BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 7, 2025; 31(37): 111435
Published online Oct 7, 2025. doi: 10.3748/wjg.v31.i37.111435
Efficacy and safety of anti-obesity drugs in metabolic dysfunction-associated steatotic liver disease: An updated review
Marcio J Concepción-Zavaleta, Jenyfer M Fuentes-Mendoza, Grupo de Investigación en Neurociencias, Metabolismo, Efectividad Clínica y Sanitaria, Universidad Científica del Sur, Lima 15067, Peru
Jhean G Gonzáles-Yovera, Department of Gastroenterology, Hospital Nacional Guillermo Almenara Irigoyén, Lima 15018, Peru
Gemma Y Ruvalcaba-Barbosa, Leonardo D Cura-Rodríguez, Josué S González-Rodríguez, Aranza I Pérez-Reyes, School of Medicine, Universidad Autónoma de San Luis Potosí, San Luis Potosi 78210, Mexico
Luis A Concepción-Urteaga, School of Medicine, Universidad Nacional de Trujillo, Trujillo 13011, La Libertad, Peru
Juan Eduardo Quiroz-Aldave, Division of Medicine, Hospital de Apoyo Chepén, Chepén 13871, Peru
José Paz-Ibarra, School of Medicine, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
José Paz-Ibarra, Department of Endocrinology, Edgardo Rebagliati Martins National Hospital, Lima 15087, Peru
ORCID number: Marcio J Concepción-Zavaleta (0000-0001-9719-1875); Jenyfer M Fuentes-Mendoza (0000-0002-4682-3999); Jhean G Gonzáles-Yovera (0000-0002-5809-3006); Gemma Y Ruvalcaba-Barbosa (0009-0003-3423-2942); Leonardo D Cura-Rodríguez (0009-0005-8324-2391); Josué S González-Rodríguez (0009-0002-9372-7315); Luis A Concepción-Urteaga (0000-0003-0462-3101); Aranza I Pérez-Reyes (0009-0003-1560-3859); Juan Eduardo Quiroz-Aldave (0000-0001-8286-095X); José Paz-Ibarra (0000-0002-2851-3727).
Co-first authors: Marcio J Concepción-Zavaleta and Jenyfer M Fuentes-Mendoza.
Author contributions: Concepción-Zavaleta MJ and Fuentes-Mendoza JM conceptualized and designed the study, supervised the process, and made critical revisions, they contributed equally to this manuscript and are co-first authors; Gonzáles-Yovera JG provided senior clinical insight and contributed to review supervision and interpretation of findings; Ruvalcaba-Barbosa GY and Cura-Rodríguez LD participated in data collection; Ruvalcaba-Barbosa GY participated in manuscript drafting and formatting; Cura-Rodríguez LD, Pérez-Reyes AI, and Paz-Ibarra J assisted in manuscript review; González-Rodríguez JS, Quiroz-Aldave JE, and Paz-Ibarra J collaborated in data interpretation; Concepción-Urteaga LA contributed to manuscript revision and institutional perspective; Pérez-Reyes AI helped in manuscript preparation; Quiroz-Aldave JE contributed to institutional coordination and manuscript supervision; Paz-Ibarra J drafted the original manuscript. All authors approved the submitted version of the 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: Marcio J Concepción-Zavaleta, MD, Grupo de Investigación en Neurociencias, Metabolismo, Efectividad Clínica y Sanitaria, Universidad Científica del Sur, 19 Panamericana Sur Km, Villa El Salvador, Lima 15067, Peru. mconcepcion@cientifica.edu.pe
Received: June 30, 2025
Revised: July 24, 2025
Accepted: August 25, 2025
Published online: October 7, 2025
Processing time: 88 Days and 1 Hours

Abstract

Obesity is a major driver of metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, metabolic dysfunction-associated steatohepatitis (MASH). As the global prevalence of obesity continues to rise, the burden of MASLD/MASH is increasing, posing significant challenges to healthcare systems. The use of anti-obesity medications (AOMs) in this population is complex due to altered hepatic metabolism, safety concerns, and potential hepatotoxicity. Recent advances in pharmacologic agents, such as glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1 RAs), dual GLP-1/glucose-gastric inhibitory polypeptide (GIP) agonists, and triple GLP-1/GIP/glucagon agonists, have shown promising metabolic effects in the general population. Among these, GLP-1 RAs (e.g., liraglutide and semaglutide) consistently demonstrate hepatic benefits, including reductions in hepatic steatosis, improvements in liver enzyme profiles, and attenuation of fibrosis progression. Tirzepatide, a dual GLP-1/GIP agonist, has shown superior weight loss effects compared to GLP-1 receptor agonist monotherapy, with emerging but still limited data on hepatic outcomes in MASLD/MASH. Retatrutide, a triple agonist, has produced the most pronounced metabolic effects to date, although its impact on liver histology remains underexplored. Other AOMs, such as bupropion-naltrexone and phentermine-topiramate, require cautious use due to potential hepatotoxicity. Importantly, advanced MASLD may alter drug pharmacokinetics, underscoring the need for individualized therapy and close monitoring. This review provides an updated synthesis of the efficacy and safety of current and emerging AOMs in patients with MASLD/MASH and highlights the urgent need for further research to define optimal pharmacological strategies in this high-risk population.

Key Words: Obesity; Metabolic dysfunction-associated steatotic liver disease; Metabolic dysfunction-associated steatohepatitis; Glucagon-like peptide-1 receptor agonists; Tirzepatide; Retatrutide; Anti-obesity medications

Core Tip: Obesity is a leading modifiable risk factor for chronic liver disease, particularly metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis. Glucagon-like peptide-1 receptor agonists and emerging dual and triple incretin agonists offer promising metabolic and hepatic benefits. However, their use in advanced liver disease raises concerns due to altered pharmacokinetics and limited safety data. This review synthesizes current evidence on the efficacy and safety of anti-obesity medications in individuals with metabolic dysfunction-associated steatotic liver disease/metabolic dysfunction-associated steatohepatitis and underscores the need for personalized treatment strategies and robust clinical trials.



INTRODUCTION

Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most prevalent chronic liver disease worldwide, affecting nearly 30% of the adult population[1]. Its clinical impact extends beyond hepatic injury, contributing substantially to cardiovascular disease, chronic kidney disease, and malignancies[2,3]. As a consequence of rising global obesity rates, the burden of MASLD is expected to escalate, straining healthcare systems across both high-income and low-income countries. Throughout this review, the term “MASLD” is used in alignment with the 2023 international consensus on updated nomenclature, which replaced the former term “non-alcoholic fatty liver disease”[4]. Likewise, “metabolic dysfunction-associated steatohepatitis” substitutes the previously used “non-alcoholic steatohepatitis”[5]. References to non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH) appear exclusively in the context of historical terminology or when citing original sources published before the nomenclature update. This approach ensures consistency, scientific accuracy, and inclusivity of earlier research findings. Despite increasing awareness, MASLD remains significantly underdiagnosed, particularly in primary care settings, due to its asymptomatic nature in early stages. To address this challenge, non-invasive diagnostic tools, such as vibration-controlled transient elastography (VCTE) and serum-based fibrosis scores like the fibrosis-4 (FIB-4) index and the NAFLD fibrosis score, have been developed to estimate fibrosis risk and reduce reliance on liver biopsy[6,7]. However, diagnostic accuracy varies, particularly in individuals with obesity or type 2 diabetes mellitus (T2DM)[8,9]. Obesity is recognized as the most powerful modifiable driver of MASLD, primarily via mechanisms involving insulin resistance (IR), adipose tissue dysfunction, and chronic low-grade inflammation[10]. Sustained weight loss has been shown to improve steatosis, inflammation, and even fibrosis, with clinical benefits observed from reductions as modest as 5%, and more profound histological improvement with ≥ 10% weight loss[10,11]. However, achieving and maintaining weight loss through lifestyle modification alone remains challenging for most patients. In this context, anti-obesity medications have emerged as a promising adjunct. These agents, which include incretin-based therapies, centrally acting agents, and novel multi-agonists, target various metabolic pathways to promote weight reduction, improve insulin sensitivity, and modulate lipid metabolism. Early clinical trials suggest that some anti-obesity medications may exert beneficial effects on hepatic outcomes in MASLD, though their role remains under investigation.

In parallel, alternative pharmacologic strategies - such as insulin sensitizers like pioglitazone - have shown histologic benefit in selected MASLD populations. However, concerns regarding side effects and long-term safety have limited their widespread adoption[12,13]. The evolving pharmacological landscape highlights the need for critical appraisal of efficacy, safety, and applicability of these agents across the MASLD spectrum. Importantly, significant knowledge gaps persist, particularly in advanced fibrosis and cirrhosis where altered pharmacokinetics and comorbidities complicate treatment. For instance, recent studies have reported changes in very-low-density lipoprotein composition and secretion in advanced disease stages, suggesting impaired hepatic synthetic function and altered lipid homeostasis[14]. These findings raise questions about the suitability of specific weight-centric therapies in late-stage MASLD. Therefore, the objective of this narrative review is to provide a comprehensive synthesis of current evidence on MASLD, including its etiology and pathophysiology, non-invasive diagnostic tools, and the efficacy and safety of anti-obesity pharmacotherapies across the disease spectrum. This work also aims to identify research gaps and propose future directions for improving care in this growing population.

LITERATURE REVIEW

A structured narrative review was conducted to assess the efficacy and safety of anti-obesity pharmacological agents in patients with MASLD. A comprehensive literature search was performed across five major databases (PubMed, Scopus, Web of Science, EMBASE, and SciELO) covering all publications up to June 30, 2025. The search strategy combined Medical Subject Headings terms and free-text keywords using Boolean operators as follows: (“anti-obesity drugs” AND “MASLD” OR “NAFLD”) AND (“efficacy” AND “safety”) AND (“GLP-1 receptor agonists” OR “tirzepatide” OR “orlistat” OR “semaglutide”). Although the term NAFLD was included in the search to ensure comprehensive retrieval of relevant literature, it is used herein to reflect the broader clinical spectrum now reclassified as MASLD, following updated nomenclature guidelines. Eligible studies included peer-reviewed articles published in English or Spanish, encompassing randomized controlled trials, cohort studies, meta-analyses, real-world evidence, and systematic reviews. The scope of this review focused on anti-obesity pharmacotherapies with primary or secondary approval for weight management, particularly those with emerging evidence in MASLD. Agents such as metformin and orlistat were considered during screening but ultimately excluded due to limited MASLD-specific evidence (e.g., metformin) or insufficient long-term weight loss efficacy in recent comparative trials (e.g., orlistat). Conference abstracts, non-peer-reviewed materials, and animal-only studies were excluded. No publication date limits were applied to capture both foundational and recent evidence. A total of 155 studies were included in this review, of which 50 (35.5%) were published within the last five years, reflecting the integration of the most up-to-date therapeutic developments.

ETIOLOGY AND PATHOPHYSIOLOGY OF MASLD
Role of IR, lipotoxicity, and inflammation

IR, lipotoxicity, oxidative stress, and inflammation form a pathophysiological continuum that drives the development and progression of MASLD. IR impairs glucose uptake and increases adipose tissue lipolysis, leading to an increased flux of free fatty acids (FFAs) to the liver (Figure 1)[5,15,16]. These FFAs exceed the hepatocyte’s capacity to safely store or oxidize lipids, resulting in lipotoxic intermediate accumulation, mitochondrial dysfunction, and endoplasmic reticulum stress. This cascade generates reactive oxygen species (ROS), which induce lipid peroxidation and damage mitochondrial DNA, proteins, and membranes[17,18]. ROS also initiate inflammatory pathways and activate damage-associated molecular patterns, intensifying hepatocellular injury[18,19]. Injured hepatocytes release cytokines and chemokines [e.g., tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β], which recruit and activate hepatic macrophages (Kupffer cells) and circulating monocytes[19-22]. This self-amplifying inflammatory loop not only perpetuates liver injury but also plays a key role in activating hepatic stellate cells (HSCs), thus linking metabolic dysfunction to fibrogenesis[20-22].

Figure 1
Figure 1 Mechanisms of obesity and liver disease. Insulin resistance leads to elevated free fatty acids, hepatic lipid accumulation, oxidative stress, and inflammation. These processes activate hepatic stellate cells and promote liver fibrosis. Image adapted from Servier Medical ART (https://smart.servier.com/), licensed under CC BY 4.0 (https://creativecommons.org/Licenses/by/4.0/). Glu: Glucose; FFA: Free fatty acids; IL: Interleukin; TNF: Tumor necrosis factor.
Activation of HSCs

Under normal physiological conditions, HSCs remain in a quiescent state. However, in response to inflammatory cytokines, ROS, and damage-associated molecular patterns released by injured hepatocytes and activated immune cells, HSCs transdifferentiate into proliferative, contractile myofibroblasts[17,20,23]. These activated cells produce large quantities of extracellular matrix proteins, particularly type I collagen, and directly contribute to hepatic fibrosis. Beyond their fibrogenic activity, HSCs also secrete proinflammatory mediators and chemokines, amplifying immune cell recruitment and sustaining hepatic inflammation[20,23]. The reciprocal interaction between HSCs and hepatic macrophages is central to the orchestration of both fibrogenesis and inflammation in MASLD[20]. Moreover, HSC activation has been linked to mitochondrial dysfunction and worsening of systemic IR[23].

Progression from steatosis to fibrosis

The development of MASLD from simple steatosis to steatohepatitis and ultimately fibrosis involves a multifactorial pathogenic cascade. Hepatic steatosis, defined by excessive triglyceride accumulation in hepatocytes, represents the initial step in this spectrum. It results primarily from IR and adipose tissue dysfunction, which drive increased FFA delivery to the liver and upregulated de novo lipogenesis[15,24]. Progression to MASLD is marked by hepatocellular stress, triggering local immune responses and oxidative injury. Resident liver immune cells detect this stress and release proinflammatory mediators such as IL-17A, which in turn promote the recruitment of myeloid-derived inflammatory cells that secrete cytokines including TNF, transforming growth factor beta, and IL-1β[25]. This sustained inflammation activates HSCs, initiating fibrogenesis through deposition of collagen and other extracellular matrix components[17]. Fibrosis is further driven by oxidative stress, mitochondrial damage, and inflammasome activation - complications of lipotoxicity and excess FFAs[15]. Moreover, genetic and epigenetic predispositions, along with alterations in the gut-liver axis, modulate the pace and severity of fibrotic progression[15,26]. Understanding the multifactorial etiology and complex pathophysiology of MASLD highlights the importance of early and accurate detection. Given its often silent progression, particularly in individuals with metabolic risk factors, timely diagnosis is essential to prevent irreversible liver damage. Many diagnostic tools are specifically designed to detect the downstream consequences of these underlying mechanisms - such as hepatic steatosis, inflammation, and fibrosis - through surrogate metabolic markers, imaging techniques, and fibrosis scoring systems. The following section outlines current strategies for identifying MASLD, including non-invasive tests (NITs).

DIAGNOSIS OF MASLD AND METABOLIC DYSFUNCTION-ASSOCIATED STEATOHEPATITIS

The diagnosis of MASLD and metabolic dysfunction-associated steatohepatitis (MASH) relies on a combination of clinical evaluation, laboratory markers, imaging techniques, and, in select cases, liver biopsy. Accurate fibrosis staging is critical for risk stratification and therapeutic decision-making.

VCTE

VCTE, commonly known as transient elastography, is a non-invasive imaging technique used to assess liver stiffness, which correlates with fibrosis in MASLD. VCTE, commonly performed with the FibroScan device, is the most validated method for assessing fibrosis in MASLD. It measures liver stiffness by calculating the velocity of a shear wave through hepatic tissue. A liver stiffness measurement (LSM) < 8.0 kPa effectively excludes advanced fibrosis, while values ≥ 8.0 kPa indicate increased risk and warrant further evaluation. LSM between 8 and 12 kPa suggest fibrotic. MASLD, and values > 12 kPa indicate advanced fibrosis, though the positive predictive value is limited[27,28]. Other elastography techniques, such as two-dimensional shear wave elastography and magnetic resonance elastography, offer comparable or superior diagnostic accuracy, but are less widely available. VCTE remains the first-line, non-invasive tool for fibrosis risk stratification and prognostic assessment in MASLD[29-31].

Non-invasive scoring systems

NITs are crucial for fibrosis risk stratification in MASLD. The FIB-4 index and the NAFLD fibrosis score are the most validated and widely endorsed scoring systems. Both rely on readily available clinical and laboratory data to estimate the likelihood of advanced fibrosis (Figures 2 and 3). The FIB-4 index, which incorporates age, aspartate transaminase, alanine transaminase (ALT), and platelet count, is recommended as the first-line NIT due to its simplicity and high negative predictive value. A FIB-4 score of < 1.3 (for adults aged < 65 years) effectively rules out advanced fibrosis, while a score of ≥ 1.3 indicates the need for further evaluation, such as the LSM or the enhanced liver fibrosis (ELF) test. For adults aged ≥ 65 years, a higher cutoff (1.9-2.0) improves specificity. FIB-4 is not validated in pediatric populations or adults aged < 35 years (Figure 2)[8,27,32]. The NAFLD fibrosis score, which includes age, body mass index, impaired fasting glucose or diabetes, aspartate transaminase/ALT ratio, platelet count, and albumin, is effective for both ruling out (low cutoff: < -1.455) and confirming (high cutoff: > 0.675) advanced fibrosis. However, it has an indeterminate range, requiring further testing in some cases (Figure 3)[9,28,33]. The FIB-4 is commonly used to assess advanced fibrosis risk, although it performs worse than the VCTE, a more direct and validated imaging-based method. Many patients with elevated LSM values (≥ 8 kPa) show normal scores, leading to false negatives and underdiagnosis, particularly in those with diabetes, obesity, or older age[8,9]. In individuals with multiple metabolic risk factors, false-negative rates may exceed 30%[9]. Its performance is age-dependent, with reduced sensitivity in younger adults and reduced specificity in the elderly[33]. Although its negative predictive value is high (particularly with cutoffs of < 1.3), the score has a low positive predictive value and a high rate of indeterminate results[28,34,35]. It performs worse than VCTE or direct serum biomarkers like the Hepascore[36-38]. Guidelines recommend starting with FIB-4, followed by confirmatory testing (e.g., VCTE, ELF) in high-risk patients. The sequential use of NAFLD fibrosis score after FIB-4 is generally discouraged[8,27].

Figure 2
Figure 2 Fibrosis-4 score: Components and interpretation flowchart. Flowchart illustrating the calculation and interpretation of the fibrosis-4 score, a non-invasive marker for advanced fibrosis in metabolic dysfunction-associated steatotic liver disease. The formula incorporates age, aspartate aminotransferase, alanine aminotransferase, and platelet count. Cut-off values are stratified to classify risk: < 1.3 indicates low risk, 1.3-2.67 is indeterminate, and > 2.67 suggests high risk, warranting further evaluation with vibration-controlled transient elastography or enhanced liver fibrosis testing. FIB-4: Fibrosis-4; AST: Aspartate aminotransferase; ALT: Alanine aminotransferase; VCTE: Vibration-controlled transient elastography; ELF: Enhanced liver fibrosis.
Figure 3
Figure 3 Non-alcoholic fatty liver disease fibrosis score: Components and interpretation flowchart. Flowchart summarizing the non-alcoholic fatty liver disease fibrosis score, which incorporates age, body mass index, fasting glucose or diabetes status, aspartate aminotransferase/alanine aminotransferase ratio, platelet count, and serum albumin. This tool stratifies patients by fibrosis risk: Values < -1.455 suggest low risk, > 0.675 indicate high risk, and intermediate values require further evaluation. The non-alcoholic fatty liver disease fibrosis score is widely used for non-invasive fibrosis screening in metabolic dysfunction-associated steatotic liver disease, especially in primary care. NAFLD: Non-alcoholic fatty liver disease; BMI: Body mass index; IFG: Impaired fasting glucose; AST: Aspartate aminotransferase; ALT: Alanine aminotransferase.
Magnetic resonance imaging-proton density fat fraction and its role in quantifying hepatic steatosis

Magnetic resonance imaging (MRI)-proton density fat fraction (PDFF) is a non-invasive and reliable method for quantifying liver fat content, with reproducibility that makes it valuable both for diagnosis and for monitoring treatment response. Studies have shown that reductions in liver fat content measured by MRI-PDFF correlate with histological improvements in steatohepatitis, notably when liver fat decreases by at least 30%[35,36]. MRI-based assessments are beneficial in clinical trials and treatment monitoring, as they provide a non-invasive means to evaluate the efficacy of interventions to reduce liver fat. For example, pharmacologic interventions have been shown to reduce liver fat content measured by MRI, although the impact on liver fibrosis may be limited[36]. MRI also enables tracking of hepatic fat reduction following lifestyle or bariatric interventions, where significant reductions in liver fat content have been observed[37,38].

Liver biopsy

Liver biopsy remains the gold standard for diagnosing and staging MASLD, though its use has become more selective with the advent of validated noninvasive diagnostic methods. Indications for liver biopsy include: (1) Diagnostic uncertainty when NITs yield inconclusive results; (2) Suspicion of advanced fibrosis or cirrhosis with equivocal noninvasive assessment; (3) Evaluation of suspected coexisting liver diseases; and (4) When biopsy can impact management decisions, such as clinical trial inclusion or advanced therapies[28,39-41]. Histological evaluation from biopsy provides detailed information on steatosis, inflammation, hepatocellular ballooning, fibrosis, and architectural changes, which can have prognostic value[28,41]. It is particularly crucial for distinguishing steatohepatitis from simple steatosis and for identifying rare histological patterns, especially in pediatric populations[40]. Despite its importance, liver biopsy has significant limitations. It is invasive, carries risks such as pain and bleeding, and can suffer from sampling and interobserver variability. Furthermore, it is not feasible for routine screening or long-term monitoring. Consequently, noninvasive biomarkers and imaging techniques, such as elastography and serum fibrosis panels (e.g., FIB-4, NAFLD fibrosis score, aspartate aminotransferase to platelet ratio index, or ELF), are prioritized for initial risk stratification and monitoring, such as inclusion in clinical trials or confirmation of non-invasive fibrosis scores and panels (e.g., FIB-4, NAFLD fibrosis score, aspartate aminotransferase to platelet ratio index, ELF)[28,42].

NON-PHARMACOLOGICAL TREATMENT FOR MASLD

Non-pharmacological management constitutes the cornerstone of treatment for MASLD. The primary interventions are lifestyle modification, including dietary changes, increased physical activity, and weight loss, with bariatric surgery considered in select cases[28,43].

Dietary modification

The most robust evidence supports a Mediterranean eating pattern, which emphasizes high intake of vegetables, fruits, whole grains, legumes, nuts, olive oil, and fish, while limiting saturated fats, refined carbohydrates, and added sugars. Caloric restriction to induce weight loss is essential, with a minimum target of 5% total body weight reduction to improve hepatic steatosis, and ≥ 10% weight loss is associated with greater histological improvement, including potential fibrosis regression. Reducing fructose and added sugars is recommended explicitly due to their association with hepatic fat accumulation and fibrosis progression. Diets should also limit saturated fat and starch, and focus on high fiber and unsaturated fats for additional cardiometabolic benefit[8,9,27,28,44].

Physical activity

Both aerobic and resistance exercise independently reduce hepatic steatosis and improve insulin sensitivity, with benefits observed even in the absence of significant weight loss. Exercise regimens should be individualized and structured, as higher engagement and intensity correlate with greater improvements in liver outcomes. Vigorous exercise may further limit progression to steatohepatitis[27,28,45].

Weight loss

Sustained weight reduction is the most effective non-pharmacological intervention for MASLD. Weight loss of at least 5% is necessary for improvement in steatosis, while ≥ 7%-10% is associated with resolution of liver steatosis and regression of fibrosis in a significant proportion of patients. Structured, individualized weight loss programs are more effective than standard counseling[8,46].

Bariatric surgery

For patients with obesity who meet established criteria for metabolic surgery, bariatric procedures (e.g., sleeve gastrectomy, Roux-en-Y gastric bypass) can result in substantial and sustained weight loss, resolution of MASLD and MASH, and improvement in fibrosis. Bariatric surgery is not recommended in decompensated cirrhosis and should be used with caution in compensated cirrhosis[8,36,47].

Alcohol and other lifestyle factors

Alcohol intake should be minimized, and complete abstinence is recommended in patients with clinically significant fibrosis (≥ F2), as alcohol is a cofactor for disease progression. Smoking cessation and management of other cardiometabolic risk factors are also important[28,36]. The foundation of MASLD management relies on comprehensive lifestyle intervention - caloric restriction with a Mediterranean-style diet, regular aerobic and resistance exercise, and weight loss of at least 5%-10%. Bariatric surgery is an option for eligible patients with obesity. These interventions not only improve liver histology but also address the high burden of cardiovascular and metabolic comorbidities in this population[43-45].

CURRENT PHARMACOLOGICAL TREATMENT FOR MASLD
GLP-1 RAs: Mechanisms and therapeutic benefits in MASLD

GLP-1 RAs promote weight loss and have demonstrated reductions in liver steatosis and inflammation in MASLD patients[42-46]. Additionally, GLP-1 RAs are associated with a reduced risk of major cardiovascular events, clinically significant portal hypertension, and all-cause mortality in patients with MASLD[48-50]. A meta-analysis of observational studies further supports an association between GLP-1 RA use and decreased risk of serious hepatic adverse outcomes, hepatic decompensation, and hepatocellular carcinoma in individuals with T2DM[51]. These agents act by modulating the satiety center in the brain, inhibiting glucagon secretion from pancreatic α-cells, and stimulating insulin production from β-cells through activation of the GLP-1 receptor. These key mechanisms are illustrated in Figure 4, highlighting their role in improving glycemic control and hepatic steatosis. Clinically relevant dosing and average weight loss effects for liraglutide and semaglutide are summarized in Table 1. In addition, hepatic outcomes such as ALT/gamma-glutamyl transpeptidase (GGT) reductions and histological resolution are detailed in Table 2[42-46]. Beyond their systemic metabolic benefits, GLP-1 RAs may exert direct intrahepatic actions that recalibrate the fibrotic and lipogenic machinery of the liver. Preclinical studies have demonstrated that these agents dampen HSC activation, a central engine of fibrosis, by silencing profibrotic signaling pathways such as transforming growth factor beta/smooth muscle actin and mad related family and curbing the expression of α-smooth muscle actin and collagen type I - molecular hallmarks of fibrogenic transformation[52]. In parallel, GLP-1 RAs have been shown to suppress the hepatic lipogenic program, notably through downregulation of sterol regulatory element-binding protein-1c and its enzymatic targets, acetyl-coenzyme A carboxylase and fatty acid synthase. This dual action not only restricts de novo lipid synthesis, but also mitigates intracellular triglyceride accumulation - an effect that unfolds independently of systemic insulin sensitivity or weight loss[53]. While the direct expression of GLP-1 RAs in hepatocytes remains a subject of debate, emerging evidence suggests a paracrine interplay involving non-parenchymal liver cells, including Kupffer cells and HSCs, which may act as intermediaries of these hepatotropic effects[54]. These findings broaden the mechanistic landscape of GLP-1 RAs, positioning them not merely as metabolic regulators, but as molecular circuit breakers capable of directly modulating hepatic injury and fibrosis in MASLD.

Figure 4
Figure 4 Mechanisms of action of glucagon-like peptide-1 receptor agonists. Glucagon-like peptide-1 receptor agonists exert their effects by acting on the hypothalamus to increase satiety and reduce appetite, delaying gastric emptying, stimulating insulin secretion from pancreatic β-cells, and suppressing glucagon release from α-cells. These mechanisms contribute to improved glycemic control and reduced hepatic steatosis. Image adapted from Servier Medical ART (https://smart.servier.com/), licensed under CC BY 4.0 (https://creativecommons.org/Licenses/by/4.0/). GLP-1 RA: Glucagon-like peptide-1 receptor agonists; MASLD: Metabolic dysfunction-associated steatotic liver disease; TNF: Tumor necrosis factor; IL: Interleukin.
Table 1 Overview of anti-obesity drug classes, mechanisms of action, and weight loss effects in metabolic dysfunction-associated steatotic liver disease.
Class
Drug
Dosage
Mechanism of action
Effect on body weight
GLP-1 RAsLiraglutide3.0 mg/dayGLP-1 receptor activation: Satiety, gastric emptying (↓)8%-10%
GLP-1 RAsSemaglutide2.4 mg/weekGLP-1 receptor activation: Satiety, gastric emptying (↓)10%-15%
Dual GLP-1/GIP agonistsTirzepatide5-15 mg/weekCombined GLP-1 and GIP receptor agonism15%-20%
Triple GLP-1/GIP/glucagon agonistsRetatrutide8-12 mg/week (in trials)GLP-1, GIP, and glucagon receptor agonistUp to 24%
FGF-21 analogsEfruxifermin28-70 mg/week (in trials)FGF21 receptor agonistApproximately 10%
CNS agentsPhentermine/topiramate7.5/46 mg/day to 15/92 mg/dayCentral appetite suppression via sympathomimetic + GABA modulation7%-9%
CNS agentsNaltrexone/bupropion8/90 mg BIDOpioid antagonist and dopamine/norepinephrine reuptake inhibitor5%-6%
Table 2 Efficacy and hepatic outcomes of anti-obesity drugs in metabolic dysfunction-associated steatotic liver disease.
Class
Drug
ALT
GGT
Histological improvement
Effect on fibrosis
MASLD-specific efficacy
Safety profile
Approval (FDA/EMA)
GLP-1 RAsLiraglutideDecreaseDecreaseNASH resolution (LEAN trial)No significant improvementHistological and biochemical improvement in NASH patientsGI side effects; contraindicated in MTC or gastroparesisApproved
GLP-1 RAsSemaglutideDecreaseDecreaseSteatohepatitis resolution (2021 phase 2)No fibrosis improvementDecrease hepatic fat and steatohepatitis resolutionWell tolerated; GI effects most commonApproved
Dual GLP-1/GIP agonistTirzepatideDecreaseDecreaseNo biopsy dataNot assessedDecrease liver fat (MRI-PDFF); improved insulin sensitivityGI symptoms; minimal hypoglycemiaFDA approved
Triple GLP-1/GIP/glucagon agonistRetatrutideDecreaseDecreaseNo biopsy dataNot assessedDecrease marked in hepatic fat in early-phase studiesGI adverse events; not tested in cirrhoticsInvestigational
FGF-21 analogEfruxiferminDecreaseDecrease↓Steatosis and ballooning (phase 2 MASH)Yes (F2-F3 patients)Biopsy-confirmed improvement in steatosis and fibrosisWell tolerated; long-term safety under investigationInvestigational
CNS agentPhentermine/topiramateUnclearUnclearNoneNonePresumed via weight loss; no liver-specific trialsNeurocognitive side effects; teratogenicityApproved (for obesity)
CNS agentNaltrexone/bupropionUnclearUnclearNoneNoneNo liver-specific dataNeuropsychiatric side effects; caution in seizuresApproved (for obesity)
Liraglutide: Evidence-based outcomes in liver disease

Liraglutide, a GLP-1 RA from the incretin class, was initially approved for the treatment of T2DM and later for weight management. It has also been shown to reduce cardiovascular risk and mortality[47]. As mentioned in the clinical practice guideline of the American Association of Clinical Endocrinology, liraglutide has also demonstrated beneficial effects in patients with NAFLD[13]. The “Liraglutide Efficacy and Action in NASH” trial demonstrated resolution of MASLD after 48 weeks of treatment, with no progression of liver fibrosis observed in 9 of 23 patients evaluated histologically[55]. In addition, liraglutide improved liver histology and significantly reduced serum levels of ALT, GGT, and other markers of hepatocellular injury[55]. As shown in Table 2, liraglutide has demonstrated resolution of MASH with improvements in ALT and GGT, although no significant impact on fibrosis was reported.

Semaglutide (approved): Therapeutic effects and clinical evidence

Semaglutide is a GLP-1 RAs primarily used for treating obesity and T2DM. In MASLD, semaglutide effectively reduces hepatic fat accumulation, improves insulin sensitivity, and decreases liver inflammation through specific mechanisms of action[56]. Clinical trials have reported significant benefits, including steatohepatitis resolution and improvements in hepatic fibrosis[57]. Gradual dose escalation is recommended to minimize gastrointestinal side effects, with liver-specific benefits observed at doses ranging from 0.1 mg to 0.4 mg daily in phase 2 studies and up to 2.4 mg weekly in phase 3 trials[57,58]. In patients with obesity and/or T2DM, semaglutide significantly reduced ALT and high-sensitivity C-reactive protein levels. Preclinical murine models of diet-induced steatotic liver disease demonstrated decreased expression of proinflammatory cytokines such as TNF-α, IL-1β, and IL-6[59]. These findings reinforce semaglutide’s therapeutic potential in MASLD management. According to Table 2, semaglutide led to resolution of steatohepatitis and a reduction in hepatic fat content, with good tolerability and minimal hepatic safety concerns.

GLP-1 RAs in cirrhosis: Safety considerations and contraindications

GLP-1 RAs have shown a favorable hepatic safety profile in patients with compensated cirrhosis (Child-Pugh A), with studies reporting reductions in hepatic decompensation (48%) and hepatocellular carcinoma (53%) compared to non-users[56,60]. A recent review confirmed these agents reduce hepatic complications in compensated cirrhosis without increasing all-cause mortality[61]. However, use in advanced cirrhosis (Child-Pugh B/C) requires caution due to limited direct evidence. One cohort study reported benefits in decompensated cirrhosis, but lacked Child-Pugh score specification, limiting conclusions[57]. Key concerns in advanced disease include: (1) Potential worsening of malnutrition via delayed gastric emptying; (2) Scarce pharmacokinetic data; and (3) Theoretical risks related to portal hypertension[60,62]. GLP-1 RAs are contraindicated in patients with a history of medullary thyroid carcinoma (boxed warning) or severe gastroparesis[42,62]. Gastrointestinal side effects (nausea, vomiting, diarrhea) occur in 20%-40% of patients, but are usually transient and dose-dependent. Regarding pancreatitis risk, extensive cohort studies and meta-analyses have found no significant association, although caution is advised in patients with a history of severe or idiopathic pancreatitis[63]. A multidisciplinary approach is strongly recommended for patients with advanced disease, including gradual dose titration and close monitoring during the first 12 weeks to optimize tolerability[57,60,62].

While GLP-1 RAs have shown a favorable short-term hepatic safety profile in patients with compensated cirrhosis, real-world data beyond two years of continuous use remain scarce. Significantly, the potential nutritional consequences of sustained GLP-1 RA therapy in this population - particularly unintended sarcopenia due to excessive or rapid weight loss - are underexplored. Although short-duration studies suggest reductions in hepatic decompensation and liver-related mortality[60-62], long-term cohort studies assessing body composition dynamics, nutrient absorption, and muscle mass preservation are lacking. This gap is particularly relevant for cirrhotic patients, whose nutritional reserve is often fragile and easily perturbed. Thus, caution is warranted when extrapolating short-term benefits to chronic use, and future prospective studies should evaluate the longitudinal impact of GLP-1 RAs on sarcopenia and functional status in this vulnerable subgroup.

Dual incretin agonists (GLP-1/GIP): Mechanisms and therapeutic potential

Dual incretin agonists targeting both GLP-1 and GIP receptors represent an emerging class of treatments for obesity and T2DM, improving postprandial metabolic regulation[42,64-67]. GLP-1 inhibits glucagon secretion at elevated glucose levels, while GIP enhances glucagon release at low glucose levels. Activating both receptors improves glycemic control and weight loss in T2DM[67,68]. Beyond glycemic regulation, these agents have been to exert beneficial hepatic effects through enhanced insulin sensitivity, reduction of hepatic fat accumulation by decreasing de novo lipogenesis and promoting fatty acid oxidation, as well as anti-inflammatory and antifibrotic actions. These combined effects contribute to improvements in liver histology and function observed in MASLD patients receiving dual incretin therapy[64-67]. While GLP-1 RAs have demonstrated hepatic benefits primarily through systemic metabolic improvements, the direct hepatic effects of incretin-based therapies remain under investigation. Notably, hepatocytes do not consistently express GLP-1 receptors, and single-cell transcriptomic studies have failed to detect meaningful GLP-1 RA expression in hepatic parenchymal cells[64,69]. This supports the view that most hepatic benefits are mediated indirectly, via improved insulin sensitivity, reduced systemic inflammation, and paracrine signaling from non-parenchymal cells such as Kupffer cells and HSCs[69]. The activation of GIP and glucagon receptors, unique to dual and triple incretin agonists, introduces additional mechanisms. GIP-1 RA improves adipose tissue function and nutrient partitioning. In contrast, glucagon receptor agonism enhances hepatic β-oxidation, reduces de novo lipogenesis, and increases energy expenditure - mechanisms that reduce intrahepatic lipid accumulation and inflammatory signaling[64,70].

In MASLD models, incretin agonists have shown the capacity to reduce HSC activation indirectly, likely by attenuating circulating cytokines (e.g., TNF-α, IL-6), lowering leptin signaling, and reducing oxidative stress[15,69]. Experimental studies have reported decreased α-smooth muscle actin and collagen type I expression in liver tissue following GLP-1 RA or dual agonist therapy. However, direct receptor-mediated antifibrotic effects on HSCs remain unconfirmed[69]. Comparatively, retatrutide, a triple GLP-1/GIP/glucagon receptor agonist, has demonstrated superior weight loss outcomes (up to 24%) relative to GLP-1 or dual agonists[71,72]. However, no biopsy-confirmed data on fibrosis regression have been published to date, and its hepatic effects have only been assessed through MRI-PDFF in early-phase studies[73]. Although weight loss of ≥ 10% has been associated with fibrosis improvement in MASLD[10], it remains unclear whether this benefit extends proportionally with greater weight loss or whether ceiling effects exist. Furthermore, triple agonists have not yet been studied in patients with advanced fibrosis or cirrhosis, limiting their clinical applicability in high-risk MASLD cohorts[74]. In summary, while incretin-based therapies exert compelling indirect metabolic and hepatic benefits, current evidence does not support a direct antifibrotic mechanism, particularly in patients with advanced disease. Further trials with biopsy-based endpoints are needed to establish the long-term hepatic outcomes of dual and triple incretin agonists[71-75].

Tirzepatide (approved): Efficacy and impact on metabolic and hepatic outcomes

Tirzepatide is a dual GIP and GLP-1 RA that regulates carbohydrate metabolism and appetite by increasing insulin secretion, reducing serum glucagon, suppressing food intake, and delaying gastric emptying, thereby improving glucose control and inducing weight loss[64,76-78]. It also enhances insulin signaling and lipolysis[70]. Clinical trials have demonstrated tirzepatide’s superiority over GLP-1 RAs in reducing glycated hemoglobin A1c (HbA1c) (> 2%) and body weight (> 15%)[79,80]. Gastrointestinal adverse effects are common, but it does not increase hypoglycemia risk[81].

The SURMOUNT trials evaluated tirzepatide’s efficacy and safety in adults with obesity or overweight, with or without T2DM[82]. Phase 3 studies (SURMOUNT-1 to SURMOUNT-4) focused on weight reduction and quality of life[82]. SURMOUNT-1 showed significant weight loss and quality-of-life improvement in obese adults[83]. The SURPASS-3 trial provided liver-specific evidence, demonstrating tirzepatide’s efficacy and safety[84,85]. MRI-PDFF data confirmed fat reduction. Tirzepatide also reduced visceral and hepatic fat, while slightly increasing abdominal subcutaneous fat, indicating a favorable redistribution of adipose tissue[86]. These findings underscore tirzepatide’s potential to improve hepatic and visceral fat profiles alongside weight reduction. Table 1 outlines its dual agonism mechanism and robust weight loss profile, while Table 2 shows its hepatic effects, including liver fat reduction and improved insulin sensitivity, although no biopsy data are yet available.

Other approved agents

Phentermine/topiramate: Appetite suppression (approved for obesity) and emerging liver implications: Phentermine is a sympathomimetic agent that increases norepinephrine release in the hypothalamus by both stimulating its release and inhibiting its reuptake, thereby enhancing synaptic availability. This mechanism suppresses appetite via β-2 adrenergic receptor activation[87]. Topiramate, primarily approved for epilepsy, contributes to weight loss by modulating sodium and calcium channels, enhancing γ-aminobutyric acid type a receptor activity, and inhibiting α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor/kainate receptor at the hypothalamic level[88,89]. Although current evidence for these agents in MASLD is limited, extrapolation from studies in the general obese population should be interpreted with caution until MASLD-specific clinical trials are available.

The combination of phentermine and topiramate has been associated with moderate weight loss (5%-9%), and clinical studies indicate that their co-administration improves tolerability by reducing individual drug concentrations while enhancing therapeutic efficacy. Lifestyle modifications are often recommended alongside pharmacotherapy to optimize outcomes[90]. Several long-term studies suggest that weight loss exceeding 10 kg can be achieved and maintained over several years[91]. In comparative analyses, this combination has demonstrated superior efficacy in achieving a ≥ 5% body weight reduction within six months compared to other therapies[92]. Although data specific to MASLD are scarce, this combination appears promising due to its capacity for significant weight loss and has shown encouraging outcomes in patients with hepatic impairment[93]. Nevertheless, as with topiramate monotherapy, concerns remain regarding teratogenic effects and a persistent risk of neurodevelopmental disorders, particularly intellectual disability, as reported in clinical trials[87,94].

Naltrexone/bupropion (approved for obesity): Reward circuit modulation and weight reduction: Naltrexone is an opioid receptor antagonist that blocks the orexigenic effect of β-endorphin via mu-opioid receptor antagonism. Bupropion, a norepinephrine and dopamine reuptake inhibitor, acts at the hypothalamic level by stimulating the proopiomelanocortin pathway, thereby enhancing dopaminergic signaling and suppressing appetite. Together, these agents produce synergistic appetite suppression, with naltrexone potentiating bupropion’s effect[95]. Through dopaminergic and noradrenergic modulation, this combination helps reduce food-related reward signaling, supporting better adherence to dietary interventions and contributing to clinically relevant weight loss[95,96]. No clinical trials to date have evaluated the efficacy of naltrexone/bupropion specifically in MASLD patients. Existing evidence is limited to populations with overweight or general obesity, underscoring the need for future studies assessing safety and therapeutic efficacy in MASLD contexts[97-99].

INTEGRATING NON-PHARMACOLOGICAL AND PHARMACOLOGICAL STRATEGIES IN MASLD MANAGEMENT

Current management of MASLD is fundamentally based on the integration of non-pharmacological and pharmacological interventions, which are considered complementary and synergistic rather than mutually exclusive. Lifestyle modification - including dietary changes, increased physical activity, and behavioral interventions - remains the cornerstone of MASLD therapy and is recommended for all patients regardless of pharmacological treatment status[28,100,101]. These interventions target the underlying metabolic derangements and are associated with improvements in hepatic steatosis, liver enzymes, and cardiometabolic risk factors. Pharmacological therapy is considered in patients who do not achieve sufficient benefit from lifestyle modification alone, particularly those with established steatohepatitis (MASH), significant fibrosis, or high-risk metabolic comorbidities such as T2DM or obesity[102,103]. In summary, current consensus in the medical literature is that pharmacological and non-pharmacological treatments for MASLD should be implemented in a complementary fashion, with lifestyle modification as the foundation and pharmacotherapy individualized based on disease severity, comorbidities, and response to non-pharmacological measures[102,103].

EMERGING CONCEPTS AND INVESTIGATIONAL THERAPIES
MASLD evidence

MASLD is a common condition marked by hepatic steatosis in the context of metabolic risk factors such as obesity, T2DM, hypertension, and dyslipidemia[96]. Its spectrum ranges from simple steatosis to MASLD, fibrosis, cirrhosis, and hepatocellular carcinoma[104-106]. Recent studies reveal distinct phenotypes driven by liver-specific genetic variants and cardiometabolic traits, contributing to increased cardiovascular risk[107,108]. This heterogeneity underscores the need for personalized therapeutic approaches. Lipidomics analyses have demonstrated significant alterations in hepatic lipid composition correlated with disease progression from MASLD to MASH[108,109]. Additionally, metabolomic studies have identified unique metabolic signatures, such as changes in lysophosphatidylcholines and xanthine levels, which are associated with inflammation and IR[110]. Genetic variants, particularly in patatin-like phospholipase domain containing 3 and transmembrane 6 superfamily member 2, confer susceptibility to MASLD and may inform precision medicine approaches[111].

Retatrutide: A triple agonist targeting GIP, GLP-1, and glucagon receptors

Retatrutide is a novel triple agonist targeting the GIP, glucagon, and GLP-1 receptors. It is currently under investigation for the treatment of obesity and T2DM, and has shown promising results in phase 2 clinical trials. These studies reported substantial reductions in body weight and improved glycemic control. In a phase 2 obesity trial, retatrutide achieved up to 24.2% weight loss over 48 weeks at a dose of 12 mg, compared to a 2.1% reduction in the placebo group[71]. In patients with T2DM, it significantly improved HbA1c levels, outperforming dulaglutide, another GLP-1 RA[72]. Retatrutide has also been associated with notable reductions in liver fat in MASLD patients[73]. However, gastrointestinal side effects are common, as expected with this class of medications[71,72,75]. Clinical applicability: Phase 2 trials predominantly included patients with early-stage fibrosis (F1-F2), and the effects on fibrosis regression or histological endpoints such as steatosis resolution remain preliminary. Ongoing phase 3 trials are expected to provide crucial data on its impact on advanced fibrosis and cirrhosis, and potential regulatory submissions are anticipated within the next few years[74,75]. While triple agonists offer substantial weight loss and liver fat reduction, current evidence derives from early-phase trials in general obesity cohorts. There is no published histological data confirming benefits on inflammation or fibrosis, and safety in cirrhotic or high-risk MASLD patients remains unknown[71-75]. As summarized in Table 2, retatrutide has shown substantial hepatic fat reduction in early trials, though its histological effects and cirrhosis safety profile remain untested.

GLP-1 and fibroblast growth factor-21 combinations: Fibrosis regression and metabolic benefits

Pegbelfermin and efruxifermin: Pegbelfermin and efruxifermin are analogs of fibroblast growth factor (FGF)-21, which are being investigated for MASLD and related liver diseases. They differ in pharmacological design, clinical progression, and outcomes[112]. Efruxifermin is an engineered Fc-FGF-21 fusion protein with enhanced pharmacokinetics, including a prolonged half-life and improved receptor selectivity. It activates FGF-R1c, FGF-R2c, and FGF-R3c, but not FGF-R4, thus avoiding elevations in low-density lipoproteins cholesterol[113,114]. Clinical trials indicate that efruxifermin is safe and well-tolerated, with mainly mild to moderate gastrointestinal side effects[113]. Clinical applicability: Efruxifermin has shown significant efficacy in reducing liver fat, improving fibrosis scores, and enhancing metabolic parameters in patients with MASLD and compensated cirrhosis. It has demonstrated fibrosis regression in F2-F3 patients. However, data on cirrhosis patients are still limited, and ongoing phase 3 trials, several limitations restrict the current applicability of FGF-21 analogs in MASLD. While efruxifermin has demonstrated encouraging reductions in liver fat and improvements in fibrosis scores, biopsy-confirmed outcomes remain short-term and may vary significantly depending on baseline disease severity and metabolic comorbidities[115,116]. Moreover, pharmacokinetic profiles in patients with hepatic impairment are poorly understood, raising concerns about altered drug metabolism, potential accumulation, and reduced therapeutic efficacy[117]. Although gastrointestinal adverse events are the most commonly reported, long-term safety remains uncertain - particularly in populations with portal hypertension or decompensated liver function[115,117]. Pegozafermin, another FGF-21 analog, has shown similar antifibrotic activity in phase 2 trials, but direct comparisons and cirrhosis-specific data are lacking[97,116]. Further research is needed to clarify optimal patient selection, safety margins, and the positioning of these agents within MASLD treatment pathways. Table 2 highlights its ability to improve steatosis and fibrosis in F2-F3 patients, supported by biopsy-confirmed outcomes in phase 2 trials.

Oral GLP-1 agents: Orforglipron and danuglipron are oral, nonpeptide GLP-1 RAs that have demonstrated meaningful reductions in HbA1c (up to 1.3%) and body weight (up to 9.9%), with an acceptable safety profile in patients with obesity and T2DM[118]. In a phase 2 trial, danuglipron significantly reduced HbA1c, fasting glucose, and body weight in adults with T2DM, although gastrointestinal adverse effects led to treatment discontinuation in some patients[119]. Danuglipron research in MASLD is limited, and one study was terminated as the sponsor opted to focus on patients with obesity and T2DM without active liver disease[120]. Orforglipron has shown efficacy and safety comparable to injectable GLP-1 RAs, offering a potentially more accessible oral alternative. However, its effects on liver disease have not been studied[121]. Clinical applicability: Orforglipron and danuglipron are promising oral alternatives to injectable GLP-1 RAs, with comparable efficacy in weight loss and IR improvement. However, their impact on liver disease has not yet been extensively studied[122,123]. Studies specifically addressing MASLD are still in early stages, and current data on their hepatic benefits remain lacking. Ongoing trials are needed to assess their effectiveness in liver-related metabolic diseases, and regulatory progress is expected soon[124,125].

Amylin analogs (cagrilintide and semaglutide)

CagriSema, a co-formulation of cagrilintide (a long-acting amylin analog) and semaglutide (a GLP-1 RA), exerts synergistic effects on glycemic control and weight loss, likely through complementary mechanisms[99,126]. Although several studies highlight its benefits for obesity and diabetes, no evidence currently supports its use in MASLD or MASH[127-129]. Recent systematic reviews have reported that CagriSema outperforms semaglutide alone in weight reduction and glycemic control, but with a higher incidence of gastrointestinal side effects, particularly vomiting[130]. To date, no clinical trials or guidelines include CagriSema as a treatment option for MASLD. Although not yet approved for MASLD, CagriSema’s synergistic action and clinical potential are outlined in Table 1[131]. Clinical applicability: Although effective for weight reduction and glycemic control, CagriSema has not been investigated explicitly in MASLD or MASH patients, and no clinical trials have included it as a treatment option for these conditions. Its gastrointestinal side effects, particularly vomiting, also raise concerns regarding its clinical application in MASLD.

Novel molecular targets: FGF-21, amylin, leptin, mitochondrial uncouplers

Recent compounds - including SR4, CZ5, OPC-163493, compound 6j, BAM15, HU6, and MB-X01Y03 - demonstrate potential for treating obesity, T2DM, and liver-related metabolic diseases, with reduced toxicity and tissue-specific action compared to classical uncouplers like 2,4-dinitrophenol[132]. Leptin analogs, such as metreleptin, have improved metabolic parameters in congenital leptin deficiency and relative leptin deficiency in MASLD; however, they have not shown sufficient efficacy to support approval for MASLD treatment[133-135].

FUTURE DIRECTIONS AND UNMET NEEDS
Lack of long-term outcome data

A critical limitation in MASLD research is the scarcity of long-term data on pharmacologic interventions, particularly concerning liver safety in patients with stage F2-F3 fibrosis and advanced disease[136,137]. Most available studies are of short or medium duration, which restricts the extrapolation of findings to chronic treatment scenarios[138]. There is an urgent need for long-term studies that assess both the sustained efficacy and safety of emerging therapies over time.

Absence of validated clinical endpoints

Recent investigations, such as the randomized phase 2a trial of retatrutide, a triple hormone receptor agonist, have shown promising reductions in liver fat and improved metabolic parameters in patients with MASLD[139]. These results suggest potential benefits in liver disease management. However, the trial also emphasized the lack of validated clinical endpoints, as long-term safety and histologic efficacy remain unknown. Moreover, the optimal balance of receptor agonism remains unclear[140]. Although incretin-based therapies have demonstrated indirect hepatic benefits through weight reduction and improved IR, their direct effects on hepatocytes and HSCs remain debated[69]. Some evidence suggests these agents may not act directly on liver parenchymal cells, further complicating endpoint validation. In summary, despite encouraging metabolic and hepatic responses to triple agonists such as retatrutide, comprehensive data on durability, safety, and mechanistic action are lacking. Future research should aim to clarify the mechanisms of action on liver tissue, identify the patient populations most likely to benefit, and establish standardized, validated clinical endpoints[141,142].

Need for combination therapy trials

Combination therapy represents a promising approach for complex metabolic conditions like MASLD. Agents such as retatrutide and CagriSema have shown synergistic effects in obesity and T2DM, yet evidence for their role in MASLD is limited, particularly in advanced disease[121,143]. Ongoing trials with efruxifermin and GLP-1 + farnesoid X receptor/FGF-21 combinations offer potential as multifaceted therapies for MASLD and deserve further investigation[144].

Priorities for future phase 3/phase 4 trials

Several late-phase clinical trials are actively evaluating new pharmacologic strategies for MASLD, with a focus on both steatosis resolution and fibrosis regression. The MAESTRO-NASH trial assessed resmetirom, a selective thyroid hormone receptor-β agonist, in patients with MASLD. While MRI-PDFF showed significant reductions in liver fat, histologic response was limited in participants who failed to achieve ≥ 30% fat reduction, highlighting the need for biomarker-based thresholds[26]. The REGENERATE study investigated obeticholic acid in patients with MASLD and fibrosis, reporting improvements in NITs and fibrosis stage, suggesting NITs may serve as surrogates for biopsy in clinical trials[104]. A recent network meta-analysis compared novel agents and found that resmetirom was most effective in reducing liver steatosis, while pegozafermin, an FGF-21 analog, produced the most significant improvements in fibrosis[108]. These findings emphasize the promise of dual-acting therapies, but also underscore the need for long-term data to validate their safety and durability.

MASLD TREATMENT ACCESS

The global burden of MASLD is significant, with new pharmacotherapies offering promising options. However, these advances raise ethical and practical concerns related to cost-effectiveness, access, and equity, particularly in low-resource settings. The high cost of novel therapies, coupled with rapid price escalation, limits access for populations with limited healthcare resources or inadequate insurance coverage. MASLD disproportionately affects individuals with diabetes and obesity, conditions more prevalent in socioeconomically disadvantaged groups, making it harder for those most in need of treatment to access it. This exacerbates existing health disparities[145]. Ethical and policy challenges arise when expensive medications such as GLP-1 RAs become the standard of care without equitable frameworks for access. In many low-income and middle-income countries, these therapies are not included in national formularies or public insurance schemes, rendering them inaccessible to a large proportion of the population. This exclusion may worsen clinical outcomes and increase long-term healthcare costs associated with liver failure, transplantation, and cardiovascular events[146,147]. To address these disparities, health systems should consider implementing tiered access strategies based on clinical severity. Risk stratification tools such as FIB-4, ELF, or VCTE could be employed to prioritize high-risk MASLD patients for advanced therapies[148]. Moreover, integration of MASLD treatment into existing chronic disease programs - such as diabetes and cardiovascular disease management - can enhance cost-efficiency and improve outcomes[149].

Policymakers should also evaluate the inclusion of high-cost therapies like GLP-1 RAs into national essential medicines lists when sufficient evidence supports their long-term cost-effectiveness[150]. Price negotiations, public-private partnerships, and outcome-based reimbursement models may reduce acquisition costs and promote broader access[151]. Additionally, global health initiatives, such as pooled procurement mechanisms or support from international organizations (e.g., World Health Organization Essential Medicines), could facilitate access to these therapies in low-income and middle-income countries[152]. Research funding and clinical trial representation must also prioritize under-resourced populations to ensure generalizability of evidence and equitable innovation[153]. Ethically, equitable access to MASLD treatments is critical, as expensive therapies and advanced diagnostics create barriers for patients in low-resource settings. While non-invasive diagnostic options are emerging, their widespread use is delayed by the need for further validation and regulatory approval, which hinders their adoption in clinical practice and trials[145]. Cost-effectiveness analyses are crucial for informing policy, particularly in resource-constrained environments, suggesting that optimized screening and treatment strategies could enhance access and outcomes. However, these strategies must be explicitly tailored to local contexts to avoid deepening existing global health inequities. Addressing these issues requires multisectoral collaboration aimed at developing scalable, affordable, and culturally appropriate models of care for MASLD[149,152].

LIMITATIONS AND GAPS
Assessment of evidence quality using Grading of Recommendations Assessment, Development and Evaluation

Although this is a narrative review, the quality of the evidence was appraised using the principles of the Grading of Recommendations Assessment, Development and Evaluation (GRADE) framework to guide a structured qualitative analysis[154]. Studies on GLP-1 RAs (liraglutide, semaglutide) demonstrated moderate quality, primarily due to consistent results across studies[13,55]. However, certain limitations, such as small sample sizes and short follow-up durations, were noted. In contrast, studies on tirzepatide and retatrutide were rated as low and very low quality, respectively, owing to the absence of long-term data and uncertainty regarding their hepatic effects. Evidence for other treatments, such as naltrexone-bupropion and phentermine-topiramate, remains limited, resulting in a low rating overall[87,88,118].

Recommendations based on GRADE

In line with the GRADE assessment, recommendations for the use of GLP-1 RAs in MASLD are considered moderate, supported by their positive effects in reducing hepatic steatosis and fibrosis[154]. However, for tirzepatide and retatrutide, recommendations must remain cautious due to the lack of robust long-term safety data[121-123]. A significant limitation in the available evidence is the absence of randomized clinical trials specifically focused on MASLD for several anti-obesity treatments. Drugs like phentermine/topiramate and naltrexone/bupropion have proven effective in weight loss; however, studies specifically targeting MASLD are sparse[87,88,118]. Most existing research includes general populations of patients with obesity or diabetes, but few studies focus on the specific impact of these treatments on liver pathology associated with MASLD. This gap in targeted evidence limits the ability to draw firm conclusions regarding the use of these agents in this patient group.

Heterogeneity of MASLD and its implications

The heterogeneity of MASLD represents another challenge when evaluating treatment efficacy and safety. MASLD manifests diversely across individuals, with some patients affected by diabetes and others not, as well as variations in fibrosis severity (e.g., early vs advanced fibrosis). This variability directly influences treatment outcomes, as patients with advanced fibrosis or cirrhosis may respond differently compared to those in the earlier stages of the disease. Furthermore, patients with diabetes may experience differential responses due to IR and other metabolic factors, which impact the efficacy of treatment. This heterogeneity highlights the need for more personalized approaches and underscores the necessity of conducting clinical trials that focus on distinct patient subgroups[154].

Exclusion of cirrhosis patients in clinical trials

An essential aspect that limits the applicability of clinical trial results to real-world clinical practice is the systematic exclusion of patients with cirrhosis from many studies. Most clinical trials investigating treatments for MASLD do not include patients with compensated or decompensated cirrhosis, creating a significant gap in our understanding of how these treatments may affect this vulnerable population. Given that MASLD can progress to cirrhosis, the lack of data regarding drug use in this patient group restricts the generalization of results and hampers clinical decision-making for cirrhosis patients in everyday settings[141]. A significant limitation of the available evidence is the systematic exclusion of patients with advanced fibrosis or cirrhosis from most clinical trials, which significantly limits the external validity and applicability of findings to real-world populations. Additionally, many studies were of short duration, typically ranging from 12 weeks to 72 weeks, which impairs our ability to draw firm conclusions about long-term safety, durability of hepatic outcomes, and progression of fibrosis. While emerging agents such as tirzepatide and retatrutide have demonstrated substantial reductions in hepatic fat content measured by MRI-PDFF, data on histologic endpoints such as inflammation and fibrosis regression remain scarce. This gap is especially relevant for informing treatment decisions in patients with advanced disease stages. Furthermore, several of the included studies were sponsored by pharmaceutical companies, which may introduce reporting bias. Independent studies and longer-term follow-up are needed to validate these findings and better define the hepatic impact of these therapies across the MASLD spectrum.

CONCLUSION

Pharmacological treatments for MASLD, particularly GLP-1 RAs and emerging incretin-based therapies, have shown promising benefits in reducing hepatic steatosis and improving metabolic health, especially in early stages. However, their role in advanced or compensated cirrhosis remains uncertain due to limited data on safety and efficacy. Future research should prioritize long-term studies including real-world populations with metabolic comorbidities to define clinical outcomes better and guide personalized treatment. Current evidence is moderate for GLP-1 RAs, but remains low for newer agents like tirzepatide and retatrutide.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Peru

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade B, Grade B, Grade B, Grade C

Novelty: Grade A, Grade A, Grade B, Grade B, Grade B, Grade B

Creativity or Innovation: Grade B, Grade B, Grade B, Grade B, Grade B, Grade C

Scientific Significance: Grade A, Grade A, Grade B, Grade B, Grade B, Grade C

P-Reviewer: Gunes Y, Professor, Türkiye; Hassan AH, PhD, Assistant Professor, Egypt; He YH, PhD, China S-Editor: Zuo Q L-Editor: A P-Editor: Lei YY

References
1.  Abdelhameed F, Kite C, Lagojda L, Dallaway A, Chatha KK, Chaggar SS, Dalamaga M, Kassi E, Kyrou I, Randeva HS. Non-invasive Scores and Serum Biomarkers for Fatty Liver in the Era of Metabolic Dysfunction-associated Steatotic Liver Disease (MASLD): A Comprehensive Review From NAFLD to MAFLD and MASLD. Curr Obes Rep. 2024;13:510-531.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 50]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
2.  Wong RJ, Tran T, Kaufman H, Niles J, Gish R. Increasing metabolic co-morbidities are associated with higher risk of advanced fibrosis in nonalcoholic steatohepatitis. PLoS One. 2019;14:e0220612.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
3.  Mellemkjær A, Kjær MB, Haldrup D, Grønbæk H, Thomsen KL. Management of cardiovascular risk in patients with metabolic dysfunction-associated steatotic liver disease. Eur J Intern Med. 2024;122:28-34.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 40]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
4.  Canivet CM, Boursier J, Loomba R. New Nomenclature for Nonalcoholic Fatty Liver Disease: Understanding Metabolic Dysfunction-Associated Steatotic Liver Disease, Metabolic Dysfunction- and Alcohol-Associated Liver Disease, and Their Implications in Clinical Practice. Semin Liver Dis. 2024;44:35-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
5.  Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, Zelber-Sagi S, Wai-Sun Wong V, Dufour JF, Schattenberg JM, Kawaguchi T, Arrese M, Valenti L, Shiha G, Tiribelli C, Yki-Järvinen H, Fan JG, Grønbæk H, Yilmaz Y, Cortez-Pinto H, Oliveira CP, Bedossa P, Adams LA, Zheng MH, Fouad Y, Chan WK, Mendez-Sanchez N, Ahn SH, Castera L, Bugianesi E, Ratziu V, George J. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J Hepatol. 2020;73:202-209.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2883]  [Cited by in RCA: 2870]  [Article Influence: 574.0]  [Reference Citation Analysis (1)]
6.  Sanyal AJ, Harrison SA, Ratziu V, Abdelmalek MF, Diehl AM, Caldwell S, Shiffman ML, Aguilar Schall R, Jia C, McColgan B, Djedjos CS, McHutchison JG, Subramanian GM, Myers RP, Younossi Z, Muir AJ, Afdhal NH, Bosch J, Goodman Z. The Natural History of Advanced Fibrosis Due to Nonalcoholic Steatohepatitis: Data From the Simtuzumab Trials. Hepatology. 2019;70:1913-1927.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 147]  [Cited by in RCA: 279]  [Article Influence: 46.5]  [Reference Citation Analysis (0)]
7.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 932]  [Cited by in RCA: 1247]  [Article Influence: 124.7]  [Reference Citation Analysis (0)]
8.  Graupera I, Thiele M, Serra-Burriel M, Caballeria L, Roulot D, Wong GL, Fabrellas N, Guha IN, Arslanow A, Expósito C, Hernández R, Aithal GP, Galle PR, Pera G, Wong VW, Lammert F, Ginès P, Castera L, Krag A; Investigators of the LiverScreen Consortium. Low Accuracy of FIB-4 and NAFLD Fibrosis Scores for Screening for Liver Fibrosis in the Population. Clin Gastroenterol Hepatol. 2022;20:2567-2576.e6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 25]  [Cited by in RCA: 161]  [Article Influence: 53.7]  [Reference Citation Analysis (0)]
9.  Viganò M, Pugliese N, Cerini F, Turati F, Cimino V, Ridolfo S, Rocchetto S, Foglio F, Terrin M, La Vecchia C, Rumi MG, Aghemo A. Accuracy of FIB-4 to Detect Elevated Liver Stiffness Measurements in Patients with Non-Alcoholic Fatty Liver Disease: A Cross-Sectional Study in Referral Centers. Int J Mol Sci. 2022;23:12489.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
10.  Milani I, Codini M, Guarisco G, Chinucci M, Gaita C, Leonetti F, Capoccia D. Hepatokines and MASLD: The GLP1-Ras-FGF21-Fetuin-A Crosstalk as a Therapeutic Target. Int J Mol Sci. 2024;25:10795.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
11.  Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, George J, Bugianesi E. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol. 2018;15:11-20.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4054]  [Cited by in RCA: 3825]  [Article Influence: 546.4]  [Reference Citation Analysis (2)]
12.  Safarikova M, Kubena AA, Frankova V, Zima T, Kalousova M. The Effects of Different Storage Conditions and Repeated Freeze/Thaw Cycles on the Concentration, Purity and Integrity of Genomic DNA. Folia Biol (Praha). 2021;67:10-15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
13.  Cusi K, Isaacs S, Barb D, Basu R, Caprio S, Garvey WT, Kashyap S, Mechanick JI, Mouzaki M, Nadolsky K, Rinella ME, Vos MB, Younossi Z. American Association of Clinical Endocrinology Clinical Practice Guideline for the Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Primary Care and Endocrinology Clinical Settings: Co-Sponsored by the American Association for the Study of Liver Diseases (AASLD). Endocr Pract. 2022;28:528-562.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 365]  [Cited by in RCA: 582]  [Article Influence: 194.0]  [Reference Citation Analysis (1)]
14.  Lucero D, Miksztowicz V, Gualano G, Longo C, Landeira G, Álvarez E, Zago V, Brites F, Berg G, Fassio E, Schreier L. Nonalcoholic fatty liver disease associated with metabolic syndrome: Influence of liver fibrosis stages on characteristics of very low-density lipoproteins. Clin Chim Acta. 2017;473:1-8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 27]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
15.  Bansal SK, Bansal MB. Pathogenesis of MASLD and MASH - role of insulin resistance and lipotoxicity. Aliment Pharmacol Ther. 2024;59 Suppl 1:S10-S22.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 33]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
16.  Berdowska I, Matusiewicz M, Fecka I. A Comprehensive Review of Metabolic Dysfunction-Associated Steatotic Liver Disease: Its Mechanistic Development Focusing on Methylglyoxal and Counterbalancing Treatment Strategies. Int J Mol Sci. 2025;26:2394.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
17.  Kuchay MS, Choudhary NS, Ramos-Molina B. Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol. 2025;328:C1637-C1666.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
18.  Rolo AP, Teodoro JS, Palmeira CM. Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis. Free Radic Biol Med. 2012;52:59-69.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 636]  [Cited by in RCA: 722]  [Article Influence: 55.5]  [Reference Citation Analysis (1)]
19.  Nakamura T, Masuda A, Nakano D, Amano K, Sano T, Nakano M, Kawaguchi T. Pathogenic Mechanisms of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD)-Associated Hepatocellular Carcinoma. Cells. 2025;14:428.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
20.  Zhong H, Liu C, Huang Z, Tan P, Chen H, Fu W. Crosstalk between Hepatic Stellate Cells and Hepatic Macrophages in Metabolic Dysfunction-Associated Steatohepatitis. Am J Pathol. 2025;195:1040-1056.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
21.  Diehl AM, Day C. Cause, Pathogenesis, and Treatment of Nonalcoholic Steatohepatitis. N Engl J Med. 2017;377:2063-2072.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 671]  [Cited by in RCA: 922]  [Article Influence: 115.3]  [Reference Citation Analysis (0)]
22.  Devasia AG, Ramasamy A, Leo CH. Current Therapeutic Landscape for Metabolic Dysfunction-Associated Steatohepatitis. Int J Mol Sci. 2025;26:1778.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
23.  Bourebaba N, Marycz K. Hepatic stellate cells role in the course of metabolic disorders development - A molecular overview. Pharmacol Res. 2021;170:105739.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
24.  Vidal-Cevallos P, Sorroza-Martínez AP, Chávez-Tapia NC, Uribe M, Montalvo-Javé EE, Nuño-Lámbarri N. The Relationship between Pathogenesis and Possible Treatments for the MASLD-Cirrhosis Spectrum. Int J Mol Sci. 2024;25:4397.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
25.  Mladenić K, Lenartić M, Marinović S, Polić B, Wensveen FM. The "Domino effect" in MASLD: The inflammatory cascade of steatohepatitis. Eur J Immunol. 2024;54:e2149641.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 30]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
26.  Mejía-Guzmán JE, Belmont-Hernández RA, Chávez-Tapia NC, Uribe M, Nuño-Lámbarri N. Metabolic-Dysfunction-Associated Steatotic Liver Disease: Molecular Mechanisms, Clinical Implications, and Emerging Therapeutic Strategies. Int J Mol Sci. 2025;26:2959.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
27.  American Diabetes Association Professional Practice Committee. 5. Facilitating Positive Health Behaviors and Well-being to Improve Health Outcomes: Standards of Care in Diabetes-2025. Diabetes Care. 2025;48:S86-S127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 33]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
28.  Rinella ME, Neuschwander-Tetri BA, Siddiqui MS, Abdelmalek MF, Caldwell S, Barb D, Kleiner DE, Loomba R. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology. 2023;77:1797-1835.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1465]  [Cited by in RCA: 1202]  [Article Influence: 601.0]  [Reference Citation Analysis (1)]
29.  Li Q, Gao Y, Wang D, Zhu X, Jia J, Liu L, Tian H, Wang L, Zhang Y, Zhang D. Prospective Comparison of 2D-Shear Wave Elastography and Vibration-Controlled Transient Elastography in Assessing Liver Fibrosis in Metabolic Dysfunction-Associated Steatotic Liver Disease. Acad Radiol. 2025;S1076-6332(25)00309.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
30.  Bertot LC, Jeffrey GP, de Boer B, Wang Z, Huang Y, Garas G, MacQuillan G, Wallace M, Smith BW, Adams LA. Comparative Accuracy of Clinical Fibrosis Markers, Hepascore and Fibroscan® to Detect Advanced Fibrosis in Patients with Nonalcoholic Fatty Liver Disease. Dig Dis Sci. 2023;68:2757-2767.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
31.  Lin H, Lee HW, Yip TC, Tsochatzis E, Petta S, Bugianesi E, Yoneda M, Zheng MH, Hagström H, Boursier J, Calleja JL, Goh GB, Chan WK, Gallego-Durán R, Sanyal AJ, de Lédinghen V, Newsome PN, Fan JG, Castéra L, Lai M, Harrison SA, Fournier-Poizat C, Wong GL, Pennisi G, Armandi A, Nakajima A, Liu WY, Shang Y, de Saint-Loup M, Llop E, Teh KK, Lara-Romero C, Asgharpour A, Mahgoub S, Chan MS, Canivet CM, Romero-Gomez M, Kim SU, Wong VW; VCTE-Prognosis Study Group. Vibration-Controlled Transient Elastography Scores to Predict Liver-Related Events in Steatotic Liver Disease. JAMA. 2024;331:1287-1297.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 60]  [Cited by in RCA: 86]  [Article Influence: 86.0]  [Reference Citation Analysis (0)]
32.  Caussy C, Vergès B, Leleu D, Duvillard L, Subtil F, Abichou-Klich A, Hervieu V, Milot L, Ségrestin B, Bin S, Rouland A, Delaunay D, Morcel P, Hadjadj S, Primot C, Petit JM, Charrière S, Moulin P, Levrero M, Cariou B, Disse E. Screening for Metabolic Dysfunction-Associated Steatotic Liver Disease-Related Advanced Fibrosis in Diabetology: A Prospective Multicenter Study. Diabetes Care. 2025;48:877-886.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
33.  Wattacheril JJ, Abdelmalek MF, Lim JK, Sanyal AJ. AGA Clinical Practice Update on the Role of Noninvasive Biomarkers in the Evaluation and Management of Nonalcoholic Fatty Liver Disease: Expert Review. Gastroenterology. 2023;165:1080-1088.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 96]  [Cited by in RCA: 100]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
34.  Tapper EB, Parikh ND. Diagnosis and Management of Cirrhosis and Its Complications: A Review. JAMA. 2023;329:1589-1602.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 51]  [Cited by in RCA: 166]  [Article Influence: 83.0]  [Reference Citation Analysis (33)]
35.  Alkhouri N, Beyer C, Shumbayawonda E, Andersson A, Yale K, Rolph T, Chung RT, Vuppalanchi R, Cusi K, Loomba R, Pansini M, Dennis A. Decreases in cT1 and liver fat content reflect treatment-induced histological improvements in MASH. J Hepatol. 2025;82:438-445.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
36.  Sheka AC, Adeyi O, Thompson J, Hameed B, Crawford PA, Ikramuddin S. Nonalcoholic Steatohepatitis: A Review. JAMA. 2020;323:1175-1183.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 473]  [Cited by in RCA: 1007]  [Article Influence: 201.4]  [Reference Citation Analysis (0)]
37.  Pooler BD, Wiens CN, McMillan A, Artz NS, Schlein A, Covarrubias Y, Hooker J, Schwimmer JB, Funk LM, Campos GM, Greenberg JA, Jacobsen G, Horgan S, Wolfson T, Gamst AC, Sirlin CB, Reeder SB. Monitoring Fatty Liver Disease with MRI Following Bariatric Surgery: A Prospective, Dual-Center Study. Radiology. 2019;290:682-690.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 27]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
38.  Bredella MA, Ghomi RH, Thomas BJ, Torriani M, Brick DJ, Gerweck AV, Misra M, Klibanski A, Miller KK. Comparison of DXA and CT in the assessment of body composition in premenopausal women with obesity and anorexia nervosa. Obesity (Silver Spring). 2010;18:2227-2233.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 137]  [Cited by in RCA: 146]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
39.  Goyal NP, Xanthakos S, Schwimmer JB. Metabolic dysfunction-associated steatotic liver disease in children. Gut. 2025;74:669-677.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
40.  Munk Lauridsen M, Ravnskjaer K, Gluud LL, Sanyal AJ. Disease classification, diagnostic challenges, and evolving clinical trial design in MASLD. J Clin Invest. 2025;135:e189953.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
41.  Büyük M, Berker N, Bağbudar S, Çavuş B, Güllüoğlu M. Hepatic progenitor cell activation and ductular reaction in metabolic dysfunction-associated steatotic liver disease (MASLD): Indicators for disease activity and the degree of fibrosis: The pilot study. Medicine (Baltimore). 2025;104:e42108.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
42.  Liu QK. Mechanisms of action and therapeutic applications of GLP-1 and dual GIP/GLP-1 receptor agonists. Front Endocrinol (Lausanne). 2024;15:1431292.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 32]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
43.  Stefan N, Yki-Järvinen H, Neuschwander-Tetri BA. Metabolic dysfunction-associated steatotic liver disease: heterogeneous pathomechanisms and effectiveness of metabolism-based treatment. Lancet Diabetes Endocrinol. 2025;13:134-148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 41]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
44.  Drucker DJ, Habener JF, Holst JJ. Discovery, characterization, and clinical development of the glucagon-like peptides. J Clin Invest. 2017;127:4217-4227.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 170]  [Cited by in RCA: 296]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
45.  Ali SMJ, Lai M. Metabolic Dysfunction-Associated Steatotic Liver Disease. Ann Intern Med. 2025;178:ITC1-ITC16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
46.  Lotfy M, Singh J, Rashed H, Tariq S, Zilahi E, Adeghate E. Mechanism of the beneficial and protective effects of exenatide in diabetic rats. J Endocrinol. 2014;220:291-304.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 29]  [Cited by in RCA: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
47.  Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB; LEADER Steering Committee;  LEADER Trial Investigators. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2016;375:311-322.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4164]  [Cited by in RCA: 4955]  [Article Influence: 550.6]  [Reference Citation Analysis (0)]
48.  Havranek B, Loh R, Torre B, Redfield R, Halegoua-DeMarzio D. Glucagon-like peptide-1 receptor agonists improve metabolic dysfunction-associated steatotic liver disease outcomes. Sci Rep. 2025;15:4947.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
49.  Elsaid MI, Li N, Firkins SA, Rustgi VK, Paskett ED, Acharya C, Reddy KR, Chiang CW, Mumtaz K. Impacts of glucagon-like peptide-1 receptor agonists on the risk of adverse liver outcomes in patients with metabolic dysfunction-associated steatotic liver disease cirrhosis and type 2 diabetes. Aliment Pharmacol Ther. 2024;59:1096-1110.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 31]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
50.  Bea S, Ko HY, Bae JH, Cho YM, Chang Y, Ryu S, Byrne CD, Shin JY. Risk of hepatic events associated with use of sodium-glucose cotransporter-2 inhibitors versus glucagon-like peptide-1 receptor agonists, and thiazolidinediones among patients with metabolic dysfunction-associated steatotic liver disease. Gut. 2025;74:284-294.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 16]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
51.  Celsa C, Pennisi G, Tulone A, Ciancimino G, Vaccaro M, Infantino G, Di Maria G, Pinato DJ, Cabibbo G, Enea M, Mantovani A, Tilg H, Targher G, Cammà C, Petta S. Glucagon-like peptide-1 receptor agonist use is associated with a lower risk of major adverse liver-related outcomes: a meta-analysis of observational cohort studies. Gut. 2025;74:815-824.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
52.  Xiao G, Zhu S, Xiao X, Yan L, Yang J, Wu G. Comparison of laboratory tests, ultrasound, or magnetic resonance elastography to detect fibrosis in patients with nonalcoholic fatty liver disease: A meta-analysis. Hepatology. 2017;66:1486-1501.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 492]  [Cited by in RCA: 643]  [Article Influence: 80.4]  [Reference Citation Analysis (0)]
53.  Kannt A, Madsen AN, Kammermeier C, Elvert R, Klöckener T, Bossart M, Haack T, Evers A, Lorenz K, Hennerici W, Rocher C, Böcskei Z, Guillemot JC, Mikol V, Pattou F, Staels B, Wagner M. Incretin combination therapy for the treatment of non-alcoholic steatohepatitis. Diabetes Obes Metab. 2020;22:1328-1338.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
54.  Lee J, Hong SW, Chae SW, Kim DH, Choi JH, Bae JC, Park SE, Rhee EJ, Park CY, Oh KW, Park SW, Kim SW, Lee WY. Exendin-4 improves steatohepatitis by increasing Sirt1 expression in high-fat diet-induced obese C57BL/6J mice. PLoS One. 2012;7:e31394.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 97]  [Cited by in RCA: 122]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
55.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1100]  [Cited by in RCA: 1484]  [Article Influence: 164.9]  [Reference Citation Analysis (1)]
56.  Tsapas A, Avgerinos I, Karagiannis T, Malandris K, Manolopoulos A, Andreadis P, Liakos A, Matthews DR, Bekiari E. Comparative Effectiveness of Glucose-Lowering Drugs for Type 2 Diabetes: A Systematic Review and Network Meta-analysis. Ann Intern Med. 2020;173:278-286.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 99]  [Cited by in RCA: 214]  [Article Influence: 42.8]  [Reference Citation Analysis (0)]
57.  Newsome PN, Sanyal AJ, Engebretsen KA, Kliers I, Østergaard L, Vanni D, Bugianesi E, Rinella ME, Roden M, Ratziu V. Semaglutide 2.4 mg in Participants With Metabolic Dysfunction-Associated Steatohepatitis: Baseline Characteristics and Design of the Phase 3 ESSENCE Trial. Aliment Pharmacol Ther. 2024;60:1525-1533.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 49]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
58.  Grunvald E, Shah R, Hernaez R, Chandar AK, Pickett-Blakely O, Teigen LM, Harindhanavudhi T, Sultan S, Singh S, Davitkov P; AGA Clinical Guidelines Committee. AGA Clinical Practice Guideline on Pharmacological Interventions for Adults With Obesity. Gastroenterology. 2022;163:1198-1225.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 132]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
59.  Niu S, Chen S, Chen X, Ren Q, Yue L, Pan X, Zhao H, Li Z, Chen X. Semaglutide ameliorates metabolism and hepatic outcomes in an NAFLD mouse model. Front Endocrinol (Lausanne). 2022;13:1046130.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 34]  [Reference Citation Analysis (0)]
60.  Kanwal F, Kramer JR, Li L, Yang YX, Cao Y, Yu X, Samuel R, Ali B, Desiderio R, Cholankeril G, Bajaj M, El-Serag HB, Asch SM. GLP-1 Receptor Agonists and Risk for Cirrhosis and Related Complications in Patients With Metabolic Dysfunction-Associated Steatotic Liver Disease. JAMA Intern Med. 2024;184:1314-1323.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 59]  [Cited by in RCA: 38]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
61.  Mahgoub S, Newsome PN. Glucagon-Like Peptide-1 Receptor Agonists in Patients With Cirrhosis: Answering the Unanswered Questions? Clin Gastroenterol Hepatol. 2024;22:1186-1187.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
62.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 25]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
63.  Patel F, Gan A, Chang K, Vega KJ. Acute Pancreatitis in a Patient Taking Semaglutide. Cureus. 2023;15:e43773.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
64.  Campbell JE, Müller TD, Finan B, DiMarchi RD, Tschöp MH, D'Alessio DA. GIPR/GLP-1R dual agonist therapies for diabetes and weight loss-chemistry, physiology, and clinical applications. Cell Metab. 2023;35:1519-1529.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 36]  [Cited by in RCA: 68]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
65.  Folli F, Finzi G, Manfrini R, Galli A, Casiraghi F, Centofanti L, Berra C, Fiorina P, Davalli A, La Rosa S, Perego C, Higgins PB. Mechanisms of action of incretin receptor based dual- and tri-agonists in pancreatic islets. Am J Physiol Endocrinol Metab. 2023;325:E595-E609.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
66.  Mathiesen DS, Bagger JI, Bergmann NC, Lund A, Christensen MB, Vilsbøll T, Knop FK. The Effects of Dual GLP-1/GIP Receptor Agonism on Glucagon Secretion-A Review. Int J Mol Sci. 2019;20:4092.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 42]  [Cited by in RCA: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
67.  Frias JP, Bastyr EJ 3rd, Vignati L, Tschöp MH, Schmitt C, Owen K, Christensen RH, DiMarchi RD. The Sustained Effects of a Dual GIP/GLP-1 Receptor Agonist, NNC0090-2746, in Patients with Type 2 Diabetes. Cell Metab. 2017;26:343-352.e2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 192]  [Cited by in RCA: 250]  [Article Influence: 31.3]  [Reference Citation Analysis (0)]
68.  Al-Zamel N, Al-Sabah S, Luqmani Y, Adi L, Chacko S, Schneider TD, Krasel C. A Dual GLP-1/GIP Receptor Agonist Does Not Antagonize Glucagon at Its Receptor but May Act as a Biased Agonist at the GLP-1 Receptor. Int J Mol Sci. 2019;20:3532.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
69.  Zhyzhneuskaya SV, Al-Mrabeh AH, Peters C, Barnes AC, Hollingsworth KG, Welsh P, Sattar N, Lean MEJ, Taylor R. Clinical utility of liver function tests for resolution of metabolic dysfunction-associated steatotic liver disease after weight loss in the Diabetes Remission Clinical Trial. Diabet Med. 2025;42:e15462.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
70.  Regmi A, Aihara E, Christe ME, Varga G, Beyer TP, Ruan X, Beebe E, O'Farrell LS, Bellinger MA, Austin AK, Lin Y, Hu H, Konkol DL, Wojnicki S, Holland AK, Friedrich JL, Brown RA, Estelle AS, Badger HS, Gaidosh GS, Kooijman S, Rensen PCN, Coskun T, Thomas MK, Roell W. Tirzepatide modulates the regulation of adipocyte nutrient metabolism through long-acting activation of the GIP receptor. Cell Metab. 2024;36:1534-1549.e7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 36]  [Reference Citation Analysis (0)]
71.  Jastreboff AM, Kaplan LM, Frías JP, Wu Q, Du Y, Gurbuz S, Coskun T, Haupt A, Milicevic Z, Hartman ML; Retatrutide Phase 2 Obesity Trial Investigators. Triple-Hormone-Receptor Agonist Retatrutide for Obesity - A Phase 2 Trial. N Engl J Med. 2023;389:514-526.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 551]  [Cited by in RCA: 474]  [Article Influence: 237.0]  [Reference Citation Analysis (0)]
72.  Rosenstock J, Frias J, Jastreboff AM, Du Y, Lou J, Gurbuz S, Thomas MK, Hartman ML, Haupt A, Milicevic Z, Coskun T. Retatrutide, a GIP, GLP-1 and glucagon receptor agonist, for people with type 2 diabetes: a randomised, double-blind, placebo and active-controlled, parallel-group, phase 2 trial conducted in the USA. Lancet. 2023;402:529-544.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 209]  [Article Influence: 104.5]  [Reference Citation Analysis (0)]
73.  Sanyal AJ, Kaplan LM, Frias JP, Brouwers B, Wu Q, Thomas MK, Harris C, Schloot NC, Du Y, Mather KJ, Haupt A, Hartman ML. Triple hormone receptor agonist retatrutide for metabolic dysfunction-associated steatotic liver disease: a randomized phase 2a trial. Nat Med. 2024;30:2037-2048.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 84]  [Article Influence: 84.0]  [Reference Citation Analysis (0)]
74.  Kaur M, Misra S. A review of an investigational drug retatrutide, a novel triple agonist agent for the treatment of obesity. Eur J Clin Pharmacol. 2024;80:669-676.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 20]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
75.  Abdul-Rahman T, Roy P, Ahmed FK, Mueller-Gomez JL, Sarkar S, Garg N, Femi-Lawal VO, Wireko AA, Thaalibi HI, Hashmi MU, Dzebu AS, Banimusa SB, Sood A. The power of three: Retatrutide's role in modern obesity and diabetes therapy. Eur J Pharmacol. 2024;985:177095.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
76.  Sun B, Willard FS, Feng D, Alsina-Fernandez J, Chen Q, Vieth M, Ho JD, Showalter AD, Stutsman C, Ding L, Suter TM, Dunbar JD, Carpenter JW, Mohammed FA, Aihara E, Brown RA, Bueno AB, Emmerson PJ, Moyers JS, Kobilka TS, Coghlan MP, Kobilka BK, Sloop KW. Structural determinants of dual incretin receptor agonism by tirzepatide. Proc Natl Acad Sci U S A. 2022;119:e2116506119.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 66]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
77.  Forzano I, Varzideh F, Avvisato R, Jankauskas SS, Mone P, Santulli G. Tirzepatide: A Systematic Update. Int J Mol Sci. 2022;23:14631.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 51]  [Reference Citation Analysis (0)]
78.  Psaltis JP, Marathe JA, Nguyen MT, Le R, Bursill CA, Marathe CS, Nelson AJ, Psaltis PJ. Incretin-based therapies for the management of cardiometabolic disease in the clinic: Past, present, and future. Med Res Rev. 2025;45:29-65.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
79.  Perry RJ, Samuel VT, Petersen KF, Shulman GI. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature. 2014;510:84-91.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 733]  [Cited by in RCA: 868]  [Article Influence: 78.9]  [Reference Citation Analysis (0)]
80.  Karagiannis T, Avgerinos I, Liakos A, Del Prato S, Matthews DR, Tsapas A, Bekiari E. Management of type 2 diabetes with the dual GIP/GLP-1 receptor agonist tirzepatide: a systematic review and meta-analysis. Diabetologia. 2022;65:1251-1261.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 59]  [Cited by in RCA: 153]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
81.  Khan DA, Banerji A, Blumenthal KG, Phillips EJ, Solensky R, White AA, Bernstein JA, Chu DK, Ellis AK, Golden DBK, Greenhawt MJ, Horner CC, Ledford D, Lieberman JA, Oppenheimer J, Rank MA, Shaker MS, Stukus DR, Wallace D, Wang J; Chief Editor(s):, Khan DA, Golden DBK, Shaker M, Stukus DR;  Workgroup Contributors:, Khan DA, Banerji A, Blumenthal KG, Phillips EJ, Solensky R, White AA;  Joint Task Force on Practice Parameters Reviewers:, Bernstein JA, Chu DK, Ellis AK, Golden DBK, Greenhawt MJ, Horner CC, Ledford D, Lieberman JA, Oppenheimer J, Rank MA, Shaker MS, Stukus DR, Wallace D, Wang J. Drug allergy: A 2022 practice parameter update. J Allergy Clin Immunol. 2022;150:1333-1393.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 272]  [Article Influence: 90.7]  [Reference Citation Analysis (0)]
82.  le Roux CW, Zhang S, Aronne LJ, Kushner RF, Chao AM, Machineni S, Dunn J, Chigutsa FB, Ahmad NN, Bunck MC. Tirzepatide for the treatment of obesity: Rationale and design of the SURMOUNT clinical development program. Obesity (Silver Spring). 2023;31:96-110.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 53]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
83.  Gudzune KA, Stefanski A, Cao D, Mojdami D, Wang F, Ahmad N, Ling Poon J. Association between weight reduction achieved with tirzepatide and quality of life in adults with obesity: Results from the SURMOUNT-1 study. Diabetes Obes Metab. 2025;27:539-550.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 10]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
84.  Malhotra A, Bednarik J, Chakladar S, Dunn JP, Weaver T, Grunstein R, Fietze I, Redline S, Azarbarzin A, Sands SA, Schwab RJ, Bunck MC. Tirzepatide for the treatment of obstructive sleep apnea: Rationale, design, and sample baseline characteristics of the SURMOUNT -OSA phase 3 trial. Contemp Clin Trials. 2024;141:107516.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 32]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
85.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 52]  [Cited by in RCA: 293]  [Article Influence: 97.7]  [Reference Citation Analysis (0)]
86.  Cariou B, Linge J, Neeland IJ, Dahlqvist Leinhard O, Petersson M, Fernández Landó L, Bray R, Rodríguez Á. Effect of tirzepatide on body fat distribution pattern in people with type 2 diabetes. Diabetes Obes Metab. 2024;26:2446-2455.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
87.  Kim A, Nguyen J, Babaei M, Kim A, Geller DH, Vidmar AP. A Narrative Review: Phentermine and Topiramate for the Treatment of Pediatric Obesity. Adolesc Health Med Ther. 2023;14:125-140.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
88.  Hampl SE, Hassink SG, Skinner AC, Armstrong SC, Barlow SE, Bolling CF, Avila Edwards KC, Eneli I, Hamre R, Joseph MM, Lunsford D, Mendonca E, Michalsky MP, Mirza N, Ochoa ER, Sharifi M, Staiano AE, Weedn AE, Flinn SK, Lindros J, Okechukwu K. Clinical Practice Guideline for the Evaluation and Treatment of Children and Adolescents With Obesity. Pediatrics. 2023;151:e2022060640.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 139]  [Cited by in RCA: 489]  [Article Influence: 244.5]  [Reference Citation Analysis (0)]
89.  Bishay RH, Tonks KT, George J, Samocha-Bonet D, Meyerowitz-Katz G, Chisholm DJ, James DE, Greenfield JR. Plasma Bile Acids More Closely Align With Insulin Resistance, Visceral and Hepatic Adiposity Than Total Adiposity. J Clin Endocrinol Metab. 2021;106:e1131-e1139.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
90.  Pramanik S, Pal P, Ray S. Non-alcoholic fatty liver disease in type 2 diabetes: Emerging evidence of benefit of peroxisome proliferator-activated receptors agonists and incretin-based therapies. World J Methodol. 2024;14:91319.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (4)]
91.  Njei B, Al-Ajlouni Y, Lemos SY, Ugwendum D, Ameyaw P, Njei LP, Boateng S. Efficacy and Safety of GLP-1 Receptor Agonists in Patients With Metabolic Dysfunction-Associated Steatotic Liver Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Cureus. 2024;16:e71366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
92.  Mallet M, Silaghi CA, Sultanik P, Conti F, Rudler M, Ratziu V, Thabut D, Pais R. Current challenges and future perspectives in treating patients with NAFLD-related cirrhosis. Hepatology. 2024;80:1270-1290.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 16]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
93.  Tsankof A, Neokosmidis G, Koureta E, Veneti S, Cholongitas E, Tziomalos K. Which is the optimal antiobesity agent for patients with nonalcoholic fatty liver disease? Front Endocrinol (Lausanne). 2022;13:984041.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
94.  Younossi ZM, Paik JM, Stepanova M, Ong J, Alqahtani S, Henry L. Clinical profiles and mortality rates are similar for metabolic dysfunction-associated steatotic liver disease and non-alcoholic fatty liver disease. J Hepatol. 2024;80:694-701.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 139]  [Cited by in RCA: 153]  [Article Influence: 153.0]  [Reference Citation Analysis (0)]
95.  Son JW, Kim S. Comprehensive Review of Current and Upcoming Anti-Obesity Drugs. Diabetes Metab J. 2020;44:802-818.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 43]  [Cited by in RCA: 82]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
96.  Telci Caklili O, Cesur M, Mikhailidis DP, Rizzo M. Novel Anti-obesity Therapies and their Different Effects and Safety Profiles: A Critical Overview. Diabetes Metab Syndr Obes. 2023;16:1767-1774.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 19]  [Reference Citation Analysis (0)]
97.  Sanyal AJ, Friedman SL, McCullough AJ, Dimick-Santos L; American Association for the Study of Liver Diseases;  United States Food and Drug Administration. Challenges and opportunities in drug and biomarker development for nonalcoholic steatohepatitis: findings and recommendations from an American Association for the Study of Liver Diseases-U.S. Food and Drug Administration Joint Workshop. Hepatology. 2015;61:1392-1405.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 247]  [Cited by in RCA: 272]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
98.  TODAY Study Group. Longitudinal Association of Depressive Symptoms, Binge Eating, and Quality of Life With Cardiovascular Risk Factors in Young Adults With Youth-Onset Type 2 Diabetes: The TODAY2 Study. Diabetes Care. 2022;45:1073-1081.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
99.  Jastreboff AM, Ryan DH, Bays HE, Ebeling PR, Mackowski MG, Philipose N, Ross L, Liu Y, Burns CE, Abbasi SA, Pannacciulli N; MariTide Phase 2 Obesity Trial Investigators. Once-Monthly Maridebart Cafraglutide for the Treatment of Obesity - A Phase 2 Trial. N Engl J Med. 2025;.  [PubMed]  [DOI]  [Full Text]
100.  Zeng J, Fan JG, Francque SM. Therapeutic management of metabolic dysfunction associated steatotic liver disease. United European Gastroenterol J. 2024;12:177-186.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 34]  [Cited by in RCA: 50]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
101.  American Diabetes Association Professional Practice Committee. 4. Comprehensive Medical Evaluation and Assessment of Comorbidities: Standards of Care in Diabetes-2025. Diabetes Care. 2025;48:S59-S85.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 31]  [Reference Citation Analysis (0)]
102.  Armandi A, Bugianesi E. Dietary and pharmacological treatment in patients with metabolic-dysfunction associated steatotic liver disease. Eur J Intern Med. 2024;122:20-27.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 24]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
103.  Cusi K, Abdelmalek MF, Apovian CM, Balapattabi K, Bannuru RR, Barb D, Bardsley JK, Beverly EA, Corbin KD, ElSayed NA, Isaacs S, Kanwal F, Pekas EJ, Richardson CR, Roden M, Sanyal AJ, Shubrook JH, Younossi ZM, Bajaj M. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) in People With Diabetes: The Need for Screening and Early Intervention. A Consensus Report of the American Diabetes Association. Diabetes Care. 2025;48:1057-1082.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
104.  Tsochatzis E, Papatheodoridis GV, Manesis EK, Kafiri G, Tiniakos DG, Archimandritis AJ. Metabolic syndrome is associated with severe fibrosis in chronic viral hepatitis and non-alcoholic steatohepatitis. Aliment Pharmacol Ther. 2008;27:80-89.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 56]  [Cited by in RCA: 59]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
105.  Fabbrini E, Sullivan S, Klein S. Obesity and nonalcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology. 2010;51:679-689.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1303]  [Cited by in RCA: 1539]  [Article Influence: 102.6]  [Reference Citation Analysis (1)]
106.  Lee HA, Kim HY. Therapeutic Mechanisms and Clinical Effects of Glucagon-like Peptide 1 Receptor Agonists in Nonalcoholic Fatty Liver Disease. Int J Mol Sci. 2023;24:9324.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 23]  [Reference Citation Analysis (0)]
107.  Friedman SL, Ratziu V, Harrison SA, Abdelmalek MF, Aithal GP, Caballeria J, Francque S, Farrell G, Kowdley KV, Craxi A, Simon K, Fischer L, Melchor-Khan L, Vest J, Wiens BL, Vig P, Seyedkazemi S, Goodman Z, Wong VW, Loomba R, Tacke F, Sanyal A, Lefebvre E. A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology. 2018;67:1754-1767.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 428]  [Cited by in RCA: 535]  [Article Influence: 76.4]  [Reference Citation Analysis (0)]
108.  Raverdy V, Tavaglione F, Chatelain E, Lassailly G, De Vincentis A, Vespasiani-Gentilucci U, Qadri SF, Caiazzo R, Verkindt H, Saponaro C, Kerr-Conte J, Baud G, Marciniak C, Chetboun M, Oukhouya-Daoud N, Blanck S, Vandel J, Olsson L, Chakaroun R, Gnemmi V, Leteurtre E, Lefebvre P, Haas JT, Yki-Järvinen H, Francque S, Staels B, Le Roux CW, Tremaroli V, Mathurin P, Marot G, Romeo S, Pattou F. Data-driven cluster analysis identifies distinct types of metabolic dysfunction-associated steatotic liver disease. Nat Med. 2024;30:3624-3633.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 23]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
109.  Mouskeftara T, Kalopitas G, Liapikos T, Arvanitakis K, Theocharidou E, Germanidis G, Gika H. A Comprehensive Analysis of Liver Lipidomics Signature in Adults with Metabolic Dysfunction-Associated Steatohepatitis-A Pilot Study. Int J Mol Sci. 2024;25:13067.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
110.  Babu AF, Palomurto S, Kärjä V, Käkelä P, Lehtonen M, Hanhineva K, Pihlajamäki J, Männistö V. Metabolic signatures of metabolic dysfunction-associated steatotic liver disease in severely obese patients. Dig Liver Dis. 2024;56:2103-2110.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
111.  Chen VL, Kuppa A, Oliveri A, Chen Y, Ponnandy P, Patel PB, Palmer ND, Speliotes EK. Human genetics of metabolic dysfunction-associated steatotic liver disease: from variants to cause to precision treatment. J Clin Invest. 2025;135:e186424.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
112.  Jeong C, Han N, Jeon N, Rhee SJ, Staatz CE, Kim MS, Baek IH. Efficacy and Safety of Fibroblast Growth Factor-21 Analogs for the Treatment of Metabolic Dysfunction-Associated Steatohepatitis: A Systematic Review and Meta-Analysis. Clin Pharmacol Ther. 2024;116:72-81.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 13]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
113.  Harrison SA, Ruane PJ, Freilich B, Neff G, Patil R, Behling C, Hu C, Shringarpure R, de Temple B, Fong E, Tillman EJ, Rolph T, Cheng A, Yale K. A randomized, double-blind, placebo-controlled phase IIa trial of efruxifermin for patients with compensated NASH cirrhosis. JHEP Rep. 2023;5:100563.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 50]  [Reference Citation Analysis (0)]
114.  Puengel T, Tacke F. Efruxifermin, an investigational treatment for fibrotic or cirrhotic nonalcoholic steatohepatitis (NASH). Expert Opin Investig Drugs. 2023;32:451-461.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
115.  Noureddin M, Rinella ME, Chalasani NP, Neff GW, Lucas KJ, Rodriguez ME, Rudraraju M, Patil R, Behling C, Burch M, Chan DC, Tillman EJ, Zari A, de Temple B, Shringarpure R, Jain M, Rolph T, Cheng A, Yale K. Efruxifermin in Compensated Liver Cirrhosis Caused by MASH. N Engl J Med. 2025;392:2413-2424.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
116.  Loomba R, Sanyal AJ, Kowdley KV, Bhatt DL, Alkhouri N, Frias JP, Bedossa P, Harrison SA, Lazas D, Barish R, Gottwald MD, Feng S, Agollah GD, Hartsfield CL, Mansbach H, Margalit M, Abdelmalek MF. Randomized, Controlled Trial of the FGF21 Analogue Pegozafermin in NASH. N Engl J Med. 2023;389:998-1008.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 263]  [Cited by in RCA: 245]  [Article Influence: 122.5]  [Reference Citation Analysis (0)]
117.  Wang Y, Yu H, Cen Z, Zhu Y, Wu W. Drug targets regulate systemic metabolism and provide new horizons to treat nonalcoholic steatohepatitis. Metabol Open. 2024;21:100267.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
118.  le Roux CW, Fils-Aimé N, Camacho F, Gould E, Barakat M. The relationship between early weight loss and weight loss maintenance with naltrexone-bupropion therapy. EClinicalMedicine. 2022;49:101436.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
119.  Karakasis P, Patoulias D, Pamporis K, Stachteas P, Bougioukas KI, Klisic A, Fragakis N, Rizzo M. Safety and efficacy of the new, oral, small-molecule, GLP-1 receptor agonists orforglipron and danuglipron for the treatment of type 2 diabetes and obesity: systematic review and meta-analysis of randomized controlled trials. Metabolism. 2023;149:155710.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 35]  [Cited by in RCA: 32]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
120.  Saxena AR, Frias JP, Brown LS, Gorman DN, Vasas S, Tsamandouras N, Birnbaum MJ. Efficacy and Safety of Oral Small Molecule Glucagon-Like Peptide 1 Receptor Agonist Danuglipron for Glycemic Control Among Patients With Type 2 Diabetes: A Randomized Clinical Trial. JAMA Netw Open. 2023;6:e2314493.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 84]  [Reference Citation Analysis (0)]
121.  Buckeridge C, Cobain S, Bays HE, Matsuoka O, Fukushima Y, Halstead P, Tsamandouras N, Sherry N, Gorman DN, Saxena AR. Efficacy and safety of danuglipron (PF-06882961) in adults with obesity: A randomized, placebo-controlled, dose-ranging phase 2b study. Diabetes Obes Metab. 2025;27:4915-4926.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
122.  Pratt E, Ma X, Liu R, Robins D, Coskun T, Sloop KW, Haupt A, Benson C. Orforglipron (LY3502970), a novel, oral non-peptide glucagon-like peptide-1 receptor agonist: A Phase 1b, multicentre, blinded, placebo-controlled, randomized, multiple-ascending-dose study in people with type 2 diabetes. Diabetes Obes Metab. 2023;25:2642-2649.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
123.  Melson E, Ashraf U, Papamargaritis D, Davies MJ. What is the pipeline for future medications for obesity? Int J Obes (Lond). 2025;49:433-451.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 87]  [Cited by in RCA: 99]  [Article Influence: 99.0]  [Reference Citation Analysis (0)]
124.  Lin RT, Sun QM, Xin X, Ng CH, Valenti L, Hu YY, Zheng MH, Feng Q. Comparative efficacy of THR-β agonists, FGF-21 analogues, GLP-1R agonists, GLP-1-based polyagonists, and Pan-PPAR agonists for MASLD: A systematic review and network meta-analysis. Metabolism. 2024;161:156043.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 16]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
125.  Giannakogeorgou A, Roden M. Role of lifestyle and glucagon-like peptide-1 receptor agonists for weight loss in obesity, type 2 diabetes and steatotic liver diseases. Aliment Pharmacol Ther. 2024;59 Suppl 1:S52-S75.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
126.  Davies MJ, Bajaj HS, Broholm C, Eliasen A, Garvey WT, le Roux CW, Lingvay I, Lyndgaard CB, Rosenstock J, Pedersen SD; REDEFINE 2 Study Group. Cagrilintide–Semaglutide in Adults with Overweight or Obesity and Type 2 Diabetes. N Engl J Med. 2025;393:648-659.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
127.  Garvey WT, Blüher M, Osorto Contreras CK, Davies MJ, Winning Lehmann E, Pietiläinen KH, Rubino D, Sbraccia P, Wadden T, Zeuthen N, Wilding JPH; REDEFINE 1 Study Group. Coadministered Cagrilintide and Semaglutide in Adults with Overweight or Obesity. N Engl J Med. 2025;393:635-647.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
128.  Bailey CJ, Flatt PR, Conlon JM. Multifunctional incretin peptides in therapies for type 2 diabetes, obesity and associated co-morbidities. Peptides. 2025;187:171380.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
129.  Kuo CC, Chuang MH, Li CH, Tsai YW, Huang PY, Kuo HT, Lai CC. Glucagon-Like Peptide-1 Receptor Agonists and Liver Outcomes in Patients With MASLD and Type 2 Diabetes. Aliment Pharmacol Ther. 2025;61:1163-1174.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 12]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
130.  Le Roux C, Mondoh A. Treatment of obesity with medications binding the glucagon-like peptide 1 receptor: what is the current state of play? Expert Opin Pharmacother. 2024;25:131-138.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
131.  Dutta D, Nagendra L, Harish BG, Sharma M, Joshi A, Hathur B, Kamrul-Hasan A. Efficacy and Safety of Cagrilintide Alone and in Combination with Semaglutide (Cagrisema) as Anti-Obesity Medications: A Systematic Review and Meta-Analysis. Indian J Endocrinol Metab. 2024;28:436-444.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
132.  Çetin E, Pedersen B, Burak MF. Paradigm shift in obesity treatment: an extensive review of current pipeline agents. Turk J Med Sci. 2025;55:1-16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
133.  Goedeke L, Shulman GI. Therapeutic potential of mitochondrial uncouplers for the treatment of metabolic associated fatty liver disease and NASH. Mol Metab. 2021;46:101178.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 70]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
134.  Vilariño-García T, Polonio-González ML, Pérez-Pérez A, Ribalta J, Arrieta F, Aguilar M, Obaya JC, Gimeno-Orna JA, Iglesias P, Navarro J, Durán S, Pedro-Botet J, Sánchez-Margalet V. Role of Leptin in Obesity, Cardiovascular Disease, and Type 2 Diabetes. Int J Mol Sci. 2024;25:2338.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 64]  [Reference Citation Analysis (0)]
135.  Metz M, Beghini M, Wolf P, Pfleger L, Hackl M, Bastian M, Freudenthaler A, Harreiter J, Zeyda M, Baumgartner-Parzer S, Marculescu R, Marella N, Hannich JT, Györi G, Berlakovich G, Roden M, Krebs M, Risti R, Lõokene A, Trauner M, Kautzky-Willer A, Krššák M, Stangl H, Fürnsinn C, Scherer T. Leptin increases hepatic triglyceride export via a vagal mechanism in humans. Cell Metab. 2022;34:1719-1731.e5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 38]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
136.  Pathak P, Liu H, Boehme S, Xie C, Krausz KW, Gonzalez F, Chiang JYL. Farnesoid X receptor induces Takeda G-protein receptor 5 cross-talk to regulate bile acid synthesis and hepatic metabolism. J Biol Chem. 2017;292:11055-11069.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 131]  [Cited by in RCA: 183]  [Article Influence: 22.9]  [Reference Citation Analysis (0)]
137.  Chen R, Petrazzini BO, Duffy Á, Rocheleau G, Jordan D, Bansal M, Do R. Trans-ancestral rare variant association study with machine learning-based phenotyping for metabolic dysfunction-associated steatotic liver disease. Genome Biol. 2025;26:50.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
138.  Ministerial Meeting on Population of the Non-Aligned Movement (1993: Bali). Non-aligned movement support for South-South collaboration in the field of population and family planning. Integration. 1994;37-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 198]  [Cited by in RCA: 199]  [Article Influence: 49.8]  [Reference Citation Analysis (0)]
139.  Chen VL, Morgan TR, Rotman Y, Patton HM, Cusi K, Kanwal F, Kim WR. Resmetirom therapy for metabolic dysfunction-associated steatotic liver disease: October 2024 updates to AASLD Practice Guidance. Hepatology. 2025;81:312-320.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 21]  [Cited by in RCA: 39]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
140.  Zhong J, Cai Z, Zhu G, Zhang J. Comparative efficacy and safety of pharmacologic therapies for metabolic dysfunction-associated steatotic liver disease over 24 weeks in reducing liver steatosis and fibrosis: A network meta-analysis. Diabetes Obes Metab. 2025;27:3309-3323.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
141.  Watt KD, Paul S, Khan MQ, Siddiqui M, Lam J, Diwan TS, Camilleri M. Obesity management for the hepatologist-What to do, how to do it and why? Hepatology. 2025;81:1607-1620.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
142.  Dulai PS, Singh S, Patel J, Soni M, Prokop LJ, Younossi Z, Sebastiani G, Ekstedt M, Hagstrom H, Nasr P, Stal P, Wong VW, Kechagias S, Hultcrantz R, Loomba R. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology. 2017;65:1557-1565.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 984]  [Cited by in RCA: 1436]  [Article Influence: 179.5]  [Reference Citation Analysis (0)]
143.  American Diabetes Association Professional Practice Committee. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Care in Diabetes-2025. Diabetes Care. 2025;48:S181-S206.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 95]  [Reference Citation Analysis (0)]
144.  Newsome PN, Ambery P. Incretins (GLP-1 receptor agonists and dual/triple agonists) and the liver. J Hepatol. 2023;79:1557-1565.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 104]  [Cited by in RCA: 95]  [Article Influence: 47.5]  [Reference Citation Analysis (0)]
145.  Le P, Tatar M, Dasarathy S, Alkhouri N, Herman WH, Taksler GB, Deshpande A, Ye W, Adekunle OA, McCullough A, Rothberg MB. Estimated Burden of Metabolic Dysfunction-Associated Steatotic Liver Disease in US Adults, 2020 to 2050. JAMA Netw Open. 2025;8:e2454707.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 40]  [Cited by in RCA: 35]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
146.  Theilade S, Christensen MB, Vilsbøll T, Knop FK. An overview of obesity mechanisms in humans: Endocrine regulation of food intake, eating behaviour and common determinants of body weight. Diabetes Obes Metab. 2021;23 Suppl 1:17-35.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
147.  Atun R, Knaul FM, Akachi Y, Frenk J. Innovative financing for health: what is truly innovative? Lancet. 2012;380:2044-2049.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 64]  [Cited by in RCA: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
148.  European Association for the Study of the Liver. EASL Clinical Practice Guidelines on non-invasive tests for evaluation of liver disease severity and prognosis - 2021 update. J Hepatol. 2021;75:659-689.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 993]  [Cited by in RCA: 1089]  [Article Influence: 272.3]  [Reference Citation Analysis (0)]
149.  Nugent R. Chronic diseases in developing countries: health and economic burdens. Ann N Y Acad Sci. 2008;1136:70-79.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 135]  [Cited by in RCA: 156]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
150.  Waldrop SW, Johnson VR, Stanford FC. Inequalities in the provision of GLP-1 receptor agonists for the treatment of obesity. Nat Med. 2024;30:22-25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 18]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
151.  Moon S, Jambert E, Childs M, von Schoen-Angerer T. A win-win solution?: A critical analysis of tiered pricing to improve access to medicines in developing countries. Global Health. 2011;7:39.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 87]  [Cited by in RCA: 81]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
152.  Ewen M, Zweekhorst M, Regeer B, Laing R. Baseline assessment of WHO's target for both availability and affordability of essential medicines to treat non-communicable diseases. PLoS One. 2017;12:e0171284.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 71]  [Cited by in RCA: 103]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
153.  Emanuel EJ, Wendler D, Killen J, Grady C. What makes clinical research in developing countries ethical? The benchmarks of ethical research. J Infect Dis. 2004;189:930-937.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 503]  [Cited by in RCA: 511]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
154.  Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, Schünemann HJ; GRADE Working Group. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008;336:924-926.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11058]  [Cited by in RCA: 15043]  [Article Influence: 884.9]  [Reference Citation Analysis (0)]