Resmetirom and semaglutide in metabolic dysfunction-associated steatohepatitis (MASH): a comparative perspective
Sections
Open Access Perspective
Resmetirom and semaglutide in metabolic dysfunction-associated steatohepatitis (MASH): a comparative perspective

Affiliation:

Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany

Email: rweiskirchen@ukaachen.de

ORCID: https://orcid.org/0000-0003-3888-0931

Ralf Weiskirchen
*

Explor Drug Sci. 2025;3:1008132 DOl: https://doi.org/10.37349/eds.2025.1008132

Received: September 03, 2025 Accepted: September 23, 2025 Published: October 30, 2025

Academic Editor: Alessandra Tolomelli, University of Bologna, Italy

The article belongs to the special issue Innovative Therapeutics in Hepato-Gastroenterology

Abstract

Metabolic dysfunction-associated steatohepatitis (MASH) is emerging as a leading cause of cirrhosis, hepatocellular carcinoma, and liver-related mortality worldwide. Among the most advanced pharmacologic candidates are resmetirom, a highly liver-selective thyroid hormone receptor-β (THR-β) agonist, and semaglutide, a long-acting glucagon-like peptide-1 receptor agonist (GLP-1 RA) already approved for diabetes and obesity. Although both agents improve hepatic steatosis, their mechanisms of action, extra-hepatic benefits, and safety signatures diverge markedly. Resmetirom, which was approved by the Food and Drug Administration (FDA) in March 2024, acts hepatocentrically to accelerate β-oxidation, lower atherogenic lipoproteins, and deliver early signals necessary for fibrosis regression, all while largely avoiding systemic thyrotoxic effects. Semaglutide acts systemically by reducing caloric load through pronounced weight loss and glycemic control, producing the highest rates of histologic MASH resolution reported to date, albeit with less direct antifibrotic efficacy and characteristic gastrointestinal tolerability issues. This comparative perspective juxtaposes the two compounds with respect to molecular pharmacology, clinical efficacy, safety, and potential clinical positioning, and proposes that, because resmetirom primarily targets hepatic lipid disposal whereas semaglutide unloads systemic caloric pressure, their complementary actions could be harnessed sequentially or in combination to achieve broader, more durable disease modification across the heterogeneous spectrum of patients with MASH.

Keywords

MASLD, therapy, drugs, thyroid hormone receptor-β agonist, GLP-1 receptor agonist

Introduction

Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as nonalcoholic steatohepatitis (NASH), is on track to become the leading cause of liver transplantation in Western countries and a significant factor in hepatocellular carcinoma (HCC), cardiovascular issues, and overall mortality [1, 2]. Despite years of research, only resmetirom is licensed for use in the United States for treating adults with non-cirrhotic MASH with moderate to advanced liver fibrosis. However, the treatment landscape has seen significant changes in the past five years, with several late-phase candidates in development. Together with resmetirom, a selective thyroid hormone receptor-β (THR-β) agonist, semaglutide, a glucagon-like peptide-1 receptor agonist (GLP-1 RA) [3, 4], has shown promising results in phase II and phase III trials. However, resmetirom and semaglutide differ significantly in their mechanisms of action, metabolic effects, safety profiles, and potential clinical applications.

Estimates from a meta-analysis suggest that the global prevalence of nonalcoholic fatty liver disease (NAFLD) is 30.1%, with a significant increase from 25.3% (1990–2006) to 38.2% (2016–2019) over the last three decades. This increase aligns with the rising global epidemic of obesity and type 2 diabetes mellitus (T2DM) [5]. In high-income countries like the United States, NASH-related cirrhosis was the second leading cause of liver transplantation among patients without HCC, and the leading cause in women without HCC [6]. Additionally, NASH-related cirrhosis is the fastest-growing cause of acute-on-chronic liver failure (ACLF), with patients experiencing longer hospital stays, increased use of dialysis and long-term care, higher in-hospital mortality rates, and higher median total health care charges [7].

The pathophysiology underlying the disorders of NAFLD and MASH is complex and not fully understood. It includes altered liver function, hepatic insulin resistance and lipotoxicity as key features, all of which are influenced by gut alterations that result in increased intestinal permeability [8]. It is a heterogeneous condition and a heightened burden of subclinical atherosclerosis is evident primarily in patients with coexisting metabolic-syndrome traits [1, 911]. Moreover, in recent years, NAFLD has evolved from an isolated liver condition to a systemic disease with significant manifestations beyond the liver. In particular, cardiovascular diseases, often with major adverse cardiovascular-cerebral events, are clinically relevant [1214]. These epidemiologic data underscore an urgent need for pharmacologic interventions that can be instituted long before end-stage disease develops.

In March 2024, the U.S. Food and Drug Administration (FDA) granted accelerated approval to resmetirom for adults with non-cirrhotic MASH and stage F2–F3 fibrosis, thereby establishing the first regulatory foothold in this therapeutic area [15].

This Perspective compares the two drugs in four areas: (i) molecular and cellular mechanisms, (ii) evidence of effectiveness, (iii) safety and tolerability, and (iv) future role in the evolving treatment plan for MASH. Instead of declaring a clear “winner”, our aim is to provide a detailed framework for clinicians, researchers, and policymakers to consider how each drug could be used in real-world scenarios once regulatory obstacles are overcome.

Baseline pharmacologic profiles of resmetirom and semaglutide

Table 1 summarizes the most relevant physicochemical and pharmacological parameters of resmetirom and semaglutide for the treatment of MASH, providing an at-a-glance comparison of their molecular size, formulation, pharmacokinetics, and key therapeutic effects. The table shows that resmetirom is a small (molecular weight about 600 g/mol), non-iodinated THR-β agonist administered once daily as an oral tablet, while semaglutide is a large (about 4 kDa) GLP-1 peptide designed for once-weekly subcutaneous injection or, with SNAC enhancement, once-daily oral administration. Additionally, they have distinct half-lives (approximately 35 h vs. 160 h), different oral bioavailability (approximately 30% vs. 1%), and unique clinical benefits. Resmetirom provides significant low-density lipoprotein cholesterol (LDL-C) and apolipoprotein B (ApoB) reduction along with early hepatic antifibrotic effects, whereas semaglutide leads to substantial weight loss and wide-ranging cardiometabolic improvements. The contrasting yet complementary profiles presented in Table 1 support the need to personalize treatment based on patient characteristics or even consider combination therapies in future studies.

 Key physicochemical and pharmacological characteristics of resmetirom and semaglutide relevant to MASH therapy.

Parameter    Resmetirom (MGL-3196)    Semaglutide
Drug class/primary target    Selective THR-β agonist    Long-acting GLP-1 receptor agonist
CAS number    920509-32-6    910463-68-2
Molecular formula    C17H12Cl2N6O4    C187H291N45O59
Molecular weight    600.45 g·mol–1    4,113.58 g·mol–1
2-D structure    See Figure 1    See Figure 2
Physical appearance    White to off-white crystalline powder    White lyophilised powder (pen) or tablets (with SNAC)
Aqueous solubility (25°C)    < 0.1 mg·mL–1 (practically insoluble)    Highly soluble ≥ 1 mg·mL–1, pH ≈ 7.4
Solubility in DMSO/buffers    DMSO ≈ 45 mg·mL–1
    EtOH ≈ 2 mg·mL–1
    Fully soluble in isotonic aqueous buffers
LogP (cLogP)*    ≈ 4.8    n/a (large peptide)
pKa (dominant)    4.7 (carboxylic acid)    Multiple; isoelectric point ≈ 4.8
Formulation in trials    Immediate-release oral tablets 40/80/100 mg    Prefilled s.c. pens 0.25–2.4 mg; oral 3/7/14 mg (SNAC-enhanced)
Typical MASH study dose    80–100 mg orally once daily    0.4 mg s.c. daily (phase II); 2.4 mg s.c. weekly in obesity
Oral bioavailability    ≈ 30%    ≈ 1% (oral); s.c. ≈ 89%
Plasma protein binding    > 99%    > 99%
Elimination half-life    ≈ 35 h    ≈ 160 h (≈ 7 days)
Main metabolism/clearance    Hepatic oxidation & conjugation; biliary/fecal excretion    Proteolysis followed by β-oxidation; renal (54%) peptide fragments
Principal route(s) of administration    Oral    s.c.; oral (with SNAC)
Key therapeutic signals in MASH↓ MRI-PDFF, ↑ MASH resolution (≈ 30%), early fibrosis improvement    MASH resolution up to 59%, ≥ 13% weight loss, fibrosis signal weight-dependent
Most frequent adverse events    Mild pruritus, transient GI discomfort, reversible ↓ TSH    Nausea, vomiting, diarrhoea, gall-bladder events, and rare pancreatitis
Development status (mid-2024)    Licensed for use in non-cirrhotic MASH in March 2024 in the USA    Phase III ESSENCE-NASH ongoing; already marketed for T2DM & obesity
Representative trade names approved by the FDA    Rezdiffra (tablets)    Rybelsus (tablets), Ozempic (injection), Wegovy (injection)
Storage    2–8°C, protected from moisture/light    2–8°C (pens); room temperature ≤ 30°C for ≤ 56 days (in-use pens)
Mechanism of action    Hepatic    Systemic
Direct improvement of insulin resistance    No    Yes
Extra-hepatic benefits    Improves atherogenic dyslipidemia by strong LDL-C & ApoB lowering    Glycaemic & cardio-nephro-metabolic benefits

ApoB: apolipoprotein B; EtOH: ethanol; FDA: Food and Drug Administration; GI: gastrointestinal; GLP-1: glucagon-like peptide-1; LDL-C: low-density lipoprotein cholesterol; MASH: metabolic dysfunction-associated steatohepatitis; MRI-PDFF: magnetic resonance imaging-proton density fat fraction; NASH: nonalcoholic steatohepatitis; s.c.: subcutaneous; SNAC: sodium N-[8-(2-hydroxybenzoyl)amino] caprylate; T2DM: type 2 diabetes mellitus; THR-β: thyroid hormone receptor-β; TSH: thyroid-stimulating hormone. * LogP (cLogP) refers to the logarithm of the octanol-water partition coefficient, a measure of the fat-solubility of a drug.

Mechanistic rationale: resmetirom

Thyroid hormones play a crucial role in regulating energy expenditure, lipid turnover, and mitochondrial respiration. In hepatocytes, triiodothyronine (T3) binding to THR-β stimulates β-oxidation, increases LDL receptor expression, improves mitochondrial function in hepatic cells, boosts very-low-density lipoprotein (VLDL) secretion, and enhances reverse cholesterol transport, thereby reducing lipotoxicity and inflammation [3, 16]. Research on humans and rodents indicates that intrahepatic hypothyroidism (resulting from elevated deiodinase 3 activity or reduced THR-β expression) is a key factor in MASH. However, administering thyroid hormone systematically exposes to the risks of cardiotoxicity and bone loss mediated by THR-α before any hepatic benefits can be seen. Resmetirom (MGL-3196) addresses this issue by selectively activating THR-β with a 28-fold affinity compared to THR-α [16].

Structurally, resmetirom (MGL-3196) is a synthetic, non-iodinated small molecule that selectively targets THR-β. Its backbone consists of two substituted phenyl rings linked by an ether bridge, creating a compact, planar scaffold (Figure 1). Halogen substituents, one chlorine atom, a para-fluoro group, and a trifluoromethyl moiety, boost lipophilicity and metabolic stability. A terminal carboxylic acid side chain mimics the acidic tail of native thyroid hormones and anchors the ligand in the receptor pocket. Altogether, these features confer high THR-β affinity while supporting once-daily oral dosing. A phase III, randomized, controlled trial of resmetirom in biopsy-confirmed NASH patients with liver fibrosis was conducted. In this trial, 322 subjects received 80 mg of resmetirom, 323 subjects received 100 mg of resmetirom, and 321 subjects received a placebo once daily for 52 weeks. The study showed that the drug was highly effective in terms of NASH resolution and improvement of liver fibrosis [17].

Chemical structures of Resmetirom (MGL-3196) and endogenous thyroid hormone triiodothyronine (T3). (A) Resmetirom, a synthetic, liver-targeted, selective thyroid-hormone-receptor-β (THR-β) agonist, consists of a tri-substituted benzyl ether core in which (i) two iodine atoms present in T3 are replaced by polar, non-halogen substituents (a trifluoromethyl group and a cyano group), and (ii) the hydrophobic phenolic ring of T3 is replaced by a 3,5-dichloro-4-(4-cyano)phenyl moiety. (B) T3 is the natural ligand for thyroid hormone receptors and features (i) a diphenyl ether backbone containing three iodine atoms at positions 3, 5, and 3’, (ii) an α-amino-propionic acid side chain conferring zwitterionic character, and (iii) a phenolic hydroxyl group essential for receptor binding. Molecular formulas, molecular weights, substance identifiers (SID), and CAS numbers for each substance are given.

Resmetirom also takes advantage of hepatic first-pass extraction to achieve a liver-to-plasma exposure ratio of approximately 8:1 [16, 18]. In animal studies, resmetirom increases the expression of genes involved in fatty acid oxidation (CPT1A, ACOX1), mitochondrial biogenesis (PPARGC1A), and cholesterol catabolism (CYP7A1), while reducing levels of lipogenic targets (SREBP-1c, FASN) [1921]. These changes at the genetic level lead to a decrease in hepatic triglyceride levels, improved insulin sensitivity, and a reduction in inflammatory and fibrotic pathways related to NF-κB and transforming growth factor-β (TGF-β). Additionally, resmetirom lowers levels of circulating LDL-C (by 20% to 30%) and ApoB (by 15% to 25%), providing a dual benefit for individuals with atherogenic dyslipidemia. Overall, the metabolic impact of resmetirom can be likened to pharmacological simulation of mild hyperthyroidism localized to hepatocytes, thus avoiding any systemic complications from excess thyroid hormone [16].

In conclusion, the liver selectivity of resmetirom is achieved through two complementary design principles: (i) a 28-fold higher binding affinity for THR-β compared to THR-α [18], and (ii) preferential first-pass hepatic extraction that results in a favorable liver-to-plasma exposure ratio. This effectively protects extra-hepatic tissues from thyromimetic stimulation, demonstrating liver-specific action without causing systemic adverse effects [16, 18].

Mechanistic rationale: semaglutide

Semaglutide is a 31-amino-acid analogue of the human incretin hormone GLP-1 that has been strategically modified to boost its stability and half-life (Figure 2). At position 8, the native alanine is replaced by the protease-resistant amino acid α-aminoisobutyric acid (Aib). This substitution makes the peptide resistant to dipeptidyl-peptidase-4 (DPP-4), whereas the C18 fatty di-acid side chain attached to Lys26 ensures high-affinity, reversible albumin binding; together, these modifications extend the elimination half-life to ~160 h and allow once-weekly dosing [22, 23].

Structure of semaglutide. The analog of human glucagon-like peptide-1 (GLP-1) is composed of 31 amino acids with the depicted sequence, which are numbered from 7–37 (according to the unprocessed GLP-1 protein). Compared to GLP-1, semaglutide carries a substitution of the alanine at position 8 with α-aminoisobutyric acid (Aib) and acylation of the lysine at position 26 with a fatty di-acid side chain for extended half-life, which is linked via a linker sequence. Moreover, it carries an amino acid substitution at position 34 (lysine to arginine), which prevents the C18 fatty acid binding moiety at the wrong site. Molecular formula, molecular weight, substance identifier (SID), and CAS number for semaglutide are given.

Furthermore, the ε-amine of the lysine at position 26 is acylated: via a glutamic acid linker, a C18 fatty di-acid (stearic acid derivative) is attached [4]. This hydrophobic side chain allows reversible binding to serum albumin, shielding the peptide from rapid renal clearance and enabling once-weekly dosing.

Apart from these substitutions, the sequence largely mirrors native GLP-1, so semaglutide retains potent agonism at the GLP-1 receptor while circulating far longer in the bloodstream, which allows for once-weekly subcutaneous or once-daily oral dosing options [4]. Originally developed for glycemic control, semaglutide promotes glucose-dependent insulin secretion, suppresses glucagon release, delays gastric emptying, and, most importantly for MASH, produces significant weight loss through the hypothalamic regulation of appetite and satiety.

In addition to calorie restriction, GLP-1 signaling has direct effects on the liver and other organs relevant to MASH. In laboratory studies, GLP-1 RAs decrease de novo lipogenesis by down-regulating SREBP-1c and up-regulating AMPK, enhance β-oxidation through PPAR-α activation, and reduce oxidative stress through cAMP-PKA-mediated pathways [21, 24]. Semaglutide also decreases endotoxemia-induced Kupffer cell activation and influences the gut-liver axis by changing bile acid pools and intestinal permeability. Moreover, semaglutide reduces appetite and increases satiety [25]. In rodent models, these cellular changes result in reduced steatosis, lobular inflammation, and ballooning. However, liver fibrosis regression is not consistently observed, suggesting that the antifibrotic effects are a result of metabolic improvement. Overall, the mechanisms of semaglutide action can be described as a systemic, weight-focused intervention that targets caloric overload, insulin resistance, and pro-inflammatory adipokine profiles, all of which contribute to the liver pathology of MASH.

Therapeutic efficacy: resmetirom

The pivotal phase II trial (MGL-3196-05, n = 125) randomized patients with biopsy-proven MASH to receive either 80 mg of resmetirom or a placebo for 36 weeks [26]. The primary endpoint, which was the relative reduction in magnetic resonance imaging-proton density fat fraction (MRI-PDFF) assessed hepatic fat, was achieved with a placebo-adjusted difference of –29%. Additionally, 32% of patients on resmetirom vs. 10% on placebo achieved a ≥ 2-point reduction in NAS without worsening fibrosis. Notably, histologic fibrosis stage improved by ≥ 1 point in 24% of patients on resmetirom compared to 10% on placebo, a signal that is not commonly seen with weight-neutral agents in such a short timeframe [26]. Moreover, LDL-C, triglycerides, ApoB, and Lp(a) all decreased significantly, highlighting cardiometabolic benefits of the compound.

Based on these findings, the double-blind phase III MAESTRO-NASH trial enrolled over 950 non-cirrhotic patients, stratified by fibrosis stage (F1–F3) and metabolic comorbidities [17]. After 52 weeks, 26% and 30% of participants who received 80 mg and 100 mg of resmetirom, respectively, achieved MASH resolution with no fibrosis worsening compared to 10% on placebo (p < 0.0001). Fibrosis improvement of ≥ 1 stage was seen in 24% (80 mg) and 26% (100 mg) compared to 14% in the placebo group (p < 0.005 for both doses) [17]. The lipid-lowering effects persisted, with LDL-C reductions of approximately 19% and ApoB by 16%. Pruritus, mostly mild, emerged as the main adverse event (see below). While 52 weeks may be considered too short to assess hard outcomes like cirrhosis or HCC incidence, the histologic improvements surpassed the traditional regulatory threshold for accelerated approval, positioning resmetirom as a frontrunner in the MASH pipeline.

Therapeutic efficacy: semaglutide

Efficacy data for semaglutide in MASH primarily come from a 72-week, dose-finding phase II RCT (n = 320) that compared 0.1, 0.2, and 0.4 mg subcutaneous semaglutide daily versus placebo [27]. The highest dose resulted in MASH resolution in 59% of patients compared to 17% on placebo (p < 0.001), a remarkable achievement not seen with previous pharmacotherapies. Median weight loss was 13%, and glycemic parameters improved in both the pre-diabetic and type 2 diabetic subgroups. However, the fibrosis endpoint (i.e., ≥ 1-stage improvement without worsening steatohepatitis) did not reach statistical significance (p = 0.10). Post-hoc analyses revealed that patients who lost more than 10% body weight experienced greater fibrosis regression, supporting the idea that weight loss, rather than a direct antifibrotic mechanism, plays a role in histologic repair with GLP-1 RA therapy [27].

In addition to liver histology, the impact of semaglutide on cardiovascular outcomes has been confirmed in large trials for diabetes (SUSTAIN-6, LEADER) and obesity (STEP-CVOT, ongoing), where major adverse cardiovascular events (MACE) were reduced by 21–26% [2830]. Since cardiovascular disease is the leading cause of death in MASH, this additional benefit could be crucial even if the effect of semaglutide on hepatic fibrosis lags behind other medications. Furthermore, the availability of an oral formulation and once-weekly dosing increases adherence options compared to competitors.

Comparative efficacy

Semaglutide achieved the highest rate of histologic MASH resolution reported to date (59% at 0.4 mg/day), while resmetirom produced an early ≥ 1-stage fibrosis improvement within 36–52 weeks, an effect rarely seen with weight-neutral agents [31, 32]. Since the fibrosis stage is the strongest independent predictor of liver-related outcomes [33], this difference should guide treatment selection and the design of future combination studies. The complementary, yet distinct, mechanistic landscapes of the two agents are summarized in Figure 3. By visually contrasting resmetirom’s hepatocentric THR-β activation with semaglutide’s system-wide incretin signaling, the figure clarifies how each drug addresses different biological “pressure points” within the MASH spectrum, focusing on hepatic lipid disposal versus systemic caloric unloading and glyco-metabolic control. It also highlights downstream consequences (e.g., LDL-C reduction, weight loss, early fibrosis signals) and the non-overlapping adverse-event profiles that must be balanced when selecting or combining therapies.

Complementary mechanistic pathways of resmetirom and semaglutide in the treatment of MASH. Resmetirom is administered as an oral tablet entering the portal circulation, accumulating in hepatocytes, and selectively activating nuclear THR-β. The downstream transcriptional effects include up-regulation of genes that govern β-oxidation, LDL-receptor expression, and bile-acid synthesis, along with the suppression of lipogenic (SREBP-1c) and profibrotic (TGF-β) pathways. The net therapeutic outcomes are a reduction of hepatic fat, early histologic fibrosis regression, and a significant lowering of LDL cholesterol (LDL-C) and apolipoprotein B (ApoB). In contrast, semaglutide is administered as a once-weekly s.c. injection. The albumin-bound drug engages GLP-1 receptors on multiple organs including the pancreas, hypothalamic centers, stomach, and adipose tissue. Both drugs have major extra-hepatic effects. Importantly, both drugs have the potential to evolve therapeutic synergy, as indicated by “hepatic lipid disposal/antifibrotic × systemic metabolic unloading/weight loss”. GI: gastrointestinal; GLP-1: glucagon-like peptide-1; GPCR: G-protein coupled receptor; LDL: low-density lipoprotein; MASH: metabolic dysfunction-associated steatohepatitis; MRI-PDFF: magnetic resonance imaging-proton density fat fraction; SREBP-1c: sterol regulatory element-binding protein-1c; t1/2: half-life; TGF-β: transforming growth factor-β; THR-β: thyroid hormone receptor-β; s.c.: subcutaneous.

Safety and tolerability

Resmetirom has demonstrated a favorable safety profile so far. Gastrointestinal (GI) complaints (10–15%) and mild pruritus (15–20%) were more common than with placebo but rarely led to discontinuation (< 3%) [34]. Heart rate, blood pressure, and bone-mineral parameters remained stable, confirming hepatic selectivity. Transient decreases in thyroid-stimulating hormone (TSH) and modest increases in free T4 were observed but normalized without intervention, even in euthyroid patients. There is no evidence of arrhythmia, osteopenia, or thyroid neoplasia, although longer surveillance is required.

Semaglutide’s safety profile has been well characterized after > 10 million patient-years of exposure in diabetes and obesity [4, 35]. Nausea (20–40%), vomiting (5–15%), and diarrhea (10–20%) are typical GLP-1 RA class effects, usually transient and dose-dependent. Rare but significant concerns include gallstone disease (1–2%), especially in the context of rapid weight loss, and acute pancreatitis (< 0.3%). Signals for retinopathy progression in patients with pre-existing diabetic retinopathy have been observed (SUSTAIN-6) but not consistently replicated. It is important to note that semaglutide’s GI profile may overlap with the baseline dyspepsia and gastroparesis found in cirrhotics, necessitating careful titration in advanced liver disease. No hepatotoxicity or adverse lipid shifts have been reported, which is a notable contrast to some other anti-obesity agents.

It should be mentioned that there are other drugs targeting the GLP-1 RA (e.g., liraglutide, tirzepatide, and survodutide) on the market that have also shown highly encouraging results, often with higher rates of MASH resolution and fibrosis improvement [36]. Therefore, it will be essential in future studies to optimize drug regimens and identify patients most likely to benefit from one or the other of these drugs, considering the specific pathophysiological, pharmacokinetic, and pharmacodynamic differences of each population group [37]. Moreover, continuous evaluation of real-world data is critical to better define the optimal positioning of oral semaglutide along the T2DM trajectory and fully exploit its potential in everyday clinical practice [38].

Therapeutic potential and clinical positioning

If resmetirom secures regulatory approval, it could emerge as the first-in-class, liver-targeted metabolic modulator for MASH. Its neutrality on body weight might be perceived as a limitation in an obesity-driven disease; however, the pronounced lipid-lowering effect and seeming antifibrotic potency offer a unique value proposition, especially for lean MASH or for patients who are refractory to lifestyle-based weight loss. Combination strategies pairing resmetirom with antifibrotic agents (e.g., FGF-21 analogues, integrin inhibitors) are under discussion and might leverage complementary mechanisms without compounding adverse events [39, 40].

Semaglutide’s rich cardiovascular and metabolic evidence base makes it a compelling candidate for first-line therapy in MASH patients with obesity, T2DM, or metabolic syndrome. The potential to achieve a > 10% weight loss aligns with lifestyle guidelines and might amplify the effects of secondary agents targeting inflammation or fibrosis. If future trials demonstrate additive benefits when combined with antifibrotic molecules (e.g., selonsertib, cilofexor) or with resmetirom itself, semaglutide could anchor a multi-drug regimen modeled on diabetes care [41]. The challenge will be to balance GI tolerability, insurance coverage, and the need for chronic injectable therapy.

Cost-effectiveness and access

Affordability and reimbursement will heavily influence real-world uptake. In the United States, the wholesale acquisition cost of weekly injectable semaglutide at anti-obesity doses exceeds USD 18,000 per year [42]. Without insurance, however, prices vary widely: compounded versions that do not carry FDA approval have been advertised for as little as USD 129 per monthly supply, whereas the on-label, FDA-approved product can cost USD 1,850 for the same time period [43]. By comparison, the annual wholesale acquisition cost of resmetirom has been set at USD 47,400; the Institute for Clinical and Economic Review (ICER) has calculated a health benefit price benchmark of USD 39,600–50,100 per year [44]. These figures exceed, by several-fold, the per-capita pharmaceutical budget in many health-care systems and underscore a critical, often overlooked question: not every individual with hepatic steatosis can, or should, receive high-cost pharmacotherapy. Cost-effectiveness analyses consistently show that drug treatment becomes economically attractive only in patients with at least stage F2 fibrosis or with high cardiovascular risk, whereas lifestyle modification and, in particular, physical activity that induces exercise-mediated anti-inflammatory effects and dietary changes are the preferred first-line options for early, uncomplicated disease [4547]. Accordingly, payers and clinicians will need to implement risk-stratified algorithms, based on non-invasive fibrosis scores, metabolic comorbidities, and patient preferences, to decide who should be treated now, who can safely defer pharmacotherapy, and how limited resources can be deployed most equitably. Expanding coverage of anti-obesity interventions to eligible individuals could generate billions of budgetary savings to the Medicare budget [48].

Real-world adherence

Adherence patterns are likely to differ between a daily oral therapy (resmetirom) and a weekly injectable therapy (semaglutide) in an otherwise asymptomatic population. While a once-weekly injection reduces the dosing frequency, challenges such as needle aversion, refrigeration requirements, and refill visits can still impact persistence. On the other hand, a once-daily oral tablet may be easily forgotten among other medications in a population without symptoms. This underscores the importance of tailored adherence aids (such as smart pens, digital pillboxes, and mobile reminders) and shared decision-making to align dosing with individual lifestyle preferences and capabilities. Implementing effective screening procedures for NAFLD, utilizing molecular diagnostics, digital reminder systems, remote biomarker monitoring, promoting clinical guidelines, and pharmacy-led adherence programs could help prevent drop-off [4951]. Conversely, GI side effects with GLP-1 RAs or mild itching with THR-β agonists could lead to discontinuation if not proactively managed through dose adjustments and patient education [52].

Both agents are positioned to address specific, partially overlapping niches in the emerging MASH treatment algorithm. Resmetirom, already approved by the FDA for adults with non-cirrhotic MASH and stage F2–F3 fibrosis, is ideal for lean or weight-stable patients with progressive fibrogenesis or atherogenic dyslipidemia. It offers significant reduction in hepatic fat, early fibrosis improvement, and lowering of LDL-C/ApoB without affecting weight. Conversely, semaglutide, with strong cardiovascular outcomes and weight-loss efficacy, may be better suited for individuals with obesity, T2DM, or high cardiometabolic risk, where it can improve histology and provide extra-hepatic benefits through calorie reduction and glycemic control.

In clinical practice, factors such as fibrosis stage, BMI level, metabolic comorbidities, tolerability (pruritus vs. GI symptoms), and insurance criteria will influence the initial choice of therapy. Future research will determine whether sequential or combination therapy can optimize long-term disease management.

However, general long-term outcome data for both agents are still scarce. Most current evidence is limited to 52–72 weeks of exposure [4]. Extended follow-up studies are therefore essential to determine whether the histologic improvements translate into durable reductions in cirrhosis, HCC, and all-cause mortality. Only multi-year trials and real-world registries will clarify the true clinical and economic benefits of semaglutide and resmetirom.

Looking ahead, NASH trials ought to incorporate a thorough evaluation of the systemic drivers, modifiers, and manifestations of the disease. This broader lens would enable a more holistic and impartial appraisal of investigational therapies, capturing cardiovascular, renal, and metabolic endpoints in addition to traditional histologic markers of liver injury, so that therapeutic value is not judged solely on liver-specific surrogates [53].

Moreover, several studies pinpoint that sex probably modulates the response to treatment [5355]. Future research should explicitly examine sex-specific efficacy and safety profiles of resmetirom and semaglutide as part of precision-medicine efforts. In parallel, systematic evaluation of how genetic polymorphisms, environmental exposures, divergent metabolic stressors, variable susceptibility to hepatocyte lipotoxicity, and differences in tissue-repair capacity shape the actions of these drugs will be crucial for advancing truly personalized therapy [53].

Extra-hepatic complications

MASH confers excess risk not only for cardiovascular disease but also for chronic kidney disease (CKD) and several extra-hepatic malignancies [56, 57]. By improving atherogenic dyslipidaemia, resmetirom might secondarily attenuate cardio-renal progression, whereas semaglutide has already shown nephro-protective and potential anti-neoplastic signals in large T2DM trials [58, 59]. Follow-up studies and disease registries will be essential to determine whether such benefits translate into lower CKD progression rates or cancer incidence in MASH cohorts.

Sex-specific precision medicine

Mounting evidence indicates that both disease biology and drug responsiveness in MASH and fibrosis are sexually dimorphic: pre-menopausal women exhibit slower fibrosis progression but may respond differently to metabolic therapies [55, 60, 61]. Therefore, the upcoming phase III and post-marketing studies should stratify efficacy and safety endpoints by sex and menopausal status. This will enable precision algorithms to match individual profiles with the most suitable single-agent or combination regimen.

Unanswered questions and research priorities

  • Durability of response: can histologic gains be maintained beyond two years, and does early improvement translate into a reduced risk of cirrhosis, HCC, or the need for a transplant? Extension and outcome studies are ongoing for both drugs.

  • Cirrhotic populations: phase III programs have thus far focused on F1–F3 fibrosis. Whether these agents benefit compensated cirrhosis (F4) in preventing decompensation remains unclear.

  • Real-world adherence: weekly injections (semaglutide) or daily oral tablets (resmetirom) appear manageable, but longitudinal adherence in asymptomatic patients may falter. Digital health tools and biomarker-guided follow-up could help improve adherence.

  • Biomarker development: non-invasive markers predicting response to either drug could avoid the need for serial biopsies. Candidate panels include MRI-PDFF, ELF, PRO-C3, and emerging transcriptomic signatures.

  • Combination therapy design: optimal sequencing, dosing, and toxicity management when using resmetirom and semaglutide together (or with other classes) remains speculative yet enticing, given their complementary mechanisms.

Conclusion

Resmetirom and semaglutide are two of the most significant advances in drug treatment for MASH. Interestingly, these two drugs come from distinct physiological paradigms: resmetirom works through hepatocentric THR-β activation, while semaglutide works through systemic incretin-mediated weight loss. Current evidence suggests that resmetirom offers potent reductions in hepatic steatosis and shows an early signal towards fibrosis reversal, along with favorable lipid-lowering effects and minimal off-target effects. On the other hand, semaglutide provides unparalleled weight loss and reduction in cardiovascular risk, leading to high rates of resolution of steatohepatitis but limited direct antifibrotic efficacy. The safety profiles of both drugs are acceptable and largely predictable, although ongoing vigilance for long-term or rare events is necessary.

Instead of competing, the two agents may ultimately complement each other, either sequentially or in combination, similar to how statins and antihypertensives coexist in cardiovascular prevention. Integration into clinical practice will rely on forthcoming phase III data, cost-effectiveness analyses, and evolving guidelines. Currently, the future looks brighter than ever for patients with MASH, as resmetirom and semaglutide challenge the long-held belief that lifestyle modification is the only therapeutic option. Their divergent, yet potentially synergistic, mechanisms of action demonstrate the multifactorial approach that a complex metabolic liver disease requires.

Taken together, these data support the use of an individualized, multi-mechanistic treatment approach. Resmetirom and semaglutide, with their complementary actions in the liver and throughout the body, offer the first real chance to customize therapy based on patient characteristics. This includes individuals who are lean with dyslipidemia or severely obese with cardio-renal comorbidities. Ongoing trials, economic assessments, and biomarker-guided care pathways will help determine the most effective way to integrate these medications, either alone or in combination, into precise strategies aimed at stopping fibrosis progression and enhancing long-term survival.

Abbreviations

Aib: α-aminoisobutyric acid

ApoB: apolipoprotein B

CKD: chronic kidney disease

FDA: Food and Drug Administration

GI: gastrointestinal

GLP-1 RA: glucagon-like peptide-1 receptor agonist

HCC: hepatocellular carcinoma

LDL-C: low-density lipoprotein cholesterol

MASH: metabolic dysfunction-associated steatohepatitis

MRI-PDFF: magnetic resonance imaging-proton density fat fraction

NAFLD: nonalcoholic fatty liver disease

NASH: nonalcoholic steatohepatitis

T2DM: type 2 diabetes mellitus

THR-β: thyroid hormone receptor-β

Declarations

Disclaimer

This comparative perspective provides a summary of current evidence and should not be misconstrued as medical advice. Clinicians should refer to official prescribing information, regulatory updates, and conduct individualized patient assessments when making therapeutic decisions.

Author contributions

RW: Conceptualization, Methodology, Data curation, Supervision, Validation, Visualization, Writing—original draft, Writing—review & editing. The author read and approved the submitted version.

Conflicts of interest

Ralf Weiskirchen, who is the Associate Editor and Guest Editor of Exploration of Drug Science, had no involvement in the decision-making or the review process of this manuscript.

Ethical approval

Not applicable.

Consent to participate

Not applicable.

Consent to publication

Not applicable.

Availability of data and materials

Not applicable.

Funding

Not applicable.

Copyright

© The Author(s) 2025.

Publisher’s note

Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

References

Targher G, Valenti L, Byrne CD. Metabolic Dysfunction-Associated Steatotic Liver Disease. N Engl J Med. 2025;393:68398. [DOI] [PubMed]
Sanyal AJ, Husain M, Diab C, Mangla KK, Shoeb A, Lingvay I, et al. Cardiovascular disease in patients with metabolic dysfunction-associated steatohepatitis compared with metabolic dysfunction-associated steatotic liver disease and other liver diseases: A systematic review. Am Heart J Plus. 2024;41:100386. [DOI] [PubMed] [PMC]
Lonardo A. Resmetirom: Finally, the Light at the End of the NASH Tunnel? Livers. 2024;4:13841. [DOI]
Weiskirchen R, Lonardo A. How 'miracle' weight-loss semaglutide promises to change medicine but can we afford the expense? Br J Pharmacol. 2025;182:165170. [DOI] [PubMed]
Younossi ZM, Golabi P, Paik JM, Henry A, Dongen CV, Henry L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): a systematic review. Hepatology. 2023;77:133547. [DOI] [PubMed] [PMC]
Younossi ZM, Stepanova M, Ong J, Trimble G, AlQahtani S, Younossi I, et al. Nonalcoholic Steatohepatitis Is the Most Rapidly Increasing Indication for Liver Transplantation in the United States. Clin Gastroenterol Hepatol. 2021;19:5809.e5. [DOI] [PubMed]
Axley P, Ahmed Z, Arora S, Haas A, Kuo Y, Kamath PS, et al. NASH Is the Most Rapidly Growing Etiology for Acute-on-Chronic Liver Failure-Related Hospitalization and Disease Burden in the United States: A Population-Based Study. Liver Transpl. 2019;25:695705. [DOI] [PubMed]
Tilg H, Adolph TE, Moschen AR. Multiple Parallel Hits Hypothesis in Nonalcoholic Fatty Liver Disease: Revisited After a Decade. Hepatology. 2021;73:83342. [DOI] [PubMed] [PMC]
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;13:13448. [DOI] [PubMed]
Lonardo A, Zheng M, Weiskirchen R. Food insecurity is an emerging risk factor for liver disease: a scoping review. Expert Rev Gastroenterol Hepatol. 2025;19:103349. [DOI] [PubMed]
Lonardo A, Targher G. Not all fat is alike in MASLD. J Hepatol. 2025;82:e2712. [DOI] [PubMed]
Zheng H, Sechi LA, Navarese EP, Casu G, Vidili G. Metabolic dysfunction-associated steatotic liver disease and cardiovascular risk: a comprehensive review. Cardiovasc Diabetol. 2024;23:346. [DOI] [PubMed] [PMC]
Batta A, Hatwal J. Excess cardiovascular mortality in men with non-alcoholic fatty liver disease: A cause for concern! World J Cardiol. 2024;16:3804. [DOI] [PubMed] [PMC]
Desai R, Alvi AT, Vasavada A, Pulakurthi YS, Patel B, Mohammed AS, et al. Sex and racial disparities in non-alcoholic fatty liver disease-related cardiovascular events: National inpatient sample analysis (2019). World J Cardiol. 2024;16:13748. [DOI] [PubMed] [PMC]
FDA News Release. FDA Approves First Treatment for Patients with Liver Scarring Due to Fatty Liver Disease. [cited 2025 Sep 16]. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-patients-liver-scarring-due-fatty-liver-disease
Suvarna R, Shetty S, Pappachan JM. Efficacy and safety of Resmetirom, a selective thyroid hormone receptor-β agonist, in the treatment of metabolic dysfunction-associated steatotic liver disease (MASLD): a systematic review and meta-analysis. Sci Rep. 2024;14:19790. [DOI] [PubMed] [PMC]
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;390:497509. [DOI] [PubMed]
Kelly MJ, Pietranico-Cole S, Larigan JD, Haynes N, Reynolds CH, Scott N, et al. Discovery of 2-[3,5-dichloro-4-(5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yloxy)phenyl]-3,5-dioxo-2,3,4,5-tetrahydro[1,2,4]triazine-6-carbonitrile (MGL-3196), a Highly Selective Thyroid Hormone Receptor β agonist in clinical trials for the treatment of dyslipidemia. J Med Chem. 2014;57:391223. [DOI] [PubMed]
Liu W, Yao F, Wang J, Shao N, Cao X, Dong Z, et al. Combination Therapy of Half-Dose Resmetirom and Metformin Attenuates Metabolic Dysfunction-Associated Steatohepatitis Through Improving Cholesterol Metabolism and Inflammation. Biomedicines. 2025;13:1315. [DOI] [PubMed] [PMC]
Arvanitakis K, Koufakis T, Cholongitas E, Francque S, Germanidis G. Insights into the results of Resmetirom trials: Can a thyroid hormone receptor agonist be the holy grail of MASH therapy? Pharmacol Ther. 2025;268:108811. [DOI] [PubMed]
Li W, Zhang X, Song J, Yang L, Wang D, Yuan G, et al. Mechanistic insights into GLP-1 receptor agonist-induced weight loss through ceRNA network analysis. Genomics. 2025;117:110988. [DOI] [PubMed]
Johnson LM, Barrick S, Hager MV, McFedries A, Homan EA, Rabaglia ME, et al. A potent α/β-peptide analogue of GLP-1 with prolonged action in vivo. J Am Chem Soc. 2014;136:1284851. [DOI] [PubMed] [PMC]
Manandhar B, Ahn J. Glucagon-like peptide-1 (GLP-1) analogs: recent advances, new possibilities, and therapeutic implications. J Med Chem. 2015;58:102037. [DOI] [PubMed] [PMC]
Ezhilarasan D. Thyromimetics and MASLD: Unveiling the Novel Molecules Beyond Resmetirom. J Gastroenterol Hepatol. 2025;40:36778. [DOI] [PubMed]
Ezhilarasan D. Mechanism of Semaglutide in MASLD Treatment: Where Is the Master Key? J Gastroenterol Hepatol. 2025;40:216375. [DOI] [PubMed]
Harrison SA, Bashir MR, Guy CD, Zhou R, Moylan CA, Frias JP, et al. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2019;394:201224. [DOI] [PubMed]
Harrison SA, Calanna S, Cusi K, Linder M, Okanoue T, Ratziu V, et al. Semaglutide for the treatment of non-alcoholic steatohepatitis: Trial design and comparison of non-invasive biomarkers. Contemp Clin Trials. 2020;97:106174. [DOI] [PubMed]
Verma S, McGuire DK, Bain SC, Bhatt DL, Leiter LA, Mazer CD, et al. Effects of glucagon-like peptide-1 receptor agonists liraglutide and semaglutide on cardiovascular and renal outcomes across body mass index categories in type 2 diabetes: Results of the LEADER and SUSTAIN 6 trials. Diabetes Obes Metab. 2020;22:248792. [DOI] [PubMed] [PMC]
Mellbin LG, Bhatt DL, David J, Ekström K, Petrie MC, Rasmussen S, et al. Semaglutide and cardiovascular outcomes by baseline HbA1c in diabetes: the SUSTAIN 6 and PIONEER 6 trials. Eur Heart J. 2024;45:13714. [DOI] [PubMed] [PMC]
Schnell O, Almandoz J, Anderson L, Barnard-Kelly K, Battelino T, Blüher M, et al. CVOT summit report 2024: new cardiovascular, kidney, and metabolic outcomes. Cardiovasc Diabetol. 2025;24:187. [DOI] [PubMed] [PMC]
Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, et al.; NN9931-4296 Investigators. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N Engl J Med. 2021;384:111324. [DOI] [PubMed]
Dichtel LE. The Glucagon-Like Peptide-1 Receptor Agonist, Semaglutide, for the Treatment of Nonalcoholic Steatohepatitis. Hepatology. 2021;74:22902. [DOI] [PubMed] [PMC]
Newsome PN, Loomba R. Therapeutic horizons in metabolic dysfunction-associated steatohepatitis. J Clin Invest. 2025;135:e186425. [DOI] [PubMed] [PMC]
Ravela N, Shackelford P, Blessing N, Yoder L, Chalasani N, Samala N. Early experience with resmetirom to treat metabolic dysfunction-associated steatohepatitis with fibrosis in a real-world setting. Hepatol Commun. 2025;9:e0670. [DOI] [PubMed] [PMC]
Sodhi M, Rezaeianzadeh R, Kezouh A, Etminan M. Risk of Gastrointestinal Adverse Events Associated With Glucagon-Like Peptide-1 Receptor Agonists for Weight Loss. JAMA. 2023;330:17957. [DOI] [PubMed] [PMC]
Singh A, Sohal A, Batta A. GLP-1, GIP/GLP-1, and GCGR/GLP-1 receptor agonists: Novel therapeutic agents for metabolic dysfunction-associated steatohepatitis. World J Gastroenterol. 2024;30:520511. [DOI] [PubMed] [PMC]
López GP. GLP-1 receptor agonists and GIP/GLP-1 co-agonists in the treatment of obesity in adolescents and the elderly. Med Clin (Barc). 2025;165:107122. [DOI] [PubMed]
Marassi M, Fadini GP. Real-world Evidence on Oral Semaglutide for the Management of Type 2 Diabetes. A Narrative Review for Clinical Practice. Clin Ther. 2025;47:10210. [DOI] [PubMed]
Suri J, Borja S, Lim JK. Combination strategies for pharmacologic treatment of non-alcoholic steatohepatitis. World J Gastroenterol. 2022;28:512940. [DOI] [PubMed] [PMC]
Ayesh H, Beran A, Suhail S, Ayesh S, Niswender K. Comparative Analysis of Resmetirom vs. FGF21 Analogs vs. GLP-1 Agonists in MASLD and MASH: Network Meta-Analysis of Clinical Trials. Biomedicines. 2024;12:2328. [DOI] [PubMed] [PMC]
Koureta E, Cholongitas E. Evolving role of semaglutide in NAFLD: in combination, weekly and oral administration. Front Pharmacol. 2024;15:1343587. [DOI] [PubMed] [PMC]
Kim N, Wang J, Burudpakdee C, Song Y, Ramasamy A, Xie Y, et al. Cost-effectiveness analysis of semaglutide 2.4 mg for the treatment of adult patients with overweight and obesity in the United States. J Manag Care Spec Pharm. 2022;28:74052. [DOI] [PubMed] [PMC]
How much does semaglutide cost per month? [Internet]. SingleCare Administrators; c2025 [cited 2025 Sep 16]. Available from: https://www.singlecare.com/blog/semaglutide-cost-per-month/
Joszt L. FDA Approves Resmetirom, First Treatment for NASH With Liver Fibrosis [Internet]. MJH Life Sciences®; c2025 [cited 2025 Sep 16]. Available from: https://www.ajmc.com/view/fda-approves-resmetirom-first-treatment-for-nash-with-liver-fibrosis
Schon H, Weiskirchen R. Exercise-Induced Release of Pharmacologically Active Substances and Their Relevance for Therapy of Hepatic Injury. Front Pharmacol. 2016;7:283. [DOI] [PubMed] [PMC]
Tapper EB, Fleming C, Rendon A, Fernandes J, Johansen P, Augusto M, et al. The Burden of Nonalcoholic Steatohepatitis: A Systematic Review of Epidemiology Studies. Gastro Hep Adv. 2022;1:104987. [DOI] [PubMed] [PMC]
Younossi ZM, Corey KE, Lim JK. AGA Clinical Practice Update on Lifestyle Modification Using Diet and Exercise to Achieve Weight Loss in the Management of Nonalcoholic Fatty Liver Disease: Expert Review. Gastroenterology. 2021;160:9128. [DOI] [PubMed]
Chen F, Su W, Ramasamy A, Zvenyach T, Kahan S, Kyle T, et al. Ten-year Medicare budget impact of increased coverage for anti-obesity intervention. J Med Econ. 2019;22:1096104. [DOI] [PubMed]
Dietrich CG, Rau M, Geier A. Screening for nonalcoholic fatty liver disease-when, who and how? World J Gastroenterol. 2021;27:580321. [DOI] [PubMed] [PMC]
Weiskirchen S, Weiskirchen R. Unraveling the future: hot topics shaping molecular diagnostics today. Expert Rev Mol Diagn. 2025;25:1116. [DOI] [PubMed]
Castera L, Alazawi W, Bugianesi E, Caussy C, Federici M, Romero-Gómez M, et al. A European Survey to Identify Challenges in the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease. Liver Int. 2025;45:e16224. [DOI] [PubMed] [PMC]
Huang X, Wu M, Huang B, Zhang Y. Gastrointestinal adverse events associated with GLP-1 receptor agonists in metabolic dysfunction-associated steatotic liver disease (MASLD): a systematic review and meta-analysis. Front Med (Lausanne). 2025;12:1509947. [DOI] [PubMed] [PMC]
Lonardo A, Ballestri S, Mantovani A, Targher G, Bril F. Endpoints in NASH Clinical Trials: Are We Blind in One Eye? Metabolites. 2024;14:40. [DOI] [PubMed] [PMC]
Burra P, Zanetto A, Germani G. Sex bias in clinical trials in gastroenterology and hepatology. Nat Rev Gastroenterol Hepatol. 2022;19:4134. [DOI] [PubMed] [PMC]
Dileo E, Saba F, Parasiliti-Caprino M, Rosso C, Bugianesi E. Impact of Sexual Dimorphism on Therapy Response in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease: From Conventional and Nutritional Approaches to Emerging Therapies. Nutrients. 2025;17:477. [DOI] [PubMed] [PMC]
Moncho F, Benlloch S, Górriz JL. The impact of metabolic dysfunction-associated steatotic liver disease (MASH) on the high risk of cardiovascular disease in CKD: interconnections and management. Clin Kidney J. 2025;18:sfaf260. [DOI] [PubMed] [PMC]
Gallo G, Nalli G, Baratta F, Desideri G, Savoia C. Metabolic Dysfunction-Associated Steatotic Liver Disease: A Silent Driver of Cardiovascular Risk and a New Target for Intervention. Int J Mol Sci. 2025;26:8081. [DOI] [PubMed] [PMC]
Luna E, Álvarez Á, Rodriguez-Sabiñón J, Villa J, Giraldo T, Martín MV, et al. Long-Term Effect of Semaglutide on the Glomerular Filtration Rate Slope in High-Risk Patients with Diabetic Nephropathy: Analysis in Real-World Clinical Practice. Pharmaceutics. 2025;17:943. [DOI] [PubMed] [PMC]
Joseph J. Comparative Effectiveness of SGLT2 Inhibitors and Semaglutide in Diabetic Nephropathy: A Retrospective Observational Study. Cureus. 2025;17:e87399. [DOI] [PubMed] [PMC]
Lonardo A, Nascimbeni F, Ballestri S, Fairweather D, Win S, Than TA, et al. Sex Differences in Nonalcoholic Fatty Liver Disease: State of the Art and Identification of Research Gaps. Hepatology. 2019;70:145769. [DOI] [PubMed] [PMC]
Jamalinia M, Lonardo A, Weiskirchen R. Sex and Gender Differences in Liver Fibrosis: Pathomechanisms and Clinical Outcomes. Fibrosis. 2024;2:10006. [DOI]
Cite this Article
Export Citation
Weiskirchen R. Resmetirom and semaglutide in metabolic dysfunction-associated steatohepatitis (MASH): a comparative perspective. Explor Drug Sci. 2025;3:1008132. https://doi.org/10.37349/eds.2025.1008132
Article Metrics

View: 60

Download: 7

Times Cited: 0