagon-like Peptide-1 Receptor Agonist Use in Patients With Liver Cirrhosis and Type 2 Diabetes. Clin Gastroenterol Hepatol. 2024;22:1255-1264.e18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
27.  Simon TG, Patorno E, Schneeweiss S. Glucagon-Like Peptide-1 Receptor Agonists and Hepatic Decompensation Events in Patients With Cirrhosis and Diabetes. Clin Gastroenterol Hepatol. 2022;20:1382-1393.e19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 28]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
28.  Frías JP, Davies MJ, Rosenstock J, Pérez Manghi FC, Fernández Landó L, Bergman BK, Liu B, Cui X, Brown K; SURPASS-2 Investigators. Tirzepatide versus Semaglutide Once Weekly in Patients with Type 2 Diabetes. N Engl J Med. 2021;385:503-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 312]  [Cited by in F6Publishing: 653]  [Article Influence: 217.7]  [Reference Citation Analysis (0)]
29.  Hartman ML, Sanyal AJ, Loomba R, Wilson JM, Nikooienejad A, Bray R, Karanikas CA, Duffin KL, Robins DA, Haupt A. Effects of Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide on Biomarkers of Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes. Diabetes Care. 2020;43:1352-1355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 174]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
30.  Gastaldelli A, Cusi K, Fernández Landó L, Bray R, Brouwers B, Rodríguez Á. Effect of tirzepatide versus insulin degludec on liver fat content and abdominal adipose tissue in people with type 2 diabetes (SURPASS-3 MRI): a substudy of the randomised, open-label, parallel-group, phase 3 SURPASS-3 trial. Lancet Diabetes Endocrinol. 2022;10:393-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 180]  [Article Influence: 90.0]  [Reference Citation Analysis (0)]
31.  Romero-Gómez M, Lawitz E, Shankar RR, Chaudhri E, Liu J, Lam RLH, Kaufman KD, Engel SS; MK-6024 P001 Study Group. A phase IIa active-comparator-controlled study to evaluate the efficacy and safety of efinopegdutide in patients with non-alcoholic fatty liver disease. J Hepatol. 2023;79:888-897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 39]  [Reference Citation Analysis (0)]
32.  Yin YH, Sang LX, Chang B. Potential therapeutic targets for nonalcoholic fatty liver disease: Glucagon-like peptide 1. World J Gastroenterol. 2023;29:6235-6238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]