Clin Mol Hepatol > Volume 31(3); 2025 > Article
Kim and Rinella: Emerging therapies and real-world application of metabolic dysfunction-associated steatotic liver disease treatment

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease, formerly referred to as non-alcoholic fatty liver disease, is the most common liver disease in Western countries and has emerged as the leading indication for liver transplantation. Metabolic dysfunction-associated steatohepatitis (MASH), a more advanced stage, carries a high risk of progression to liver fibrosis, cirrhosis, liver failure, and hepatocellular carcinoma. Until recently, lifestyle intervention remained the mainstay of MASH management, with no pharmacological treatments specifically approved. However, advances in understanding its pathophysiological mechanisms have fueled numerous clinical trials, culminating in the Food and Drug Administration’s (FDA) approval of resmetirom as the first treatment for MASH in 2024. Additionally, many investigational drugs are nearing FDA approval or progressing through late-stage clinical trials. This review examines the current therapeutic landscape, highlights strategies for identifying patients suitable for liver-directed therapies in real-world settings, and discusses the challenges that remain.

INTRODUCTION

Metabolic dysfunction-associated steatotic liver disease (MASLD), previously referred to as non-alcoholic fatty liver disease (NAFLD), is the most prevalent liver disease in the USA [1], and has an estimated global prevalence of 30% [2,3]. Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as non-alcoholic steatohepatitis (NASH), represents a more severe form of MASLD and is the leading indication for liver transplantation among women and individuals over 65 years old [4]. MASLD is linked to other metabolic conditions such as obesity, type 2 diabetes mellitus (T2DM), hypertension, and dyslipidemia, and it poses a substantial burden on both patient health and worldwide healthcare systems [5]. Moreover, the growing prevalence of MASLD imposes significant clinical, economic, and societal challenges [6]. Until recently, given the lack of approved therapies, therapeutic strategies have primarily focused on lifestyle modifications and optimization of comorbidities. While lifestyle interventions can be effective, they are challenging to maintain, which limits their overall impact [7]. Therefore, as fibrosis improvement is crucial to bend the arc of disease progression, and sustained weight loss through lifestyle intervention is achieved by only a minority, pharmacological therapies are needed to meaningfully impact liver-related outcomes. After decades of research, the Food and Drug Administration (FDA) approved the first treatment for MASH in March 2024, resmetirom, a thyroid hormone receptor-β (THR-β) agonist [8]. Several drug candidates are currently in the pipeline to enrich the armamentarium of treatment for MASH. In this review, we will focus on the identification of the appropriate target population and outline the current treatment landscape for drugs in development.

PATHOPHYSIOLOGY OF MASLD

The pathogenesis of MASLD is multifactorial, encompassing genetic predisposition, insulin resistance, impaired lipid and carbohydrate metabolism, oxidative stress, inflammation, and apoptosis [9]. Environmental factors, such as diet and physical inactivity, primarily increase the risk of hepatic steatosis. Excessive caloric intake beyond metabolic demand results in adipose tissue fat overload, promoting inflammation and insulin resistance in adipose tissue [10]. Insulin resistance impairs lipolysis regulation in adipose tissue, resulting in an increased hepatic fatty acid influx [11]. Positive energy balance also triggers hepatic de novo lipogenesis, resulting in carbohydrate-derived hepatic lipid accumulation [12]. Hepatic fatty acids undergo either mitochondrial β-oxidation or triglyceride synthesis via re-esterification. Overwhelmed fatty acid disposal mechanisms lead to lipotoxic species formation, triggering endoplasmic reticulum stress, oxidative stress, mitochondrial dysfunction, and inflammasome activation, ultimately driving the MASH phenotype characterized by hepatocellular injury, inflammation, and fibrosis [9]. Moreover, dysbiosis of the gut microbiota compromises intestinal barrier integrity and increases permeability, facilitating the translocation of lipopolysaccharides and pro-inflammatory factors into the portal circulation, thereby promoting hepatic inflammation and injury. Furthermore, dysbiosis disrupts bile acid metabolism, exacerbating hepatic inflammation and oxidative stress [13]. Additionally, genetic factors, such as variations in the PNPLA3, TM6SF2, and HSD167B13 genes, have been identified as key risk factors that increase susceptibility to MASLD and progression to advanced liver disease and hepatocellular carcinoma [14]. These factors collectively serve as crucial risk factors, or “multiple hits”, that trigger the onset of hepatic steatosis and drive its progression to inflammation and fibrosis [9], while the sustained low-grade systemic inflammation promotes cardiovascular disease [15] and tumor development [16]. The aforementioned mechanisms serve as potential targets for pharmacological intervention (Fig. 1). Approaches targeting insulin sensitivity, free fatty acid metabolism, mitochondrial function, inflammation, fibrosis, and gut-liver axis signaling hold therapeutic potential.

LIFESTYLE MODIFICATION

Lifestyle modifications, particularly weight reduction through dietary adjustments and increased physical activity, remain the foundational strategy in the management of MASLD [17] and have been the only available options for several years prior to the approval of the first pharmacological therapy, resmetirom, in 2024. Even small amounts of weight loss (<5%) can improve liver enzymes though more substantial weight loss is needed to resolve steatohepatitis (approximately 7%) and >10% is needed to improve fibrosis [18]. While several studies have supported this concept, the impact of weight loss varies between individuals and can be impacted by many factors, including genetic polymorphisms [19,20]. Unfortunately, the sustainability of weight loss in patients is limited due to the challenge of lifestyle modification maintenance throughout the years [7]. While bariatric surgery has been shown to induce MASH regres-sion, attenuate hepatic fibrosis progression, and promote sustained weight reduction [21], its applicability is restricted to a subset of patients meeting specific criteria (body mass index ≥40 kg/m2 regardless of metabolic comorbidities or a body mass index ≥35 kg/m2 with metabolic comorbidities). Given these limitations, pharmacological treatments remain indispensable for the effective management of MASLD population.

PHARMACOLOGICAL AGENTS

Pharmacological agents should be tailored according to individual patient profile and may include liver directed therapy (resmetirom) and/or therapies to control metabolic comorbidities, such as T2DM, dyslipidemia, and obesity [22].

Resmetirom – the only drug conditionally approved by the FDA for MASH

Resmetirom, an oral, once-daily, liver-targeted THR-β selective agonist, is the first and currently the only drug to have received, on 14 March 2024, accelerated FDA approval in the USA for treating non-cirrhotic MASH with moderate-to-advanced liver fibrosis (corresponding to F2–F3 fibrosis stages) [8]. THR-β is predominantly expressed in the liver and plays a key role in regulating hepatic lipid metabolism through inducing fatty acid oxidation, mitophagy, lipol-ysis, and cholesterol clearance in the liver [23]. In the interim analysis of the registration trial, MAESTRO-NASH, which included 966 patients with biopsy proven MASH and significant fibrosis (F2–F3), both doses of resmetirom demonstrated statistical significance on the dual primary surrogate histological endpoints after 52 weeks of treatment [24]. Fibrosis improvement (at least 1 stage) with no worsening of NAFLD activity score (NAS) was achieved in 24% and 26% of patients in resmetirom 80 mg and 100 mg groups, respectively, versus 14% in the placebo group (P<0.001). MASH resolution with no worsening of fibrosis was achieved in a significantly higher proportion in the resmetirom 80 mg and 100 mg groups (26% and 30%, respectively) compared to the placebo group (10%, P<0.001). Resmetirom also met its key secondary endpoint and showed a significant decrease in low-density lipoprotein cholesterol in the resmetirom 80 mg and 100 mg groups (–14% and −16%, respectively) compared to a slight increase in the placebo group (+0.1%). Resmetirom was also associated with significant reduction in the hepatic fat fraction (magnetic resonance imaging-derived proton density fat fraction [MRI-PDFF]) with a relative change from baseline of −35% and −47% in patients treated by resmetirom 80 mg and 100 mg, respectively, versus −9% in the placebo group. The reduction in hepatic fat fraction was predictive of histological response as 88% of patients with fibrosis improvement and 96% of patients with MASH resolution had a relative reduction of at least 30% on MRI-PDFF. Resmetirom demonstrated an acceptable safety/tolerability profile with minimal impact on central and peripheral thyroid hormones. Serious adverse events were rare and comparable across all treatment groups, with the most common adverse events being mild and transient nausea, vomiting, and diarrhea. MAESTRONASH, together with additional phase 3 trials, are still ongoing to assess the progression to major adverse liver outcomes in MASH patients with and without cirrhosis. Resmetirom was granted conditional approval by the FDA under the accelerated approval pathway, permitting its market availability while its long-term clinical efficacy is being evaluated in ongoing trials. The final approval remains dependent on the demonstration of clinical benefit in these studies, and approval may be rescinded if the trials fail to meet their predefined endpoints.

Glucagon-like peptide-1 receptor agonists and dual agonists – FDA-approved drugs for obesity and/or T2DM

Semaglutide

Glucagon-like peptide-1 receptor agonists (GLP-1 Ras) promote insulin release from pancreatic β cells after food ingestion. They also delay gastric emptying, reduce appetite, and contribute to a decrease in hepatic steatosis [25]. Semaglutide, a GLP-1 RA approved by the FDA for treating T2DM and/or obesity, has been shown to promote weight loss and improve glycemic control in individuals with these conditions [26]. Since insulin resistance is a common feature of both T2DM and obesity, and a key driver in the pathogenesis of MASH, semaglutide has been studied as a treatment option for MASH. In November 2024, semaglutide met its primary endpoint in the ongoing phase 3 registrational trial, ESSENCE, which enrolled 1,200 patients with biopsy-proven MASH and significant fibrosis (F2–F3) [27]. At 72 weeks, an interim analysis of the first 800 randomized participants with biopsy results demonstrated that a significantly higher proportion of patients achieved at least 1 stage improvement in fibrosis with no worsening of MASH in the once-weekly subcutaneous semaglutide 2.4 mg group (37%) compared to the placebo group (23%, P<0.0001). MASH resolution with no worsening of fibrosis was achieved in 63% of patients treated with semaglutide versus 34% in the placebo group (P<0.0001). Semaglutide had a significant weight loss benefit with a relative weight decrease of 11% in the semaglutide group versus 2% in the placebo group. As previously shown, semaglutide was also associated with significant benefit for glucose and lipid control. In addition to the benefits observed in this MASH registration trial, the cardiovascular efficacy of semaglutide has been shown in outcomes trials conducted in the T2DM population [28] The safety/tolerability profile of this drug candidate has been widely investigated in the context of a high cardiovascular risk profile population and has been demonstrated to be safe and well tolerated. Common adverse events are gastrointestinal disorders such as nausea, diarrhea, constipation, and vomiting.
A recent study using a USA claims database demonstrated that MASLD cirrhosis patients with T2DM who received GLP-1 RAs had a reduced risk of long-term major adverse liver outcomes compared to those not receiving GLP-1 RAs [29]. Similarly, a Swedish registry suggested that GLP-1 RAs reduce the risk of major adverse liver outcomes in patients with chronic liver disease and T2DM [30]. The ongoing ESSENCE trial will further characterize the potential benefit of semaglutide to prevent major adverse liver outcomes in MASH patients with F2–F3 fibrosis.

Other GLP-1-based therapies

Several additional drug candidates are in development, such as dual GLP-1/glucose-dependent insulinotropic polypeptide (GIP)-RAs (tirzepatide [31]) or dual GLP-1/glucagon RAs (survodutide [32]) and have shown promising phase 2b results (Table 1). GIP, another incretin regulating postprandial glucose and lipid metabolism, can further stimulate insulin secretion, reduce food intake via central nervous system modulation, and enhance perfusion in subcutaneous white adipose tissue, thereby improving postprandial triglyceride clearance and increasing insulin sensitivity [33]. Given that tirzepatide, a dual GIP and GLP-1 RA recently approved for T2DM, has demonstrated superior glycemic control and weight loss compared to a GLP-1 RA [34], its ability to reduce hepatic fat and visceral adipose tissue further underscores its potential advantage in treating MASH over GLP-1 RAs [35]. Though the role of GIP agonism in MASH therapy, independent of body weight loss, is not well understood, a phase 2b trial involving participants with MASH and moderate to severe fibrosis showed that once-weekly subcutaneous tirzepatide administered for 52 weeks was significantly more effective than placebo in achieving MASH resolution without worsening of fibrosis (44% with tirzepatide 5 mg, 56% with tirzepatide 10 mg, 62% with tirzepatide 15 mg vs. 10% with placebo; P<0.001 for all) [31]. Glucagon receptor activation reduces hepatic glycogen levels, increases gluconeogenesis, decreases hepatic lipogenesis, and boosts mitochondrial turnover and oxidative function in the liver, thereby increasing energy expenditure [36]. In phase 2b trials, once-weekly subcutaneous injections of survodutide, a dual GLP-1/glucagon RA, for 48 weeks were superior to placebo in improving MASH without worsening of fibrosis and in reducing hepatic fat content. The safety profile was generally tolerable, with gastrointestinal side effects commonly observed, similar to other GLP-1 RAs [32].

Pharmacotherapies with positive phase 2b data

Several additional drug candidates have completed their phase 2b and have either already started their phase 3 program or are pending phase 3 initiation. Among this potential armamentarium of drugs, the subcutaneous fibroblast growth factor 21 (FGF21) analogs, including efruxifermin [37] and pegozafermin [38], appear appealing. FGF21 directly inhibits de novo lipogenesis and lipotoxicity, enhances fatty acid oxidation, and reduces inflammation and fibrosis in the liver. Moreover, FGF21 stimulates adiponectin production in adipocytes, potentially augmenting its metabolic, anti-inflammatory, and antifibrotic properties [39]. Efruxifermin, a long-acting Fc-FGF21 fusion protein with a half-life of approximately 3 days, improved fibrosis without worsening of MASH in 30% and 49% of patients receiving 28 mg and 50 mg, respectively, compared to 19% in the placebo group, according to a recent analysis of sequential liver biopsy data from the 96-week phase 2b HARMONY trial. MASH resolution without fibrosis worsening was achieved in 40% and 37% of patients in the 28 mg and 50 mg efruxifermin groups, respectively, versus 19% in the placebo group [40]. In the 96-week phase 2b SYMMETRY trial involving patients with compensated cirrhosis (F4), 39% of patients in the efruxifermin 50mg group (P=0.009) demonstrated significant improvement in fibrosis by ≥1 stage without worsening of MASH at week 96, compared to 29% in the efruxifermin 28 mg group and 15% in the placebo group [41]. Pegozafermin, a long-acting glyco-pegylated recombinant analogue of FGF21, significantly improved fibrosis and MASH resolution in patients with stage 2–3 fibrosis, along with a notable reduction in liver fat content in a phase 2b trial. It also enhanced insulin sensitivity and provided benefits for lipid metabolism. While nonclinical and early clinical studies have suggested potential bone effects with FGF21 analogues, this trial detected no evidence of reduced bone density or fractures; however, longer-term studies are warranted [38]. Efimosfermin alfa (BOS-580), a long-acting, once-monthly Fc-FGF21 fusion protein, demonstrated encouraging outcomes in a 24-week study. It improved fibrosis without worsening of MASH (45% vs. 21%, P=0.038) and resolved MASH without worsening of fibrosis (68% vs. 29%, P=0.002). Beyond these histological improvements, efimosfermin significantly improved fibrosis biomarkers, reduced non-invasive markers of hepatic injury and hepatic fat, and enhanced glycemic control. The study also confirmed its favorable tolerability profile, with adverse events mostly limited to mild to moderate gastrointestinal symptoms [42].
Peroxisome proliferator-activated receptors (PPARs), a subfamily of nuclear hormone receptors, act as ligand-activated transcription factors and include three isoforms: PPARα, PPARβ/δ, and PPARγ. PPARα, mainly expressed in the liver, regulates hepatic fatty acid metabolism, plasma lipoprotein levels, and FGF21 secretion, while PPARβ/δ, found in skeletal muscle and adipose tissue, influences fatty acid oxidation and insulin sensitivity. PPARγ, predominantly in adipose tissue, plays a key role in regulating lipid metabolism, insulin resistance, immune function, and adipocyte differentiation [43,44]. Lanifibranor, an oral pan-PPAR agonist, demonstrated superior efficacy in achieving MASH resolution without fibrosis worsening (49% vs. 22%, P<0.01) and improving fibrosis by at least one stage without MASH worsening (48% vs. 29%, P<0.05), despite potential safety concerns, including weight gain, hemoglobin decrease, bone fractures, and vesical cancer [45].
Denifanstat (TVB-02640), an oral fatty acid synthase inhibitor, prevents steatosis by blocking de novo lipogenesis in hepatocytes, reduces inflammation by inhibiting immune cell activation, and prevents fibrosis by reducing de novo lipogenesis in stellate cells, thereby blocking stellate cell activation [46]. The 52-week phase 2b trial of once-daily oral denifanstat demonstrated statistically significant and clinically meaningful benefits, including resolution of MASH without fibrosis progression (38% vs. 16%, P<0.01) and improvement in fibrosis without worsening of MASH (41% vs. 18%, P<0.01), as confirmed by paired liver biopsy results [47]. PXL065, a novel oral agent that retains the efficacy of pioglitazone while lacking PPARγ activity, inhibits mitochondrial pyruvate carrier and long-chain acyl-CoA synthetase 4. In a phase 2b trial, it demonstrated fibrosis improvement by ≥1 stage without worsening of MASH following 36 weeks of treatment (38% [7.5 mg], 50% [15 mg, P=0.06], and 31% [22.5 mg] vs. 17% for placebo) [48]. Icosabutate, an oral, structurally enhanced fatty acid that targets free-fatty acid receptor 1 and 4, which are highly expressed in Kupffer cells, demonstrated histological improvements along with enhanced insulin sensitivity and lipid-related benefits in a phase 2b trial, despite not meeting its primary endpoint (Table 1) [49].
Careful consideration is required when interpreting the results of clinical trials. First, participants in MASH trials are typically non-cirrhotic MASH patients, either F1–F3 or F2–F3, with a NAS of ≥4. These criteria have led to significant variability in MASH activity and fibrosis stages among enrolled patients [50]. The impact of disease heterogeneity on the response to MASH therapies remains unclear, but drug efficacy may vary across individuals and disease stages. Second, histological surrogate endpoints have been established in clinical trials for MASH because the slow progression of the disease makes long-term observation of liver-related clinical outcomes impractical. For accelerated FDA approval, at least one of the following histologic endpoints must be met: (1) MASH resolution without worsening of liver fibrosis, or (2) fibrosis improvement without worsening of MASH [51]. However, MASH activity alone has limited prognostic value for clinical outcomes [52]. Moreover, liver biopsy is subject to sampling variability, which limits comprehensive liver assessment and may lead to inaccurate evaluations of treatment efficacy [53]. Additionally, inter- and intraobserver inconsistencies further complicate the reliable assessment of drug efficacy in MASH trials [54].

NON-INVASIVE IDENTIFICATION OF MASH PATIENTS WITH F2–F3 FIBROSIS

The introduction of approved MASH therapy is considerably transforming the clinical management of MASLD patients and should bring additional real-world evidence on this patient population, especially on the non-invasive diagnosis and follow-up of patients. Although pivotal clinical trials for resmetirom relied on histologic diagnosis, liver biopsy is not practical for routine clinical use [55]. Fortunately, the current FDA label does not mandate liver biopsy as a diagnostic criterion. However, careful attention is required to ensure the accurate selection of the target population. Based on the baseline characteristics of patients included in the phase 3 registration trials of resmetirom, an expert panel has published their recommendations for patient selection and follow-up [56]. After excluding alternate diagnoses through comprehensive history-taking and additional testing, which may occasionally require a liver biopsy, non-invasive biomarkers can effectively identify the population eligible for resmetirom treatment. To select presumed MASH, steatosis assessment using vibration-controlled transient elastography (VCTE)-controlled attenuation parameter (CAP) score (≥280 dB/m) or MRI-PDFF (≥5%), along with elevated aspartate aminotransferase (AST) (>17 IU/L for females and >20 IU/L for males), is recommended as an initial screening approach. As outlined in Figure 2, MASH F2–F3 patients may present with a liver stiffness measurement (LSM) of 10–15 kPa via VCTE or 3.3–4.2 kPa via magnetic resonance elastography (MRE). The threshold recommended by the expert panel is based on the interquartile range from the MAESTRO-NASH trial data. To avoid overtreatment in patients at the lower end of the spectrum, the panel set a lower threshold of 10 kPa for VCTE, considering the reduced specificity of VCTE values in individuals with obesity.
More recently, the American Association for the Study of Liver Diseases (AASLD) released guidance for clinicians on the use of resmetirom, including updated non-invasive criteria for patient selection [57]. The AASLD guidance suggests broader eligibility criteria, recommending LSM values of 8–15 kPa by VCTE or 3.1–4.4 kPa by MRE. This expansion reflects findings from the MAESTRO-NASH trial, in which approximately half of the participants had LSM by VCTE outside the interquartile range of 10–15 kPa. Ultimately, the assessment of a patient’s fibrosis status remains at the discretion of the prescribing clinician, who must integrate both expert opinion and AASLD guidance to make an informed treatment decision.
Higher liver stiffness suggests more advanced fibrosis, and such patients should be approached with caution or excluded. As a single non-invasive test (NIT) is insufficient, the expert panel suggests performing several NITs and considering the entire patient profile before making any decisions. It is of paramount importance to exclude patients with potential cirrhosis, which may be indicated by several lab parameters and/or signs of portal hypertension.
Laboratory safety parameters should be assessed after three months of treatment, then NITs may confirm improvements after 6–12 months of treatment. The application of NITs in the assessment of treatment response is an emerging area of interest. Substantial evidence has established the association between improvements in NITs and changes in fibrosis stage in MASH [58]. A systematic review and meta-analysis of participants from NASH clinical trials identified the combination of MRI-PDFF and alanine aminotransferase (ALT) response as a predictor of histological response [59]. In addition, a decline in VCTE stiffness has been suggested to correlate with histologic response [60]. In the MAESTRO-NASH trial, a reduction of at least 20% in liver stiffness, measured by MRE, was associated with improvements in histologic fibrosis, while a ≥30% decrease in hepatic steatosis, measured by MRI-PDFF, was linked to more favorable histologic outcomes compared to placebo. In contrast, ALT, CAP, and VCTE demonstrated weaker correlations with histopathological responses [61]. However, more robust data are needed to establish NITs as reliable surrogates for histologic improvement following MASH pharmacotherapy.
From a clinical perspective, it is essential not only to stratify patients by fibrosis severity and MASH activity but also to identify those most vulnerable to disease-related adverse clinical outcomes. Substantial additional evidence is being developed, testifying to a collective effort to link NITs to major adverse liver outcomes. Recently, Lin and colleagues [62] demonstrated the high accuracy of VCTE-based scores in predicting major adverse liver outcomes in clinical practice, based on data from over 16,000 patients. Future analyses of data from MASH clinical trials may offer further insights into the role of NITs in treatment monitoring and predicting clinical outcomes.

CHALLENGES IN THE APPLICATION TO REAL CLINICAL PRACTICE

While the introduction of new therapeutic drugs for MASLD represents progress, it also comes with challenges. Firstly, a recent consensus process involving multiple scientific societies has established new terminology, replacing NAFLD with MASLD [2]. Nevertheless, the differential definitions of NAFLD and MASLD could potentially lead to uncertainty in clinical applications. From the perspective of incorporating the new MASLD nomenclature criteria into existing clinical data from patients with biopsy-proven NAFLD, the results remain relevant, as reflected in the strong concordance between traditional NAFLD and the newly defined MASLD [63,64]. However, the dynamic nature of metabolic risk factors and alcohol intake must be considered as they influence the definition of MASLD. Further research is needed to extrapolate clinical trial results to current MASLD patients. On the other hand, MASLD diagnosis is based on affirmative criteria rather than exclusion, with relatively simple and widely used parameters defining MASLD, facilitating earlier identification of at-risk patients [2]. Moreover, refining diagnostic criteria and patient classification will be beneficial for achieving favorable clinical trial outcomes and tailoring therapeutic approaches. Additionally, the newly introduced definition of metabolic alcohol-associated liver disease (MetALD) represents an overlap of MASLD with significant alcohol use and was frequently excluded from clinical trials under the previous NAFLD definition. Given the lack of evidence on dedicated treatment approaches and interventions for this patient group, careful consideration is needed when applying MASLD-based clinical trial results to MetALD patients. To establish evidence-based treatment strategies, specialized clinical trials focusing on the MetALD population are urgently needed.
Second, many standard NITs focus on the detection of advanced fibrosis and exhibit suboptimal performance with an area under the receiver operating characteristic curve <0.8 in identifying at-risk MASH [65]. Diagnosing at-risk MASH is challenging as it requires assessing hepatic steatosis, fibrosis, and inflammation. The most promising NITs for this purpose include FibroScan AST score, MRI-AST score, MRE combined with fibrosis-4 index, and MRI using iron-corrected T1, with further optimization ongoing [58]. However, these NITs also lack mechanistic biomarkers that account for biological variations among patients and the wide range of MASLD phenotypes. Therefore, circulating microRNAs (miRs) and proteomics-based biomarkers are increasingly recognized as promising candidates for clinical application. Recent evidence suggests that specific miRs exhibit significant alterations in the sera of patients with atrisk MASH and hold promise as early biomarkers for diagnosing and predicting MASLD progression [66]. The NIS4® and its modified version, NIS2+TM, comprise serum biomarkers, including miR-34a, demonstrating good performance in identifying at-risk MASH [67]. In addition, a proteomics-based signature has been identified for detecting at-risk MASH and monitoring therapy-induced histological changes [68]. Ongoing research in this field presents promising opportunities for advancing personalized treatment approaches and refining diagnostic tools.
Third, the economic impact of MASH therapies requires thorough consideration as these treatments become integrated into clinical practice. Although resmetirom may introduce additional financial burdens for patients, including drug costs, it also has the potential to slow or even reverse disease progression. The progression of MASH is a significant driver of healthcare costs, with expenses rising exponentially as the disease advances [69]. In a simulated cohort with an average life expectancy of about 21 years, a medical intervention for MASLD was considered cost-effective in reducing liver fibrosis and mortality only if the annual drug cost remained below $12,000 [70]. Another early economic evaluation suggested that resmetirom could be cost-effective for treating MASH and fibrosis compared to a placebo, assuming a willingness-to-pay threshold of US $100,000 per quality-adjusted life year [71]. A recent budget impact analysis from the perspective of USA private insurers assessed the financial implications of incorporating resmetirom for adults with non-cirrhotic MASH and moderate-to-advanced liver fibrosis [72]. While its inclusion in the formulary led to a moderate budget increase over three years, primarily driven by drug costs, it also contributed to reducing expenses related to disease progression. However, the limited time frame constrains the evaluation of long-term benefits, such as cost savings from slowed disease progression and the impact of future treatment competition. To fully evaluate the financial and clinical sustainability of resmetirom, long-term cost-effectiveness analyses are necessary.
Early trials suggest that many MASH therapies not only improve liver health but also provide metabolic and cardiovascular advantages, since metabolic dysfunction is fundamentally linked to the pathophysiology of MASH. Given that cardiac disease continues to be the primary cause of mortality in individuals with MASLD, treatments that enhance cardiovascular disease profiles are of particular interest. However, the cost and accessibility of these therapies will play a crucial role in their widespread adoption. Given its high cost, several strategies could improve the affordability and accessibility of resmetirom, similar to approaches used to lower the prices of human immunodeficiency virus and hepatitis C virus medicines [73]. Pricing strategies include tiered pricing models (implementing region-specific pricing based on economic status), value-based pricing (pricing based on real-world effectiveness in improving liver outcomes), and subscription-based models (implementing a flat-rate pricing model). Additional approaches include expanding insurance coverage, patient assistance programs, voluntary licensing agreements for generic production, and research into alternative dosing regimens to reduce costs while maintaining efficacy.

FUTURE DIRECTIONS

The therapeutic efficacy of monotherapy remains suboptimal, as evidenced by clinical investigations demonstrating that approximately 60% of subjects exhibit insufficient treatment response or fail to achieve predetermined primary endpoints [74]. Considering the sophisticated pathophysiology of MASH involving various contributing pathways and the complex cross-talk among target molecules within different signaling networks, combination therapy is likely to become the standard of care in this disease, similar to T2DM and hypertension. Combination approaches offer potential advantages, such as boosting biological potency, enhancing clinical efficacy, and optimizing side effect profiles by targeting converging pathways, which can strengthen biological responses, counter compensatory mechanisms, and address both upstream and downstream pathogenic drivers [75]. Including treatment regimens with verified extrahepatic clinical benefits, such as significant weight loss, achieved with GLP-1 RAs [27], adds an additional advantage to combination therapy, as weight loss may further enhance therapeutic response. Achieving an optimal combination therapy may necessitate the integration of two or more pharmacotherapies with complementary mechanisms of action. Several potential therapeutic combination options are under investigation [76]. However, substantial challenges remain in developing combination approaches, as early clinical trial data have been negative. The efficacy of each drug in combination therapy has yet to be fully established. Identifying two optimal synergistic pathways among various interconnected mechanisms, navigating complex regulatory requirements, determining optimal dosing, managing an increased risk of side effects, and enrolling a large number of patients all present significant clinical and translational hurdles [77].
Among potential combinations, adding GLP-1 RAs to THR-β agonists appears physiologically promising, as it may enhance antifibrotic effects through additional metabolic benefits such as weight loss and improved glucose control. Adding an FGF21 agonist to resmetirom could increase glucose uptake by adipose tissue while simultaneously preventing its release through lipolysis. Given the potent effects of FGF21 and the limitation of its subcutaneous administration, an induction regimen with FGF21 followed by long-term oral resmetirom therapy may offer a viable treatment strategy [78]. In real-world applications of combination therapy, GLP-1 RAs will serve as the backbone, as they are already widely prescribed for T2DM and obesity, which are common in patients with MASH [77]. The combination of FGF21 analogs and GLP-1 RA presents an intriguing possibility by potentiating the insulin-sensitizing effect of FGF21 [78] with the insulin-secretion effect of GLP-1 [25]. which would likely improve glycemic and lipid control. The combined administration of efruxifermin (an FGF21 analogue) and GLP-1 RA demonstrated superior efficacy to GLP-1 RA monotherapy in patients with MASH and T2DM. This dual therapy significantly reduced hepatic fat content and enhanced multiple clinical parameters, including markers of liver injury, fibrosis, glucose metabolism, and lipid profiles, while maintaining a safety and tolerability profile comparable to GLP-1 RA alone [79]. A phase 2 trial combining semaglutide (GLP-1 RA) with firsocostat (acetyl-CoA carboxylase inhibitor) and/or cilofexor (farnesoid X receptor agonist) successfully achieved its primary endpoints, demonstrating greater relative liver fat reduction, improvement in liver biochemistry and non-invasive fibrosis markers, and a favorable safety profile [80]. Large-scale, late-phase clinical trials with long-term follow-up are needed to validate these promising findings. Further mechanistic studies are essential to understand the drug-drug interactions and potential synergistic effects before this therapeutic approach can be incorporated into clinical practice. It remains unclear whether combination therapy should be used during the initial induction phase, followed by monotherapy for maintenance, or reserved only for patients who do not respond to monotherapy.
Despite the recent development of new drugs, a significant proportion of patients have not responded to therapy, with response rates of only 20% to 30% in clinical trials demonstrating MASH resolution and fibrosis regression [24,45,81]. Considering that MASLD is collectively influenced by genetic, metabolic, and environmental factors, contributing to its heterogeneity [9], precision medicine is gaining interest in optimizing individual patient care. Genomic and metabolomic technologies provide insights into individual patient variations, allowing for more targeted therapeutic approaches based on predicted treatment responses. To select the most effective combination therapy, it is crucial to adopt a personalized strategy that includes non-invasive testing, metabolic evaluations, and genetic analysis to tailor treatment to the patient’s unique characteristics [82].

CONCLUSION

Given the global burden of MASLD and MASH, along with their associated morbidity and mortality, these conditions remain a substantial and growing public health concern. The establishment of targeted pharmacological therapies is crucial for addressing the ongoing health challenge and improving outcomes for affected individuals. Resmetirom is the first FDA-approved therapy targeting MASH, showing effectiveness in reversing liver fibrosis and steatohepatitis, in addition to lipid benefits. Incretin-based therapies, such as GLP-1 and dual receptor agonists, also show promise for addressing MASH with fibrosis. Several agents in late-phase 2 or phase 3 trials appear promising but still require additional validation. Using NITs with high discriminatory ability to identify patients with at-risk MASH and significant fibrosis can help select those most likely to benefit from MASH pharmacotherapy, and guide treatment response monitoring. In the near future, MASH treatment is anticipated to undergo a paradigm shift, likely involving combination therapies and precision medicine that specifically target the primary drivers of each patient’s condition.

FOOTNOTES

Authors’ contribution
HY Kim and M. E. Rinella were responsible for conception, design, analysis, interpretation, and drafting of the manuscript. All authors approved the final version of the manuscript.
Conflicts of Interest
M. E. Rinella has served as a scientific consultant for Akero, 89Bio, Boehringer Ingelheim, Intercept Pharmaceuticals, Histoindex, Madrigal, NGM Biopharmaceuticals, Novo Nordisk, Eli Lilly, Sagimet Biosciences, Sonic Incytes, Cytodyn, and GSK. Additionally, M E. Rinella is a member of the scientific executive boards of Akero, Madrigal, and Novo Nordisk.

Figure 1.
Pathogenesis of metabolic dysfunction-associated steatotic liver disease and potential targets of drugs. Excessive caloric intake exceeding metabolic needs leads to fat accumulation in adipose tissue, which promotes inflammation and insulin resistance. Insulin resistance disrupts the regulation of lipolysis in adipose tissue, leading to an increased influx of fatty acids into the liver. A positive energy balance further stimulates hepatic de novo lipogenesis, causing the accumulation of lipids derived from carbohydrates in the liver. Overburdened fatty acid disposal pathways contribute to the formation of lipotoxic species in the liver, ultimately promoting hepatic inflammation and fibrosis. Genetic factors increase susceptibility to MASLD and its progression to advanced liver disease. Additionally, gut microbiota dysbiosis promotes the translocation of lipopolysaccharides and pro-inflammatory factors into the portal circulation and disrupts bile acid metabolism, ultimately driving hepatic inflammation and injury. ACC, acetyl-CoA carboxylase; ACLS4, acyl-CoA synthetase long chain family member 4; DGAT2, diacylglycerol acyltransferase 2; FFA, free fatty acid; FGF21, fibroblast growth factor 21; ER, endoplasmic reticulum; FASN, fatty acid synthase; FXR, farnesoid X receptor; GC, glucagon; GIP, glucose-dependent insulinotropic polypeptide; GLP1, Glucagon-like peptide-1; LPS, lipopolysaccharide; MASLD, metabolic dysfunction-associated steatotic liver disease; MPC, mitochondrial pyruvate carrier; PPAR, peroxisome proliferator-activated receptor; TG, triglyceride; THR-β, thyroid hormone receptor-β; SEFA, structurally enhanced fatty acid.

cmh-2025-0083f1.jpg
Figure 2.
Suggested algorithm for patient selection and monitoring for resmetirom utilizing non-invasive fibrosis tests. ALT, alanine aminotransferase; DILI, drug-induced liver injury; ELF, Enhanced liver fibrosis; LSM, liver stiffness measurement; MASLD, metabolic dysfunction-associated steatotic liver disease; MRE, magnetic resonance elastography; MRI-PDFF, magnetic resonance imaging-derived proton density fat fraction; NIT, non-invasive test; PHTN, portal hypertension; VCTE, vibration-controlled transient elastography. Adapted from the article of Noureddin et al. (Clin Gastroenterol Hepatol 2024;22:2367-2377) [56].

cmh-2025-0083f2.jpg
Table 1.
Drugs in phase 3 development or with phase 2b results
Drug name/therapeutic class Trial phase Treatment duration/administration route/dosage Histology endpoints results Additional benefits Safety/tolerability
Efruxifermin [40] 2b 96 wk Intention-to-treat results Lipid control Well tolerated
FGF 21 analog Subcutaneous MASH resolution w/o worsening of fibrosis Glucose control Mild gastrointestinal disorders
Weekly 28 mg, 50 mg - Placebo: 19% (n=43) Potential bone metabolism safety concern
- 28 mg QW: 40% (n=40), P<0.05
- 50 mg QW: 37% (n=43), P<0.05
Fibrosis improvement w/o worsening of MASH
- Placebo: 19% (n=43)
- 28 mg QW: 30% (n=40)
- 50 mg QW: 49% (n=43), P<0.01
Pegozafermin [38] 2b 24 wk Intention-to-treat results Lipid control Well tolerated
FGF21 analog Subcutaneous MASH resolution w/o worsening of fibrosis Glucose control Mild gastrointestinal disorders
Weekly 15 mg, Weekly 30 mg, Biweekly 44 mg - Placebo: 2% (n=61) Potential bone metabolism safety concern
- 15 mg QW: 37% (n=14), P<0.001
- 30 mg QW: 23% (n=66), P<0.001
- 44 mg Q2W: 26% (n=51), P<0.001
Fibrosis improvement w/o worsening of MASH
- Placebo: 7% (n=61)
- 15 mg QW: 22% (n=14)
- 30 mg QW: 26% (n=66), P<0.01
- 44 mg Q2W: 27% (n=51), P<0.01
Efimosfermin [42] 2b 24 wk Paired biopsy results Lipid control Well tolerated
FGF21 analog Subcutaneous MASH resolution w/o worsening of fibrosis Glucose control Mild gastrointestinal disorders
Monthly 300 mg - Placebo: 29% (n=34) Potential bone metabolism safety concern
- 300 mg Q4W: 68% (n=31), P<0.01
Fibrosis improvement w/o worsening of MASH
- Placebo: 21% (n=34)
- 300 mg Q4W: 45% (n=31), P<0.05
Lanifibranor [45] 2b 24 wk Intention-to-treat results Lipid control Well tolerated
Pan-PPAR agonist Oral MASH resolution w/o worsening of fibrosis Glucose control Mild gastrointestinal disorders
Daily 800 mg, 1,200 mg - Placebo: 22% (n=81) Weight gain and fluid retention
- 800 mg: 39% (n=83), P<0.05 Anemia
- 1,200 mg: 49% (n=83), P<0.01 Vesical cancer?
Fibrosis improvement w/o worsening of MASH
- Placebo: 29% (n=81)
- 800 mg: 34% (n=83)
- 1,200 mg: 48% (n=83), P<0.05
Tirzepatide [31] 2b 52 wk Subcutaneous Intention-to-treat results Lipid control Well tolerated
Dual GLP-1/GIP receptor agonist Weekly 5 mg, 10 mg, 15 mg MASH resolution w/o worsening of fibrosis Glucose control Mild gastrointestinal disorders
- Placebo: 10% (n=48) Weight loss
- 5 mg: 44% (n=47), P<0.001
- 10 mg: 56% (n=47), P<0.001
- 15 mg: 62% (n=48), P<0.001
Fibrosis improvement w/o worsening of MASH
- Placebo: 30% (n=48)
- 5 mg: 55% (n=47)
- 10 mg: 51% (n=47)
- 15 mg: 51% (n=48)
Survodutide [32] 2b 48 wk Intention-to-treat results (post-hoc analysis in F2–F3 patients) Lipid control Well tolerated
Dual GLP-1/glucagon receptor agonist Subcutaneous MASH resolution w/o worsening of fibrosis Glucose control Mild gastrointestinal disorders
Weekly 2.4 mg, 4.8 mg, 6 mg - Placebo: 10% (n=60) Weight loss
- 2.4 mg: 45% (n=53), P<0.001
- 4.8 mg: 46% (n=59), P<0.001
- 6.0 mg: 45% (n=51), P<0.001
Fibrosis improvement w/o worsening of MASH
- Placebo: 22% (n=60)
- 2.4 mg: 38% (n=53)
- 4.8 mg: 34% (n=59)
- 6.0 mg: 43% (n=51)
Denifanstat [47] 2b 52 wk Paired biopsy results Lipid benefit Well tolerated
Fatty acid synthase inhibitor Oral MASH resolution w/o worsening of fibrosis Hair loss
Daily 50 mg - Placebo: 16% (n=45)
- 50 mg: 38% (n=81), P<0.01
Fibrosis improvement w/o worsening of MASH
- Placebo: 18% (n=45)
- 50 mg: 41% (n=81), P<0.01
PXL065 [48] 2b 36 wk Paired biopsy results Glucose control Well tolerated
Deuterium-stabilized R-pioglitazone Oral MASH resolution w/o worsening of fibrosis Mild gastrointestinal disorders
Daily 7.5 mg, 15 mg, 22.5 mg - Placebo: 30% (n=23) Weight gain and fluid retention
- 7.5 mg: 38% (n=21) Anemia
- 15 mg: 36% (n=22)
- 22.5 mg: 31% (n=26)
Fibrosis improvement w/o worsening of MASH
- Placebo: 17% (n=23)
- 7.5 mg: 38% (n=21)
- 15 mg: 50% (n=22)
- 22.5 mg: 31% (n=26)
Icosabutate [49] 2b 52 wk Paired biopsy results Lipid control Well tolerated
Free-fatty acid receptor 1 and 4 agonist Oral MASH resolution w/o worsening of fibrosis Glucose control
Daily 300 mg, 600 mg - Placebo: 14% (n=59)
- 300 mg: 19% (n=57)
- 600 mg: 26% (n=62)
Fibrosis improvement w/o worsening of MASH
- Placebo: 12% (n=59)
- 300 mg: 26% (n=57)
- 600 mg: 23% (n=62)

FGF21, fibroblast growth factor 21; GIP, glucose-dependent insulinotropic polypeptide; GLP-1, Glucagon-like peptide-1; MASH, metabolic dysfunction-associated steatohepatitis; PPAR, peroxisome proliferator-activated receptor; QW, once weekly; Q2W, every two weeks; Q4W, every four weeks; w/o, without.

Abbreviations

AASLD
American Association for the Study of Liver Diseases
ALT
alanine aminotransferase
AST
aspartate aminotransferase
CAP
controlled attenuation parameter
FDA
Food and Drug Administration
FGF21
fibroblast growth factor 21
GIP
glucose-dependent insulinotropic polypeptide
GLP-1 RA
glucagon-like peptide-1 receptor agonists
LSM
liver stiffness measurement
MASH
metabolic dysfunction-associated steatohepatitis
MASLD
metabolic dysfunction-associated steatotic liver disease
MetALD
metabolic alcohol-associated liver disease
miRs
microRNAs
MRE
magnetic resonance elastography
MRI-PDFF
magnetic resonance imaging-derived proton density fat fraction
NAFLD
non-alcoholic fatty liver disease
NAS
NAFLD activity score
NASH
non-alcoholic steatohepatitis
NIT
non-invasive test
PPAR
Peroxisome proliferator-activated receptor
T2DM
type 2 diabetes mellitus
THR-β
thyroid hormone receptor-β
VCTE
vibration-controlled transient elastography

REFERENCES

1. Williams CD, Stengel J, Asike MI, Torres DM, Shaw J, Contreras M, et al. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: a prospective study. Gastroenterology 2011;140:124-131.
pmid
2. Rinella ME, Lazarus JV, Ratziu V, Francque SM, Sanyal AJ, Kanwal F, et al.; NAFLD Nomenclature consensus group. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Hepatology 2023;78:1966-1986.
pmid
3. Younossi ZM, Golabi P, Paik JM, Henry A, Van Dongen C, et al. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology 2023;77:1335-1347.
crossref pmid
4. Noureddin M, Vipani A, Bresee C, Todo T, Kim IK, Alkhouri N, et al. NASH leading cause of liver transplant in women: updated analysis of indications for liver transplant and ethnic and gender variances. Am J Gastroenterol 2018;113:1649-1659.
crossref pmid pmc pdf
5. Kim CH, Younossi ZM. Nonalcoholic fatty liver disease: a manifestation of the metabolic syndrome. Cleve Clin J Med 2008;75:721-728.
crossref pmid
6. Perumpail BJ, Khan MA, Yoo ER, Cholankeril G, Kim D, Ahmed A. Clinical epidemiology and disease burden of nonalcoholic fatty liver disease. World J Gastroenterol 2017;23:8263-8276.
crossref pmid pmc
7. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol 2024;81:492-542.
crossref pmid pdf
8. U.S. Food and Drug Administration (FDA). FDA Approves first treatment for patients with liver scarring due to fatty liver disease. FDA web site, <https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-patients-liver-scarring-due-fatty-liver-disease>. Accessed 14 Mar 2024.

9. Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med 2018;24:908-922.
crossref pmid pmc pdf
10. Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest 2019;129:3990-4000.
crossref pmid pmc
11. Lomonaco R, Ortiz-Lopez C, Orsak B, Webb A, Hardies J, Darland C, et al. Effect of adipose tissue insulin resistance on metabolic parameters and liver histology in obese patients with nonalcoholic fatty liver disease. Hepatology 2012;55:1389-1397.
crossref pmid
12. Sanders FWB, Acharjee A, Walker C, Marney L, Roberts LD, Imamura F, et al. Hepatic steatosis risk is partly driven by increased de novo lipogenesis following carbohydrate consumption. Genome Biol 2018;19:79.
crossref pmid pmc pdf
13. Zhou M, Lv J, Chen X, Shi Y, Chao G, Zhang S. From gut to liver: exploring the crosstalk between gut-liver axis and oxidative stress in metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2025;30:101777.
crossref pmid
14. Zhang J, Li Y, Yang L, Ma N, Qian S, Chen Y, et al. New advances in drug development for metabolic dysfunction-associated diseases and alcohol-associated liver disease. Cell Biosci 2024;14:90.
crossref pmid pmc pdf
15. Lawler PR, Bhatt DL, Godoy LC, Lüscher TF, Bonow RO, Verma S, et al. Targeting cardiovascular inflammation: next steps in clinical translation. Eur Heart J 2021;42:113-131.
crossref pmid pdf
16. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature 2008;454:436-444.
crossref pmid pdf
17. Hydes TJ, Ravi S, Loomba R, E Gray M. Evidence-based clinical advice for nutrition and dietary weight loss strategies for the management of NAFLD and NASH. Clin Mol Hepatol 2020;26:383-400.
crossref pmid pmc pdf
18. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, Torres-Gonzalez A, Gra-Oramas B, Gonzalez-Fabian L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology 2015;149:367-378.e5 quiz e14-5.
crossref pmid
19. Le P, Tatar M, Dasarathy S, Alkhouri N, Herman W, Deshpande A, et al. Projection of the clinical burden of NAFLD in US adults from 2020-2050: a modeling study. Paper presented at: The Liver Meeting. 2023 Nov 10-14. Boston, MA, USA: AASLD abstract 2245.

20. Rachakonda V, Wills R, DeLany JP, Kershaw EE, Behari J. Differential impact of weight loss on nonalcoholic fatty liver resolution in a North American cohort with obesity. Obesity (Silver Spring) 2017;25:1360-1368.
crossref pmid pdf
21. Lassailly G, Caiazzo R, Ntandja-Wandji LC, Gnemmi V, Baud G, Verkindt H, et al. Bariatric surgery provides long-term resolution of nonalcoholic steatohepatitis and regression of fibrosis. Gastroenterology 2020;159:1290-1301.e5.
crossref pmid
22. Rinella ME, Neuschwander-Tetri BA, Siddiqui MS, Abdelmalek MF, Caldwell S, Barb D, et al. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology 2023;77:1797-1835.
crossref pmid
23. Ritter MJ, Amano I, Hollenberg AN. Thyroid hormone signaling and the liver. Hepatology 2020;72:742-752.
crossref pmid pdf
24. Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, et al.; MAESTRO-NASH Investigators. A phase 3, randomized, controlled trial of resmetirom in NASH with liver fibrosis. N Engl J Med 2024;390:497-509.
pmid
25. Nauck MA, Quast DR, Wefers J, Pfeiffer AFH. The evolving story of incretins (GIP and GLP-1) in metabolic and cardiovascular disease: a pathophysiological update. Diabetes Obes Metab 2021;23 Suppl 3:5-29.
pmid
26. Davies M, Færch L, Jeppesen OK, Pakseresht A, Pedersen SD, Perreault L, et al.; STEP 2 Study Group. Semaglutide 2·4 mg once a week in adults with overweight or obesity, and type 2 diabetes (STEP 2): a randomised, double-blind, double-dummy, placebo-controlled, phase 3 trial. Lancet 2021;397:971-984.
crossref pmid
27. Newsome P, Sanyal A, Kliers I, Ostergaard L, Long M, Kjaer M, et al. Phase 3 ESSENCE trial: semaglutide in metabolic dysfunction-associated steatohepatitis (MASH). Paper presented at: The Liver Meeting. 2024 Nov 15-19. San Diego, CA, USA: AASLD abstract 5018.

28. Husain M, Bain SC, Jeppesen OK, Lingvay I, Sørrig R, Treppendahl MB, et al. Semaglutide (SUSTAIN and PIONEER) reduces cardiovascular events in type 2 diabetes across varying cardiovascular risk. Diabetes Obes Metab 2020;22:442-451.
crossref pmid pmc pdf
29. Elsaid MI, Li N, Firkins SA, Rustgi VK, Paskett ED, Acharya C, et al. 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.
crossref pmid
30. Wester A, Shang Y, Toresson Grip E, Matthews AA, Hagström H. Glucagon-like peptide-1 receptor agonists and risk of major adverse liver outcomes in patients with chronic liver disease and type 2 diabetes. Gut 2024;73:835-843.
crossref pmid
31. Loomba R, Hartman ML, Lawitz EJ, Vuppalanchi R, Boursier J, Bugianesi E, et al.; SYNERGY-NASH Investigators. Tirzepatide for metabolic dysfunction-associated steatohepatitis with liver fibrosis. N Engl J Med 2024;391:299-310.
crossref pmid
32. Sanyal AJ, Bedossa P, Fraessdorf M, Neff GW, Lawitz E, Bugianesi E, et al.; 1404-0043 Trial Investigators. A phase 2 randomized trial of survodutide in MASH and fibrosis. N Engl J Med 2024;391:311-319.
crossref pmid
33. Samms RJ, Coghlan MP, Sloop KW. How may GIP enhance the therapeutic efficacy of GLP-1? Trends Endocrinol Metab 2020;31:410-421.
crossref pmid
34. Ludvik B, Giorgino F, Jódar E, Frias JP, Fernández Landó L, Brown K, et al. Once-weekly tirzepatide versus once-daily insulin degludec as add-on to metformin with or without SGLT2 inhibitors in patients with type 2 diabetes (SURPASS-3): a randomised, open-label, parallel-group, phase 3 trial. Lancet 2021;398:583-598.
crossref pmid
35. 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.
crossref pmid
36. Boland ML, Laker RC, Mather K, Nawrocki A, Oldham S, Boland BB, et al. Resolution of NASH and hepatic fibrosis by the GLP-1R/GcgR dual-agonist Cotadutide via modulating mitochondrial function and lipogenesis. Nat Metab 2020;2:413-431.
crossref pmid pmc pdf
37. Harrison SA, Frias JP, Neff G, Abrams GA, Lucas KJ, Sanchez W, et al.; HARMONY Study Group. Safety and efficacy of once-weekly efruxifermin versus placebo in non-alcoholic steatohepatitis (HARMONY): a multicentre, randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Gastroenterol Hepatol 2023;8:1080-1093.
crossref pmid
38. Loomba R, Sanyal AJ, Kowdley KV, Bhatt DL, Alkhouri N, Frias JP, et al. Randomized, controlled trial of the FGF21 analogue pegozafermin in NASH. N Engl J Med 2023;389:998-1008.
crossref pmid pmc
39. Geng L, Lam KSL, Xu A. The therapeutic potential of FGF21 in metabolic diseases: from bench to clinic. Nat Rev Endocrinol 2020;16:654-667.
crossref pmid pdf
40. Akero Therapeutics. Corporate presentation. Akero web site, <https://ir.akerotx.com/static-files/c76e5581-39ab-4d46-a6c7-b5893f7a2210>. Accessed 20 Jan 2025.

41. Akero Therapeutics. Akero Therapeutics reports preliminary topline results showing statistically significant reversal of compensated cirrhosis (F4) due to MASH—by both completer and ITT analyses—at week 96 in phase 2b SYMMETRY study. Akero web site, <https://ir.akerotx.com/news-releases/newsrelease-details/akero-therapeutics-reports-preliminary-topline-results-showing>. Accessed 20 Jan 2025.

42. Noureddin M, Kowdley K, Clawson A, Odrljin T, Bryant M, Jeglinski B, et al. Once-monthly efimosfermin alfa (BOS-580) in metabolic dysufnction-associated steatoheatitis with F2/F3 fibrosis: results from a 24 week, randomized, double-blind, placebo-controlled, phase 2 trial [Abstract]. Hepatology 2024;80:S1-S2011.
pmid
43. Lefere S, Puengel T, Hundertmark J, Penners C, Frank AK, Guillot A, et al. Differential effects of selective- and pan-PPAR agonists on experimental steatohepatitis and hepatic macrophages. J Hepatol 2020;73:757-770.
crossref pmid
44. Wang Y, Nakajima T, Gonzalez FJ, Tanaka N. PPARs as metabolic regulators in the liver: lessons from liver-specific PPARnull mice. Int J Mol Sci 2020;21:2061.
crossref pmid pmc
45. Francque SM, Bedossa P, Ratziu V, Anstee QM, Bugianesi E, Sanyal AJ, et al.; NATIVE Study Group. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. N Engl J Med 2021;385:1547-1558.
crossref pmid
46. O’Farrell M, Duke G, Crowley R, Buckley D, Martins EB, Bhattacharya D, et al. FASN inhibition targets multiple drivers of NASH by reducing steatosis, inflammation and fibrosis in preclinical models. Sci Rep 2022;12:15661.
pmid pmc
47. Loomba R, Bedossa P, Grimmer K, Kemble G, Bruno Martins E, McCulloch W, et al. Denifanstat for the treatment of metabolic dysfunction-associated steatohepatitis: a multicentre, double-blind, randomised, placebo-controlled, phase 2b trial. Lancet Gastroenterol Hepatol 2024;9:1090-1100.
crossref pmid
48. Harrison SA, Thang C, Bolze S, Dewitt S, Hallakou-Bozec S, Dubourg J, et al. Evaluation of PXL065 - deuterium-stabilized (R)-pioglitazone in patients with NASH: a phase II randomized placebo-controlled trial (DESTINY-1). J Hepatol 2023;78:914-925.
crossref pmid
49. Harrison SA, Alkhouri N, Benun J, Ortiz-Lasanta G, Rudraraju M, Steineger HH, et al. Icosabutate in NASH/MASH with fibrosis: results from a randomised, multicentre, double-blind, placebo controlled, phase 2b trial (Icona) [Abstract]. Hepatology 2024;79:E44-E45.

50. Harrison SA, Allen AM, Dubourg J, Noureddin M, Alkhouri N. Challenges and opportunities in NASH drug development. Nat Med 2023;29:562-573.
crossref pmid pdf
51. U.S. Food and Drug Administration (FDA). Noncirrhotic nonalcoholic steatohepatitis with liver fibrosis: developing drugs for treatment. U.S. FDA web site, <https://www.fda.gov/regulatory-information/search-fda-guidance-documents/noncirrhotic-nonalcoholic-steatohepatitis-liver-fibrosis-developing-drugs-treatment>. Accessed 20 Jan 2025.

52. Hagström H, Nasr P, Ekstedt M, Hammar U, Stål P, Hultcrantz R, et al. Fibrosis stage but not NASH predicts mortality and time to development of severe liver disease in biopsy-proven NAFLD. J Hepatol 2017;67:1265-1273.
crossref pmid
53. Vuppalanchi R, Unalp A, Van Natta ML, Cummings OW, Sandrasegaran KE, Hameed T, et al. Effects of liver biopsy sample length and number of readings on sampling variability in nonalcoholic Fatty liver disease. Clin Gastroenterol Hepatol 2009;7:481-486.
crossref pmid
54. Kuwashiro T, Takahashi H, Hyogo H, Ogawa Y, Imajo K, Yoneda M, et al. Discordant pathological diagnosis of non-alcoholic fatty liver disease: a prospective multicenter study. JGH Open 2019;4:497-502.
crossref pmid pmc pdf
55. Harrison SA, Dubourg J. Liver biopsy evaluation in MASH drug development: think thrice, act wise. J Hepatol 2024;81:886-894.
crossref pmid
56. Noureddin M, Charlton MR, Harrison SA, Bansal MB, Alkhouri N, Loomba R, et al. Expert panel recommendations: practical clinical applications for initiating and monitoring resmetirom in patients with MASH/NASH and moderate to noncirrhotic advanced fibrosis. Clin Gastroenterol Hepatol 2024;22:2367-2377.
crossref pmid
57. Chen VL, Morgan TR, Rotman Y, Patton HM, Cusi K, Kanwal F, et al. Resmetirom therapy for metabolic dysfunction-associated steatotic liver disease: October 2024 updates to AASLD practice guidance. Hepatology 2025;81:312-320.
crossref pmid
58. Tincopa MA, Loomba R. Non-invasive diagnosis and monitoring of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Lancet Gastroenterol Hepatol 2023;8:660-670.
crossref pmid
59. Huang DQ, Sharpton SR, Amangurbanova M, Tamaki N, Sirlin CB, Loomba R; NAFLD Research Study Group. Clinical utility of combined MRI-PDFF and ALT response in predicting histologic response in nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2023;21:2682-2685.e4.
crossref pmid
60. Petta S, Sebastiani G, Viganò M, Ampuero J, Wai-Sun Wong V, Boursier J, et al. Monitoring occurrence of liver-related events and survival by transient elastography in patients with nonalcoholic fatty liver disease and compensated advanced chronic liver disease. Clin Gastroenterol Hepatol 2021;19:806-815.e5.
crossref pmid
61. Loomba R, Schattenberg J, Schattenberg JM, Taub R, Labriola D, Noureddin M, et al. 521 ALT, FibroScan VCTE, and FibroScan Cap as predictive markers of resmetirom biopsy response. Gastroenterology 2024;166:S-1550.
crossref
62. Lin H, Lee HW, Yip TC, Tsochatzis E, Petta S, Bugianesi E, et al.; VCTE-Prognosis Study Group. Vibration-controlled transient elastography scores to predict liver-related events in steatotic liver disease. JAMA 2024;331:1287-1297.
pmid pmc
63. Song SJ, Lai JC, Wong GL, Wong VW, Yip TC. Can we use old NAFLD data under the new MASLD definition? J Hepatol 2024;80:e54-e56.
crossref pmid
64. 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.
crossref pmid
65. Vali Y, Lee J, Boursier J, Petta S, Wonders K, Tiniakos D, et al.; Liver Investigation: Testing Marker Utility in Steatohepatitis (LITMUS) consortium investigators. Biomarkers for staging fibrosis and non-alcoholic steatohepatitis in non-alcoholic fatty liver disease (the LITMUS project): a comparative diagnostic accuracy study. Lancet Gastroenterol Hepatol 2023;8:714-725.
crossref pmid
66. Liu CH, Ampuero J, Gil-Gómez A, Montero-Vallejo R, Rojas Á, Muñoz-Hernández R, et al. miRNAs in patients with non-alcoholic fatty liver disease: a systematic review and meta-analysis. J Hepatol 2018;69:1335-1348.
crossref pmid
67. Harrison SA, Ratziu V, Magnanensi J, Hajji Y, Deledicque S, Majd Z, et al. NIS2+™, an optimisation of the blood-based biomarker NIS4® technology for the detection of at-risk NASH: a prospective derivation and validation study. J Hepatol 2023;79:758-767.
crossref pmid
68. Sanyal AJ, Williams SA, Lavine JE, Neuschwander-Tetri BA, Alexander L, Ostroff R, et al. Defining the serum proteomic signature of hepatic steatosis, inflammation, ballooning and fibrosis in non-alcoholic fatty liver disease. J Hepatol 2023;78:693-703.
crossref pmid
69. Fishman JC, Qian C, Kim Y, Rochon H, Szabo SM, Sun R, et al. Cost burden of cirrhosis and liver disease progression in metabolic dysfunction-associated steatohepatitis: a US cohort study. J Manag Care Spec Pharm 2024;30:929-941.
crossref pmid pmc
70. Rustgi VK, Duff SB, Elsaid MI. Cost-effectiveness and potential value of pharmaceutical treatment of nonalcoholic fatty liver disease. J Med Econ 2022;25:347-355.
crossref pmid
71. Javanbakht M, Fishman J, Moloney E, Rydqvist P, Ansaripour A. Early cost-effectiveness and price threshold analyses of resmetirom: an investigational treatment for management of nonalcoholic steatohepatitis. Pharmacoecon Open 2023;7:93-110.
crossref pmid pdf
72. Fishman J, Kim Y, Parisé H, Bercaw E, Smith Z. Budget impact of resmetirom for the treatment of adults with non-cirrhotic non-alcoholic steatohepatitis (NASH) with moderate to advanced liver fibrosis (consistent with stages F2 to F3 fibrosis). J Med Econ 2024;27:1108-1118.
crossref pmid
73. Assefa Y, Hill PS, Williams OD. Access to hepatitis C virus treatment: Lessons from implementation of strategies for increasing access to antiretroviral treatment. Int J Infect Dis 2018;70:65-68.
crossref pmid
74. Ratziu V, Friedman SL. Why do so many nonalcoholic steatohepatitis trials fail? Gastroenterology 2023;165:5-10.
crossref pmid
75. Ratziu V, Charlton M. Rational combination therapy for NASH: insights from clinical trials and error. J Hepatol 2023;78:1073-1079.
crossref pmid
76. Noureddin M. MASH clinical trials and drugs pipeline: an impending tsunami. Hepatology 2024 Mar 19;doi: 10.1097/HEP.0000000000000860.
crossref
77. Do A, Zahrawi F, Mehal WZ. Therapeutic landscape of metabolic dysfunction-associated steatohepatitis (MASH). Nat Rev Drug Discov 2025;24:171-189.
crossref pmid pdf
78. Harrison SA, Rolph T, Knott M, Dubourg J. FGF21 agonists: an emerging therapeutic for metabolic dysfunction-associated steatohepatitis and beyond. J Hepatol 2024;81:562-576.
crossref pmid
79. Harrison SA, Frias JP, Lucas KJ, Reiss G, Neff G, Bollepalli S, et al. Safety and efficacy of efruxifermin in combination with a GLP-1 receptor agonist in patients with NASH/MASH and type 2 diabetes in a randomized phase 2 study. Clin Gastroenterol Hepatol 2025;23:103-113.
crossref pmid
80. Alkhouri N, Herring R, Kabler H, Kayali Z, Hassanein T, Kohli A, et al. Safety and efficacy of combination therapy with semaglutide, cilofexor and firsocostat in patients with non-alcoholic steatohepatitis: a randomised, open-label phase II trial. J Hepatol 2022;77:607-618.
crossref pmid
81. Loomba R, Abdelmalek MF, Armstrong MJ, Jara M, Kjær MS, Krarup N, et al.; NN9931-4492 investigators. Semaglutide 2·4 mg once weekly in patients with non-alcoholic steatohepatitisrelated cirrhosis: a randomised, placebo-controlled phase 2 trial. Lancet Gastroenterol Hepatol 2023;8:511-522.
crossref pmid pmc
82. Caddeo A, Romeo S. Precision medicine and nucleotide-based therapeutics to treat steatotic liver disease. Clin Mol Hepatol 2025;31(Suppl):S76-S93.
crossref pmid pdf

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: cmh_journal@ijpnc.com
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 3204
TOTAL : 2897706
Close layer