Next Article in Journal
Salivary Metabolomics Discloses Metabolite Signatures of Oral Leukoplakia with and Without Dysplasia
Previous Article in Journal
Annexin–Membrane Interactions Across Eukaryotic Domains of Life—A Comparative Approach
Previous Article in Special Issue
Levels of Growth Differentiation Factor 15 Correlated with Metabolic Dysfunction-Associated Steatotic Liver Disease in Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacological Treatment of MASLD: Contemporary Treatment and Future Perspectives

by
Krzysztof Drygalski
Department of Hypertension and Diabetology, Medical University of Gdansk, ul. Mariana Smoluchowskiego 17, 80-214 Gdańsk, Poland
Int. J. Mol. Sci. 2025, 26(13), 6518; https://doi.org/10.3390/ijms26136518
Submission received: 18 May 2025 / Revised: 25 June 2025 / Accepted: 3 July 2025 / Published: 7 July 2025
(This article belongs to the Special Issue Steatotic Liver Disease: From Bench to Bedside and Back)

Abstract

Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly NAFLD, is the most prevalent chronic liver disease worldwide. Strongly linked to obesity, type 2 diabetes, and metabolic syndrome, MASLD poses a growing health burden. Despite its high prevalence and risk of progression, no pharmacological treatment is currently approved. This narrative review provides an overview of emerging pharmacological treatments under clinical investigation, with a particular focus on agents recently evaluated in randomized clinical trials. A systematic search of the ClinicalTrials.gov database through to April 2025 was conducted to identify relevant studies. Investigational drugs were categorized by their molecular mechanisms, and data on efficacy, safety, and clinical development phases were summarized. The most extensively studied drug classes include GLP-1 receptor agonists, PPAR agonists, and FXR agonists, as well as inhibitors of ACC and DGAT. These therapies have shown promising effects on hepatic steatosis, liver enzyme levels, and metabolic markers and may be introduced into clinical practice in the near future.
Keywords:
MASLD; NAFLD; MASH; NASH; treatment

1. Introduction

Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is the most common chronic liver disease worldwide, with an estimated prevalence affecting approximately 25–30% of the global population [1]. The prevalence is significantly higher among individuals with obesity, type 2 diabetes mellitus (T2DM), and other cardiometabolic risk factors constituting metabolic syndrome [1]. The recently redefined terminology from NAFLD to MASLD underscores the central role of metabolic dysfunction in disease pathogenesis, offering a clearer and more inclusive framework for diagnosis and management [2].
MASLD is characterized by hepatic steatosis, defined as triglyceride accumulation in more than 5% of hepatocytes, and may progress to steatohepatitis and hepatic fibrosis if left untreated. In a subset of patients, this progression can lead to cirrhosis and, more rarely, hepatocellular carcinoma (HCC) as a late-stage complication [2]. Nevertheless, hepatocyte steatosis remains the central point of the disease, affecting liver function through multiple molecular pathways [3]. The condition is typically asymptomatic in its early stages and frequently discovered incidentally during imaging studies or blood tests performed for unrelated reasons. This often leads to delayed diagnosis, by which time the steatosis has been silently progressing for months or years, subtly impairing aspects of metabolic homeostasis such as glycemic control and lipid profiles [4].
The pathophysiology of MASLD is complex and multifactorial. Insulin resistance is a central mechanism, promoting increased adipose tissue lipolysis and hepatic influx of free fatty acids. These metabolic alterations lead to hepatocellular lipid accumulation and subsequent lipotoxicity, oxidative stress, mitochondrial dysfunction, and inflammation [5,6,7]. On the other hand, lipid accumulation in hepatocytes itself promotes hepatic insulin resistance. Further disease progression from simple steatosis involves an interplay of various metabolic disturbances. This is best captured by the “multiple-hit” theory, which posits that liver steatosis and its progression to steatohepatitis, and fibrosis result from the cumulative, and often simultaneous, impact of several pathogenic factors (Figure 1) [8,9]. Beyond steatosis and lipotoxicity as the first “hit”, other “hits” include oxidative and endoplasmic reticulum stress, mitochondrial dysfunction, and alterations in the gut–liver axis—particularly dysbiosis and increased intestinal permeability that facilitate translocation of bacterial endotoxins [10,11]. These stimuli collectively activate hepatic immune responses, perpetuate chronic inflammation, and trigger fibrosis. Genetic predispositions like polymorphisms in PNPLA3 or TM6SF2 genes further modulate individual susceptibility and disease course [12,13]. This multifactorial model underscores the heterogeneity of MASLD and highlights the need for comprehensive management strategies that extend beyond simple lipid reduction to include modulation of inflammation and oxidative stress, body weight management, or glycemic control.
Aside from liver-specific consequences such as cirrhosis and HCC, MASLD carries significant systemic risks. It is now recognized as a major contributor to morbidity and mortality through its strong association with cardiovascular diseases—the leading cause of death worldwide—as well as obesity, T2DM, and chronic kidney disease [1,2]. In fact, MASLD is the leading cause of liver transplantation in women and the second leading cause, after alcohol-related liver disease, in men in the United States [14].
The diagnosis of MASLD is based on imaging evidence of hepatic steatosis in individuals with metabolic risk factors, following the exclusion of other potential causes of liver fat accumulation. Ultrasonography is the most widely used screening tool, owing to its broad availability and low cost; however, its sensitivity is limited, particularly in detecting mild steatosis. More advanced imaging techniques, such as transient elastography (e.g., FibroScan) and MRI-based modalities, provide greater sensitivity and the added benefit of assessing liver stiffness [2,15,16]. Although liver biopsy remains the gold standard for diagnosing steatohepatitis and staging fibrosis, its use is generally reserved for specific clinical situations due to its invasive nature. To assist in the non-invasive evaluation of liver fibrosis and guide treatment strategies, clinical scoring systems like FIB-4, along with transient elastography, have been developed to facilitate risk stratification (Figure 2) [2].
Given the high global burden, complex pathophysiology, and systemic impact of MASLD, effective therapeutic strategies and clear treatment recommendations are urgently needed. While lifestyle modification remains the cornerstone of management, long-term adherence is often challenging, and no pharmacological therapies have yet received widespread regulatory approval for MASLD. However, in March 2024 the FDA approved resmetirom, an oral thyroid hormone receptor-β (THR-β) agonist, in combination with diet and exercise for the treatment of adults with non-cirrhotic non-alcoholic steatohepatitis (NASH) with moderate to advanced liver fibrosis (consistent with stage F2 to F3 fibrosis) [17]. Nevertheless, there is still no approved treatment for MASLD/NAFLD itself.
In recent years, there has been growing interest in developing new drugs that target the key molecular mechanisms driving the disease. Numerous randomized clinical trials (RCT) have evaluated novel agents with various mechanisms of action, ranging from insulin sensitizers and lipid metabolism modulators to anti-inflammatory and antifibrotic compounds. In this review, I summarize the current treatment landscape of MASLD and critically assess recent clinical trial data to highlight emerging pharmacological options that may soon become part of routine clinical practice.

2. Contemporary Treatment Recommendations

Effective management of MASLD primarily targets the underlying metabolic disturbances driving disease progression. Lifestyle intervention—particularly achieving weight loss through tailored dietary adjustments and regular physical activity—remains the foundation of treatment [18]. Evidence shows that even modest reductions in body weight can lead to significant improvements in liver fat content, inflammation, and fibrosis. Importantly, the focus is not on rapid or drastic weight loss, but rather on a gradual, sustainable approach that promotes long-term health benefits. A steady reduction of approximately 0.5 to 1 kg per week is generally recommended, with an initial goal of losing 5–10% of baseline body weight over the course of three to six months [19,20]. This weight should ideally be maintained for a similar period before pursuing additional gradual reductions if necessary [13,16]. Despite the proven benefits, maintaining these lifestyle changes can be difficult in real-world settings, particularly among individuals with comorbidities [15]. Consequently, there has been growing interest in adjunct pharmacological treatments. The following sections will review the available evidence on physical activity, dietary approaches, and emerging pharmacotherapies in the treatment of MASLD.

3. Dietary Interventions in MASLD

Dietary modification remains a fundamental component in the management of MASLD, with strong evidence supporting its role in reducing hepatic steatosis, improving metabolic parameters, and preventing disease progression. The effect of dietary interventions and lifestyle changes in MASLD has been profoundly reviewed recently [21]. Given that excess caloric intake, insulin resistance, and poor diet quality contribute significantly to intrahepatic fat accumulation, dietary changes aimed at creating a caloric deficit are central to treatment. Most guidelines recommend a hypocaloric diet with a daily energy deficit of 500–1000 kcal, targeting gradual, sustainable weight loss [22,23]. A reduction of 5–10% in body weight over 3–6 months has been shown to yield significant improvements in liver fat, insulin sensitivity, and inflammation, with weight maintenance or further reduction as the next step.
Among the dietary patterns studied, the Mediterranean diet is the most consistently recommended by international guidelines (EASL-EASD-EASO, ESPEN, and APASL), owing to its high content of monounsaturated and polyunsaturated fats, fiber, antioxidants, and polyphenols [13,22,23,24]. It has demonstrated efficacy in reducing liver fat, improving glycemic control, and lowering cardiovascular risk [25,26]. Other dietary strategies with potential benefits include high-protein diets—especially those emphasizing plant-based proteins—and low-carbohydrate ketogenic diets, which may improve insulin sensitivity and reduce liver fat content [27,28,29]. Intermittent fasting, which focuses on meal timing rather than content, has also shown effects in reducing hepatic steatosis and improving metabolic markers, though comparative data with other diets are limited [30,31,32,33].
Ultimately, dietary treatment must be individualized to patient preferences, metabolic status, and comorbid conditions. The key to success lies in achieving and maintaining a negative energy balance through a sustainable, healthy diet. While the Mediterranean diet currently offers the strongest overall evidence base, flexible adaptation of its principles to accommodate individual needs appears to be the most practical and effective approach in clinical practice.

4. Physical Activity and Lifestyle Modifications in MASLD

Lifestyle modification, particularly increased physical activity, plays a pivotal role in the management of MASLD and, similarly to dietary interventions, is recommended as a first-line intervention by most clinical guidelines. Regular exercise has been shown to improve insulin sensitivity, reduce hepatic fat accumulation, and lower markers of inflammation and oxidative stress—key drivers in MASLD progression [34]. Aerobic exercise, such as brisk walking, cycling, or swimming, is especially effective at reducing visceral adiposity and intrahepatic triglyceride content, with benefits observed even in the absence of significant weight loss. Studies have demonstrated that physical activity alone can decrease liver fat by 2% to 50%, depending on intensity and duration [35,36,37]. The most commonly recommended regimen includes moderate-intensity aerobic activity for 150–200 min per week, complemented by resistance training to preserve lean muscle mass and enhance metabolic health.

5. Pharmacological Treatment

Despite lifestyle modification remaining the cornerstone of MASLD management, pharmacotherapy is increasingly recognized as a critical adjunct, especially in patients with biopsy-confirmed metabolic steatohepatitis (MASH), significant fibrosis (≥F2), or coexisting cardiometabolic comorbidities. Several agents have demonstrated histological and biochemical effects by targeting core pathophysiological pathways: oxidative stress, insulin resistance, lipotoxicity, and inflammation. Although no drug is currently globally approved specifically for MASLD, some drugs are locally recommended or used off-label in patients diagnosed with liver steatosis (Table 1).
  • Pioglitazone (30–45 mg/day)
Pioglitazone, a thiazolidinedione, acts as a selective peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist. It promotes adipocyte differentiation and enhances peripheral insulin sensitivity, thereby reducing the flux of free fatty acids (FFAs) to the liver. Pioglitazone also decreases hepatic de novo lipogenesis and inflammatory cytokine expression while increasing adiponectin levels, which has anti-inflammatory and insulin-sensitizing properties. RCTs, including PIVENS, have shown histological improvements in steatosis, lobular inflammation, and hepatocellular ballooning in patients with MASH, especially those with T2DM [13,15,38,39,40]. However, its use may be limited by adverse effects such as weight gain and fluid retention [38,39,40].
2. 
Vitamin E (800 IU/day, d-α-tocopherol)
Vitamin E is a lipid-soluble antioxidant that reduces oxidative damage within hepatocytes, a key driver in the progression from steatosis to steatohepatitis [15]. It neutralizes reactive oxygen species (ROS) and downregulates pro-inflammatory signaling pathways. The PIVENS trial demonstrated significant improvement in steatohepatitis but not fibrosis among non-diabetic adults with biopsy-proven NASH [38]. Despite moderate improvements in transaminases and histology, concerns remain about long-term safety, including a possible increased risk of prostate cancer in older men and hemorrhagic stroke [41,42]. Thus, it is not recommended in patients with diabetes or advanced cirrhosis.
3. 
Ursodeoxycholic Acid (UDCA, 13–15 mg/kg/day)
UDCA is a hydrophilic bile acid with cytoprotective, anti-apoptotic, and anti-inflammatory properties. It stabilizes hepatocyte membranes, reduces hepatic transaminase levels in serum, and protects hepatocytes from oxidative stress [43,44,45]. While extensively used in cholestatic liver diseases, evidence for its efficacy in MASLD is limited and inconclusive. Some small trials have reported improvements in liver enzymes and steatosis, but histological benefits are uncertain [45,46,47,48]. It is worth noting that most of the evidence for UDCA use comes from patients with NASH or cholestatic liver diseases.

6. Review of New Drugs Currently Tested in RCTs

In recent years, a growing number of investigational drugs have been studied, targeting various metabolic pathways involved in MASLD pathogenesis. The following section presents a review of contemporary RCTs, primarily phase 2 studies, evaluating the efficacy and safety of these novel therapeutic agents. The ClinicalTrials.gov database was searched up to April 2025 for clinical trials reporting a reduction in liver steatosis with any drugs used in the treatment of NAFLD/MASLD. Experimental therapies were grouped by their molecular mechanisms of action (Table 2, Figure 3).

7. Acetyl-CoA Carboxylase (ACC) Inhibitors

Acetyl-CoA carboxylase (ACC) plays a pivotal role in hepatic lipid metabolism by catalyzing the conversion of acetyl-CoA to malonyl-CoA, the first step in de novo lipogenesis. This process contributes to hepatic triglyceride accumulation, a hallmark of MASLD. ACC exists in two isoforms: ACC1, primarily involved in lipogenesis, and ACC2, which regulates fatty acid oxidation by inhibiting carnitine palmitoyltransferase 1 (CPT1). Inhibition of ACC reduces de novo lipogenesis and may enhance fatty acid oxidation, thereby decreasing hepatic steatosis [49,94].
In the NCT01431521 study, MK-4074, a dual ACC1/ACC2 inhibitor, administered at 200 mg twice daily for four weeks, resulted in a −36% reduction in intrahepatic fat, significantly outperforming pioglitazone [49]. The treatment was well tolerated and did not increase the rate of adverse events, although it had no effect on ALT or AST levels. Another ACC inhibitor, PF-05221304, was evaluated in several trials (NCT03248882, NCT03776175, and NCT04399538), either as monotherapy or in combination with diacylglycerol acyltransferases 2 (DGAT2) inhibitor PF-06865571 [50,51,52]. PF-05221304 showed a dose-dependent reduction in liver fat content—up to −40% in some regimens—and was also associated with reductions in ALT. However, its use led to elevations in serum triglycerides, a known adverse effect of ACC inhibition. Importantly, combining PF-05221304 with a DGAT2 inhibitor, PF-06865571, did not mitigate this triglyceride elevation. These findings highlight both the therapeutic potential and limitations of ACC inhibitors in MASLD, emphasizing the need for strategies that can offset the associated metabolic side effects.

8. Amino Acids, Carnitine, and N-Acetylcysteine

N-acetylcysteine and carnitine have been proposed as therapeutic agents in MASLD due to their roles in mitochondrial function, oxidative stress reduction, and metabolic regulation. N-acetylcysteine replenishes intracellular glutathione stores, reducing oxidative stress—a key driver of hepatocellular injury in MASLD [95,96]. Carnitine, on the other hand, is essential for mitochondrial fatty acid transport and β-oxidation, may enhance lipid metabolism, and mitigates hepatic steatosis [97,98,99].
These concepts were tested in the phase 2 clinical trial NCT04073368, which evaluated the safety and efficacy of two investigational amino acid-based formulations—AXA1125 and AXA1957—in patients with NASH [53]. AXA1125 includes leucine, isoleucine, valine, arginine, glutamine, and N-acetylcysteine, while AXA1957 contains leucine, isoleucine, arginine, glutamine, serine, and carnitine. Both formulations were designed to reduce inflammation and restore mitochondrial function. However, the addition of branched amino acids raises some concerns about the molecular effects of the tested formulations, since both leucine and isoleucine restriction were reported to ameliorate insulin resistance and NAFLD [100,101].
Nevertheless, over the treatment period, both AXA1125 and AXA1957 led to reductions in hepatic steatosis of −22.9% and −20.3%, respectively, and serum ALT levels of −21.9% and −20.6%, respectively, indicating a favorable impact on liver fat content and hepatocellular injury. These effects support the hypothesis that N-acetylcysteine and carnitine supplementation may help modulate oxidative stress and β-oxidation implicated in MASLD pathogenesis.

9. AMPK Activators

AMP-activated protein kinase (AMPK) serves as a central regulator of cellular energy homeostasis, influencing pathways related to lipid metabolism, glucose uptake, and inflammation [102,103]. Out of many downstream effects of AMPK activation, the most important in MASLD are inhibition of ACC, which promotes ß-oxidation; inhibition of sterol regulatory element-binding protein 1 (SREBP-1c), which downregulates fatty acid synthase (FAS) and de novo lipogenesis; and inhibition of β-Hydroxy β-methylglutaryl-CoA (HMG-CoA) reductase, which reduces cholesterol synthesis and promotion of glucose transporter 1 and 4 translocation towards cell membranes. Thus, AMPK activation has been proposed as a therapeutic strategy to mitigate hepatic steatosis and improve insulin sensitivity.
In the STAMP-NAFLD trial (NCT03763877), the direct AMPK activator PXL770 was evaluated in a randomized, double-blind, placebo-controlled phase 2a study involving 120 patients with hepatic steatosis [54]. Over 12 weeks of treatment, patients receiving PXL770 at 250 mg twice daily and 500 mg once daily achieved statistically significant reductions in liver fat content of −14.3% and −14.7%, respectively, as well as significant improvements in ALT, AST, HbA1c, and FIB-4 scores. These metabolic and hepatic benefits were observed without significant effects on lipid profiles or body weight, and the treatment was well tolerated. Separately, the NCT00063232 study evaluated metformin, an indirect AMPK activator, in patients with biopsy-confirmed NASH [55]. Treatment with 1 g of metformin twice daily for 48 weeks resulted in significant reductions in histological NASH activity, increased the number of patients with normal ALT levels, and improved insulin resistance as measured by HOMA-IR (−3.3 U). Collectively, these findings underscore the potential of AMPK-targeted therapies in MASLD, both in improving hepatic steatosis and modulating systemic metabolic dysfunction.

10. Bile Acid Metabolism

Bile acids are not only essential for lipid digestion but also act as signaling molecules regulating metabolic pathways through receptors like the farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5) [4,104]. Disruptions in bile acid homeostasis can contribute to hepatic steatosis, inflammation, and fibrosis. FXR activation in the liver suppresses lipogenic genes via indirect SREBP-1c inhibition and promotes fatty acid oxidation via PPARα, while in the intestine it regulates fibroblast growth factor 19 (FGF19), a hormone involved in bile acid synthesis feedback [105,106]. Although ileal bile acid transporter (IBAT) inhibition may reduce intestinal FXR signaling, increased hepatic bile acid production could enhance hepatic FXR activity, supporting improved lipid and glucose metabolism [107]. Additionally, elevated bile acid levels in the colon stimulate TGR5 receptors on enteroendocrine L-cells, leading to increased secretion of glucagon-like peptide-1 (GLP-1), which may provide additional effects in treatments targeting bile acid metabolism in MASLD [108].
Therapeutic strategies targeting bile acid metabolism, such as stearoyl-CoA desaturase 1 (SCD1) inhibition by aramchol and IBAT inhibition by elobixibat, aim to restore metabolic balance and mitigate liver damage in MASLD [109]. In the phase 2b ARREST trial (NCT02279524), 247 patients with biopsy-confirmed NASH received 400 mg or 600 mg of aramchol or placebo daily for 52 weeks [56]. Although the primary endpoint—a significant placebo-corrected reduction in liver triglyceride content—was narrowly missed (−3.1%; p = 0.066), notable secondary outcomes included NASH resolution without fibrosis worsening in 16.7% of patients in the 600 mg group versus 5% in the placebo group and fibrosis improvement in −29.5% compared to −17.5%. Aramchol also led to reductions in ALT (−17.3 U/L), AST (−10.8 U/L), and HbA1c (−0.13% points) and was well tolerated, supporting its role in modulating both hepatic inflammation and metabolic dysfunction.
Elobixibat (NCT04006145), by contrast, targets the IBAT disrupting enterohepatic circulation of bile acids and promoting hepatic bile acid synthesis. In a 16-week phase 2 trial involving 47 patients with NAFLD or NASH, daily elobixibat (5 mg) significantly lowered LDL cholesterol levels by −22 mg/dL on average. Despite the fact that the study did not assess its effect on liver steatosis, it supports the idea of targeting bile acid metabolism as a potential pharmacodynamic target in MASLD.

11. Diacylglycerol Acyltransferase Inhibitors

DGAT1 and DGAT2 catalyze the final step in triglyceride synthesis by esterifying diacylglycerol with acyl-CoA. DGAT2 is primarily active in hepatocytes, contributing to de novo lipogenesis and hepatic triglyceride accumulation, while DGAT1 functions in the intestine and peripheral tissues, facilitating dietary fat absorption and chylomicron creation. In MASLD, excessive activity of these enzymes leads to hepatic steatosis, lipotoxicity, and progression to steatohepatitis and fibrosis [110,111,112].
The NCT01811472 trial evaluated LCQ908, a DGAT1 inhibitor, in patients with hepatic steatosis and elevated triglycerides. Treatment led to a reduction in liver fat content of −2.9% and reduced ALT by −9.1 U/L, body weight by −2.5 kg, and waist circumference by −4.2 cm, while having no effect on AST, GGT, or serum triglyceride levels. The drug had a safety profile similar to placebo but was characterized by higher incident levels of diarrhea and abdominal pain.
In contrast, the DGAT2 inhibitor PF-06865571 was studied in NCT04399538 in combination with the ACC inhibitor clesacostat [52]. This trial included patients with a BMI between 25 and 40 kg/m2 and features of metabolic syndrome. The combination therapy resulted in a reduction in hepatic steatosis of up −60.3% but was associated with an increase in serum triglycerides of up to +27.9% and adverse events such as mild gastrointestinal disturbances and iron deficiency. Similarly, in the NCT03776175 trial, PF-06865571—administered alone or in combination with clesacostat—led to reductions in liver fat content of up to −40.1% in overweight or obese individuals [50].

12. Fatty Acid Synthase Inhibitors

Fatty acid synthase (FAS) is a key enzyme in the de novo lipogenesis pathway, responsible for the synthesis of palmitate. In MASLD, upregulated de novo lipogenesis contributes to hepatic steatosis and further lipotoxicity and inflammation. By inhibiting FAS, TVB-2640 reduces palmitate production, thereby decreasing the accumulation of toxic lipid species such as diacylglycerols and ceramides. This reduction in lipotoxic intermediates can alleviate hepatocellular stress, diminish inflammatory responses, and impede the activation of hepatic stellate cells, which are central to fibrogenesis [113,114].
The FASCINATE-1 trial (NCT03938246) evaluated TVB-2640, a first-in-class oral FASN inhibitor, in patients with NAFLD and stage 1–3 fibrosis [57]. In this 12-week phase 2a trial, patients with ≥8% liver fat and a BMI ≤ 40 kg/m2 received TVB-2640 at 25 mg or 50 mg daily or placebo. TVB-2640 significantly reduced liver fat in a dose-dependent manner, with the 50 mg group achieving a −28.1% reduction versus a +4.5% increase in the placebo group. Additionally, 61% of patients in the 50 mg group achieved a ≥30% liver fat reduction. The drug also decreased ALT levels by −20.5% and was well tolerated, with only mild adverse events reported.

13. Free Fatty Acid Receptor 4 Agonist

FFAR4, also known as GPR120, is a G-protein-coupled receptor activated by long-chain fatty acids, including omega-3 fatty acids. Activation of FFAR4 has been shown to exert anti-inflammatory effects by modulating macrophage polarization towards an anti-inflammatory M2 phenotype and inhibiting pro-inflammatory signaling pathways. In the context of MASLD, FFAR4 activation may reduce hepatic inflammation and fibrosis by attenuating inflammatory responses and improving insulin sensitivity [115,116].
The ICONA trial (NCT04052516) evaluated icosabutate, an oral free fatty acid receptor 1 and 4 (FFAR1/FFAR4) agonist, in patients with biopsy-confirmed MASH and fibrosis stages F1–F3 [58]. In this phase 2b study, 187 patients were randomized to receive 300 mg, 600 mg, or placebo daily for 52 weeks. Although the trial did not meet its primary endpoint—MASH resolution without fibrosis worsening—the 600 mg group showed a higher response rate than the placebo group (23.9% vs. 14.5%); however, the change was not statistically significant. Secondary outcomes included significant reductions in ALT (−30.1 U/L), AST (−18.5 U/L), GGT (−32.7 U/L), HOMA-IR (−3.8 U), bilirubin (−1.5 µM/L), hsCRP (−3.4 mg/L), and the fibrosis marker PRO-C3 (−11.8 µg/L). Fibrosis improvement of at least one stage occurred in 29.3% and 23.9% of patients receiving 300 mg and 600 mg, respectively, compared to 11.3% with placebo. However, no reduction in liver fat content was observed. These results suggest that while FFAR agonism may not directly reduce steatosis, it may provide antifibrotic and anti-inflammatory benefits in MASLD.

14. Fibroblast Growth Factor 21 Signaling

FGF21 is an endocrine hormone predominantly produced by the liver, playing a crucial role in regulating glucose and lipid metabolism. It exerts its effects by binding to fibroblast growth factor receptors (FGFRs), particularly FGFR1c, in complex with the co-receptor β-Klotho (KLB). Activation of this signaling pathway enhances insulin sensitivity, promotes fatty acid oxidation, reduces lipogenesis, and exerts anti-inflammatory effects [117,118].
The NCT03976401 trial assessed efruxifermin, a long-acting FGF21 analog, in patients with a BMI > 25 kg/m2 and hepatic fat content ≥ 10% [59,60,61]. Efruxifermin treatment led to a significant reduction in hepatic steatosis of −14.1% and reduced ALT levels by −32.3 U/L, liver stiffness by −37.6 kPa, and fibrosis biomarkers such as pro-C3 by −8.4 µg/L. However, gastrointestinal adverse events, including nausea, vomiting, and diarrhea, were reported. Another FGF21 analog, pegbelfermin (BMS-986036), was investigated in the NCT02413372 phase 2 trial involving patients with a BMI ≥ 25 kg/m2 [62,63]. Over a 16-week period, daily 10 mg or weekly 20 mg subcutaneous injections significantly reduced hepatic fat content (−6.8%) compared to placebo (−1.3%). The treatment was well tolerated, with only mild gastrointestinal symptoms reported. In contrast, NCT04583423 evaluated MK-3655, a humanized monoclonal antibody that activates FGFR1c/β-Klotho signaling by binding KLB [64]. This study included patients with NASH and a BMI between 25 and 50 kg/m2 but failed to demonstrate a significant therapeutic effect. Despite MK-3655’s lack of benefit, FGF21 analogs like efruxifermin and pegbelfermin show potential in improving steatosis, inflammation, and fibrosis in MASLD.

15. Farnesoid X Receptor Agonists

FXR is a nuclear receptor activated by bile acids, playing a pivotal role in regulating bile acid synthesis, lipid and glucose metabolism, and inflammatory response. FXR promotes ß-oxidation via PPARα activation and suppresses de novo lipogenesis via SREBP-1c inhibition by a small heterodimer partner (SHP) acting as a mediator. In the context of MASLD, FXR activation has been shown to reduce hepatic steatosis, inflammation, and fibrosis, making FXR agonists promising therapeutic agents [4,104].
Obeticholic acid (OCA), the most extensively studied FXR agonist, was evaluated in the FLINT trial (NCT01265498), where non-cirrhotic NASH patients receiving 25 mg daily for 72 weeks experienced notable improvements in liver histology, including reductions in steatosis, inflammation, and fibrosis [65,66,67,68]. Biochemical improvements included decreases in ALT (−38 U/L) and AST (−27 U/L). However, treatment was associated with pruritus and increases in alkaline phosphatase, HOMA-IR, and total and LDL cholesterol, without significant effects on triglycerides, HbA1c, waist circumference, or blood pressure. A separate phase 2 study (NCT00501592) in patients with NAFLD and T2DM demonstrated that 25–50 mg of OCA daily for six weeks reduced ALT (−9 U/L) and GGT (−39 U/L) [69].
Other FXR agonists under investigation include tropifexor, vonafexor, and EDP-305. Tropifexor (NCT04328077) showed dose-dependent reductions in ALT of up to −18%. Vonafexor (NCT03812029), evaluated in the LIVIFY trial, led to reductions in liver fat of up to −6.3% and reduced ALT by −16.3 U/L, GGT by −40.6 U/L, weight by up to −2.5 kg, and waist circumference by up to −2.2 cm [70]. EDP-305, a non-bile acid FXR agonist, at a 2.5 mg dose (NCT03421431) led to significant reductions in ALT of up to −26.1 U/L and liver fat of up to −6.4% [71]. Notably, EDP-305 also decreased ApoA1 levels, though it had no significant effects on lipid profiles or glycemic control.
Collectively, these studies highlight the capacity of FXR agonists to improve liver enzymes, steatosis, and fibrosis in MASLD. However, the consistent occurrence of pruritus and, in some cases, unfavorable changes in lipid profiles underscore the need for optimized dosing strategies and long-term evaluation to ensure both efficacy and tolerability.

16. GLP-1 and Dual GLP-1/GIP Agonists

GLP-1 receptor agonists enhance insulin secretion, suppress glucagon release, slow gastric emptying, and promote satiety, leading to weight loss and improved glycemic control. These effects contribute to reduced hepatic steatosis and inflammation and may provide a therapeutic alternative for MASLD patients [119,120].
Semaglutide, a GLP-1 receptor agonist, was evaluated in several phase 2 trials. In the NCT02970942 trial, patients with biopsy-proven NASH and fibrosis stages F1–F3 received daily subcutaneous injections for 72 weeks [74]. The highest dose (0.4 mg/day) led to NASH resolution without worsening of fibrosis in 59% of patients compared to 17% with placebo, along with significant reductions in steatosis scores, ALT (up to −60%), AST (up to −50%), GGT (up to −52%), HbA1c (up to 1.2% points) and body weight (up to −12.3 kg). However, in NCT03987451, where semaglutide was tested at a higher dose of 2.4 mg weekly in patients with compensated cirrhosis, it failed to produce significant improvements in fibrosis or NASH resolution, though beneficial effects on liver fat (−38%), liver enzymes, HbA1c (−1.4% points), body weight (−8.6 kg), and BMI (−3.1 units) were still observed [72,73].
Other trials reinforced the metabolic effects of semaglutide. In NCT04216589, conducted in patients with NAFLD and HIV, semaglutide led to reductions in steatosis (−4.2%), HbA1c, weight (−7.8 kg), BMI (−2.8 units), and waist circumference (−6.7 cm) [75]. Similarly, NCT04944992 demonstrated improvements in hepatic fat content (−42.3%), weight (−7.1 kg), and lipid parameters—including reductions in total cholesterol (−8.0%), LDL (−6.9%), triglycerides (−23.3%), and apoB (−9.8%), alongside an increase in HDL (+3.6%)—in overweight and obese patients with NAFLD [76].
In contrast, the SYNERGY-NASH trial (NCT04166773) investigated tirzepatide, a dual GLP-1/GIP receptor agonist, in patients with biopsy-confirmed MASH and fibrosis stages F2–F3 [77]. Weekly administration of tirzepatide at doses of 5, 10, and 15 mg for 52 weeks led to MASH resolution without worsening of fibrosis in up to 62% of patients and fibrosis improvement in over 50% of cases across all dose groups—substantially outperforming placebo. Tirzepatide also led to reductions in steatosis (−11.3%) and body weight (−17.9 kg).
Altogether, these studies illustrate that GLP-1 receptor agonists like semaglutide consistently improve steatosis, metabolic markers, and liver enzymes in MASLD, although effects on fibrosis may be more limited, particularly in advanced disease stages. Tirzepatide, through combined GLP-1 and GIP receptor activity, appears to offer superior efficacy in both NASH resolution and fibrosis regression, underscoring the potential of dual incretin-based therapies in MASLD management.

17. Dual GLP-1/Glucagon Receptor Agonists

Glucagon receptor activation plays a multifaceted role in hepatic metabolism. It promotes lipolysis, enhances basal energy expenditure, and influences liver lipid metabolism [121,122,123]. In the context of MASLD, glucagon receptor agonism may complement GLP-1 receptor agonism by directly stimulating hepatic fatty acid oxidation and reducing lipogenesis, leading to decreased hepatic steatosis. Moreover, glucagon receptor activation may attenuate hepatic inflammation and fibrosis, further contributing to its therapeutic potential in MASLD.
Dual GLP-1 and glucagon receptor agonists are an emerging class of therapeutics in MASLD that combine the metabolic benefits of GLP-1 receptor activation with the lipid-lowering and energy-expending effects of glucagon signaling. In the phase 2a trial NCT04944992, efinopegdutide was compared to semaglutide in patients with NAFLD [76]. Administered once weekly at a dose of 10 mg for 24 weeks, efinopegdutide produced a 72.7% reduction in liver fat content—significantly greater than the 42.3% reduction observed with semaglutide. In addition to reducing steatosis and body weight (−8.5 kg), efinopegdutide also led to decreases in total (−15.2%), LDL (−13.0 %), and HDL cholesterol (−8.1%); triglycerides (−30.9%); and apolipoprotein B (−14.7 %). These lipid-modifying effects suggest that glucagon receptor agonism enhances hepatic lipid metabolism beyond what is achieved with GLP-1 agonism alone.
In the NCT04771273 trial, survodutide—another dual GLP-1/glucagon receptor agonist—was evaluated in patients with biopsy-proven MASH and fibrosis stages F1–F3 [78]. After 48 weeks of treatment, survodutide was superior to placebo in achieving MASH resolution without fibrosis progression and demonstrating meaningful improvements in liver fat content (−13 %). The third agent in this class, cotadutide, was assessed in the NCT04019561 trial involving patients with NAFLD or NASH and fibrosis stages F1–F3 [79]. Over 19 weeks, cotadutide significantly reduced hepatic steatosis (−4.8%), ALT (−15.5%), AST (−14.0%), body weight (−2.9 kg), and BMI (−1.0 units).
Altogether, these trials highlight the potential of dual GLP-1/glucagon receptor agonists in treating MASLD through complementary mechanisms that reduce liver fat, improve cardiometabolic markers, and address liver inflammation. Their robust metabolic effects, alongside preliminary histological benefits, position this class as a promising option for future MASLD therapy.

18. Ketohexokinase Inhibition

Ketohexokinase (KHK), also known as fructokinase, is the key enzyme responsible for phosphorylating fructose to fructose-1-phosphate. The process promotes de novo lipogenesis and contributes to hepatic fat accumulation. In the context of MASLD, excessive dietary fructose intake and elevated hepatic KHK activity have been linked to increased triglyceride synthesis, steatosis, and insulin resistance [124,125]. Inhibiting KHK may therefore reduce hepatic lipid accumulation and improve metabolic parameters [126].
The phase 2a trials NCT03969719 and NCT03256526 evaluated PF-06835919, an oral KHK inhibitor, in 164 adults with NAFLD and T2DM [80,81]. Participants were randomized to receive 150 mg, 300 mg, or placebo daily for 16 weeks. Treatment with PF-06835919 led to dose-dependent reductions in liver fat content, with a 19% reduction in the 300 mg group and 17% in the 150 mg group, compared to only 5% in the placebo group. ALT levels also decreased, supporting a hepatoprotective effect. Although both active treatment arms showed significant reductions in HbA1c from baseline, these were not statistically significant versus placebo, and no significant changes were observed in HOMA-IR, suggesting a limited effect on insulin sensitivity. The drug was well tolerated, with mostly mild gastrointestinal symptoms and no serious adverse events reported.

19. Mitochondrial Uncoupling

Mitochondria serve as the principal sites of cellular energy production, generating adenosine triphosphate (ATP) via oxidative phosphorylation. This process involves the transfer of electrons through the mitochondrial electron transport chain (ETC), which drives the translocation of protons across the inner mitochondrial membrane, thereby establishing an electrochemical proton gradient. ATP synthase subsequently utilizes this gradient to catalyze the phosphorylation of ADP to ATP. Mitochondrial uncoupling refers to the dissipation of the proton gradient independent of ATP synthesis, typically through proton leak pathways that permit the re-entry of protons into the mitochondrial matrix. This uncoupling process reduces the efficiency of oxidative phosphorylation, resulting in increased substrate oxidation and energy expenditure, with excess energy released as heat. These effects can contribute to decreased lipid accumulation and improved metabolic homeostasis [127,128].
HU6, a controlled-release oral prodrug of 2,4-dinitrophenol (DNP), exerts its effects by selectively and mildly uncoupling oxidative phosphorylation. Unlike old uncouplers, which pose significant toxicity risks due to uncontrolled mitochondrial disruption, HU6 is designed to achieve a wider therapeutic window of uncoupling that safely enhances mitochondrial energy dissipation [129,130].
The phase 2a clinical trial NCT04874233 evaluated HU6, an oral prodrug of the mitochondrial uncoupler 2,4-dinitrophenol (DNP), in individuals with NAFLD [82]. In the study, 80 adults with a BMI of 28–45 kg/m2 and ≥8% liver fat were randomized to receive daily oral doses of HU6 (150, 300, or 450 mg) or placebo for 61 days [82]. HU6 significantly reduced liver fat content at all doses, with mean relative reductions of −26.8% (150 mg), −35.6% (300 mg), and −33.0% (450 mg), compared to a +5.4% increase in the placebo group.
These findings indicate that mitochondrial uncoupling via HU6 can effectively reduce hepatic steatosis and support weight loss in patients with MASLD. However, the treatment was associated with a slightly higher incidence of diarrhea and flushing.

20. Nonsteroidal Anti-Inflammatory Drugs

Acetylsalicylic acid (ASA)’s beneficial effects in MASLD are attributed to its anti-inflammatory and antiplatelet properties. It irreversibly inhibits cyclooxygenase enzymes (COX-1 and COX-2), leading to decreased synthesis of pro-inflammatory prostaglandins and thromboxanes. This inhibition reduces hepatic inflammation, which is implicated in the progression of liver steatosis to steatohepatitis and fibrosis. Furthermore, ASA has been shown to suppress the transforming growth factor-beta1 (TGF-β1)/Smad signaling pathway, which plays a crucial role in hepatic fibrogenesis. By attenuating this pathway, ASA may help prevent the development of liver fibrosis [131,132,133,134].
The phase 2 clinical trial NCT04031729 evaluated the efficacy of low-dose ASA (81 mg daily) over six months in adults with MASLD without cirrhosis [83]. This randomized, double-blind, placebo-controlled study demonstrated that ASA significantly reduced hepatic fat content compared to placebo. Specifically, participants receiving it experienced a mean absolute reduction in liver fat of −6.6%, whereas the placebo group showed a mean increase of +3.6%. Additionally, 43% of ASA-treated individuals achieved at least a 30% relative reduction in liver fat, compared to 13% in the placebo group.

21. PPAR Agonists

PPARs are nuclear hormone receptors that regulate gene expression involved in lipid metabolism and glucose homeostasis [13,38,39,40]. There are three isoforms: PPAR-α, PPAR-γ, and PPAR-δ. PPARs are key transcription factors that coordinate hepatic metabolism in response to nutritional cues. Among them, PPARα plays a central role in adapting liver function during feeding and fasting transitions. In the fasted state, PPARα is activated by fatty acids released from adipose tissue and promotes their hepatic uptake and mitochondrial β-oxidation, partly through transcriptional activation of transport proteins (e.g., CD36 and SLC27A1) and enzymes such as CPT1A. This pathway supports energy production and ketogenesis, while also inducing the hepatokine FGF21, which enhances systemic glucose and lipid metabolism. Additionally, PPARα activates autophagy (notably lipophagy), helping maintain energy balance. During the fed state, PPARα contributes to triglyceride synthesis by promoting acetyl-CoA transport and activating lipogenic regulators like SREBP1C. PPARγ and PPARδ also contribute, with PPARγ enhancing lipid storage via mTORC1 signaling and PPARδ boosting β-oxidation and HDL formation. Together, PPARs orchestrate lipid and glucose metabolism to maintain hepatic and systemic energy homeostasis.
Lanifibranor, a pan-PPAR agonist activating α, γ, and δ isoforms, was investigated in two trials (NCT03008070 and NCT03459079) [88,89,90,91]. In the phase 2b NATIVE trial (NCT03008070), 49% of patients receiving 1200 mg daily achieved NASH resolution without worsening fibrosis, compared to 27% with placebo, and 55% showed at least one-stage fibrosis improvement. Lanifibranor also significantly reduced ALT (−26.1 U/L), AST (−15.1 U/L), hsCRP (−2.0 mg/L), HOMA-IR (−5.8 units), HbA1c (−0.4% points), and triglycerides (−43.4 mg/dL). In the NCT03459079 trial, the drug additionally reduced hepatic steatosis (−8.7%) and HbA1c (−0.7% points), while increasing HDL cholesterol by +7.6 mg/dL.
Saroglitazar, a dual PPAR-α/γ agonist, was studied in the trials NCT03061721 and NCT03863574 [86,87]. In the NCT03061721, daily 4 mg dosing significantly reduced liver fat content (−4.2%), ALT levels (−44.9%), and fibrosis scores. The drug was characterized by a favorable safety profile. These findings were corroborated by NCT03863574, where saroglitazar led to reductions in ALT (−29.6 U/L), AST (−16.3 U/L), and GGT (−28.4 U/L).
Finally, dR-pioglitazone, a deuterium-stabilized R-stereoisomer of pioglitazone and selective PPAR-γ agonist, was evaluated in NCT04321343. This study demonstrated reductions in fibrosis scores, hepatic steatosis (up to −21.3%), ALT (up to −13.6 U/L), AST (up to −10.2 U/L), GGT (up to −21.8 U/L), HbA1c (up to −0.2% points), and HOMA-IR (up to −0.21 units), highlighting its potential for targeting insulin resistance and hepatic injury in patients with biopsy-proven MASLD [84,85].

22. SGLT1 and SGLT2 Inhibitors

Sodium-glucose co-transporter 1 and 2 (SGLT1/2) are key glucose transporters in the intestine and kidneys, and molecular targets of antidiabetic drugs, flozins. SGLT1 inhibition in the intestine reduces postprandial glucose absorption, while SGLT2 inhibition in the kidneys promotes urinary glucose excretion. Together, these effects reduce glucose and insulin levels, enhance fatty acid β-oxidation, lower insulin resistance, and promote caloric deficit, all relevant to MASLD pathophysiology [24,135,136].
The phase 2a clinical trial NCT03205150 investigated licogliflozin, a dual SGLT1/SGLT2 inhibitor [92]. In this randomized, placebo-controlled study, patients received either 30 mg or 150 mg of licogliflozin daily for 12 weeks. The 150 mg group showed significant reductions in ALT (−30.4 U/L) and improvements in hepatic steatosis (−6.9%) as measured by MRI-PDFF, along with reductions in AST (−17.0 U/L) and body weight (−4.5 kg). The treatment was generally well tolerated, though gastrointestinal side effects, especially diarrhea, were more frequent at the higher dose.

23. Testosterone

The therapeutic rationale for testosterone supplementation in MASLD is supported by growing evidence that low testosterone levels in men are associated with increased visceral adiposity, insulin resistance, and hepatic steatosis. Testosterone enhances insulin sensitivity by improving insulin signaling pathways and decreasing adipose tissue-derived inflammation [137]. Additionally, testosterone promotes hepatic fatty acid oxidation while downregulating de novo lipogenesis, contributing to a reduction in intrahepatic triglyceride accumulation. Its anti-inflammatory effects further help suppress hepatic production of pro-inflammatory cytokines and reduce oxidative stress, both of which play a role in the transition from simple steatosis to steatohepatitis and fibrosis [138,139,140].
The phase 2 LiFT trial (NCT04134091) evaluated LPCN 1144, an oral prodrug of testosterone, in men with biopsy-confirmed NASH and fibrosis stages F1–F3 [93]. In this 36-week, randomized, placebo-controlled trial, participants received LPCN 1144 either alone or in combination with vitamin E. Both treatment arms showed significant reductions in liver fat content by week 12, with mean absolute reductions of up to −9.2%. By week 36, both groups achieved statistically significant NASH resolution without worsening fibrosis. Additionally, treatment led to reductions in fibrosis, ALT (−22.9 U/L), AST (−12.0 U/L), ALP (−8.5 U/L), and GGT (−13.4 U/L). LPCN 1144 was well tolerated, with adverse event rates similar to placebo.
These findings suggest that testosterone replacement through LPCN 1144 may serve as a potential therapeutic approach in MASLD, particularly in hypogonadal men, by targeting hepatic steatosis, liver injury, and fibrosis. Further studies are needed to confirm long-term outcomes and clarify its role in broader patient populations.

24. THR-β Agonists

The therapeutic effects of thyroid hormone receptor beta (THR-β) agonists like TERN-501 in MASH are attributed to their ability to enhance hepatic lipid metabolism through mitochondrial biogenesis and β-oxidation. THR-β is predominantly expressed in the liver, where its activation promotes fatty acid oxidation and reduces hepatic lipogenesis—key mechanisms relevant to MASLD [141]. Unlike non-selective thyroid hormone therapies, THR-β agonists aim to provide metabolic benefits without systemic hormonal side effects.
The DUET trial (NCT05415722) evaluated TERN-501, a selective THR-β agonist, in patients with MASH. In this 12-week, randomized, double-blind, placebo-controlled phase 2a study, TERN-501 was administered at varying doses as monotherapy or in combination with the FXR agonist TERN-101. The primary endpoint was a change in liver fat content as measured by MRI-PDFF. TERN-501 monotherapy significantly reduced liver fat content across all dose groups, reaching −44.8% in the 6 mg group. The treatment was well tolerated, with only a slightly higher incidence of pruritus.

25. Conclusions

The growing body of evidence from recent RCTs underscores the remarkable diversity of molecular pathways implicated in MASLD pathogenesis. A wide array of investigational drugs has emerged, targeting key molecular mechanisms involved in disease progression. Among these, GLP-1 receptor agonists, PPAR agonists, and FXR agonists, as well as inhibitors of ACC and DGAT, have been the most extensively studied and demonstrate the most promising therapeutic potential to date. Notably, GLP-1 receptor agonists, as well as dual GLP-1/GIP and GLP-1/glucagon receptor agonists, appear to exert the most comprehensive effects on MASLD, simultaneously improving steatosis, insulin resistance, inflammation, and body weight. While several agents have shown encouraging results in terms of hepatic fat reduction, metabolic improvement, and even histological benefits, the majority of available data come from small, early-phase trials, primarily phase 2 studies. The current landscape therefore reflects an exciting but still preliminary stage of drug development in MASLD. Robust, large-scale phase 3 trials are still needed to confirm these findings, evaluate long-term safety and efficacy, and ultimately establish evidence-based pharmacologic treatments for widespread clinical use.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

During the preparation of this manuscript, the author used Chat GPT, 4.0, for the purposes of grammar and language checking, as well as for the creation of graphics. The author also used Servier graphics. The author has reviewed and edited the output and takes full responsibility for the content of this publication.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ACCAcetyl-CoA carboxylase
ALPAlkaline phosphatase
ALTAlanine aminotransferase
AMPKAMP-activated protein kinase
ASAAcetylsalicylic acid
ASTAspartate transaminase
ATPAdenosine triphosphate
COXCyclooxygenase
CPT 1Carnitine palmitoyltransferase 1
DGATDiacylglycerol acyltransferase
DNP2,4-dinitrophenol
FASFatty acid synthase
FFARFree fatty acid receptor
FXRFarnesoid X receptor
FGFFibroblast growth factor
GIPGastric inhibitory polypeptide
GLP-1Glucagon-like peptide-1
HMG-CoAβ-Hydroxy β-methylglutaryl-CoA
IBATIleal bile acid transporter
KHXKetohexokinase
KLBCo-receptor β-Klotho
MASLDMetabolic dysfunction-associated steatotic liver disease
NAFLDNon-alcoholic fatty liver disease
NASHNon-alcoholic steatohepatitis
OCAObeticholic acid
PPARPeroxisome proliferator-activated receptor
RCTRandomized clinical trial
SCD1Stearoyl-CoA desaturase 1
SHPSmall heterodimer partner
SREBP-1cSterol regulatory element-binding protein 1
TGF-ß1Transforming growth factor-beta1
TGRTakeda G protein-coupled receptor
THR-ßThyroid hormone receptor ß

References

  1. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global Epidemiology of Nonalcoholic Fatty Liver Disease—Meta-analytic Assessment of Prevalence, Incidence, and Outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, G.Y.; Brandman, D. A Clinical Update on MASLD. JAMA Intern. Med. 2025, 185, 105. [Google Scholar] [CrossRef]
  3. Bessone, F.; Razori, M.V.; Roma, M.G. Molecular Pathways of Nonalcoholic Fatty Liver Disease Development and Progression. Cell. Mol. Life Sci. 2019, 76, 99–128. [Google Scholar] [CrossRef] [PubMed]
  4. Grander, C.; Grabherr, F.; Tilg, H. Non-Alcoholic Fatty Liver Disease: Pathophysiological Concepts and Treatment Options. Cardiovasc. Res. 2023, 119, 1787–1798. [Google Scholar] [CrossRef]
  5. Malhi, H.; Gores, G. Molecular Mechanisms of Lipotoxicity in Nonalcoholic Fatty Liver Disease. Semin. Liver Dis. 2008, 28, 360–369. [Google Scholar] [CrossRef] [PubMed]
  6. Kasumov, T.; Li, L.; Li, M.; Gulshan, K.; Kirwan, J.P.; Liu, X.; Previs, S.; Willard, B.; Smith, J.D.; McCullough, A. Ceramide as a Mediator of Non-Alcoholic Fatty Liver Disease and Associated Atherosclerosis. PLoS ONE 2015, 10, e0126910. [Google Scholar] [CrossRef]
  7. Mota, M.; Banini, B.A.; Cazanave, S.C.; Sanyal, A.J. Molecular Mechanisms of Lipotoxicity and Glucotoxicity in Nonalcoholic Fatty Liver Disease. Metabolism 2016, 65, 1049–1061. [Google Scholar] [CrossRef]
  8. Tilg, H.; Moschen, A.R. Evolution of Inflammation in Nonalcoholic Fatty Liver Disease: The Multiple Parallel Hits Hypothesis. Hepatology 2010, 52, 1836–1846. [Google Scholar] [CrossRef]
  9. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The Multiple-Hit Pathogenesis of Non-Alcoholic Fatty Liver Disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
  10. Thuy, S.; Ladurner, R.; Volynets, V.; Wagner, S.; Strahl, S.; Königsrainer, A.; Maier, K.-P.; Bischoff, S.C.; Bergheim, I. Nonalcoholic Fatty Liver Disease in Humans Is Associated with Increased Plasma Endotoxin and Plasminogen Activator Inhibitor 1 Concentrations and with Fructose Intake. J. Nutr. 2008, 138, 1452–1455. [Google Scholar] [CrossRef]
  11. Le Roy, T.; Llopis, M.; Lepage, P.; Bruneau, A.; Rabot, S.; Bevilacqua, C.; Martin, P.; Philippe, C.; Walker, F.; Bado, A.; et al. Intestinal Microbiota Determines Development of Non-Alcoholic Fatty Liver Disease in Mice. Gut 2013, 62, 1787–1794. [Google Scholar] [CrossRef] [PubMed]
  12. Arab, J.P.; Arrese, M.; Trauner, M. Recent Insights into the Pathogenesis of Nonalcoholic Fatty Liver Disease. Annu. Rev. Pathol. Mech. Dis. 2018, 13, 321–350. [Google Scholar] [CrossRef] [PubMed]
  13. Rinella, M.E.; Neuschwander-Tetri, B.A.; Siddiqui, M.S.; Abdelmalek, M.F.; Caldwell, S.; Barb, D.; Kleiner, D.E.; Loomba, R. AASLD Practice Guidance on the Clinical Assessment and Management of Nonalcoholic Fatty Liver Disease. Hepatology 2023, 77, 1797–1835. [Google Scholar] [CrossRef]
  14. Kwong, A.J.; Kim, W.R.; Lake, J.R.; Schladt, D.P.; Schnellinger, E.M.; Gauntt, K.; McDermott, M.; Weiss, S.; Handarova, D.K.; Snyder, J.J.; et al. OPTN/SRTR 2022 Annual Data Report: Liver. Am. J. Transplant. 2024, 24, S176–S265. [Google Scholar] [CrossRef]
  15. Kanwal, F.; Shubrook, J.H.; Adams, L.A.; Pfotenhauer, K.; Wai-Sun Wong, V.; Wright, E.; Abdelmalek, M.F.; Harrison, S.A.; Loomba, R.; Mantzoros, C.S.; et al. Clinical Care Pathway for the Risk Stratification and Management of Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology 2021, 161, 1657–1669. [Google Scholar] [CrossRef]
  16. Cusi, K.; Isaacs, S.; Barb, D.; Basu, R.; Caprio, S.; Garvey, W.T.; Kashyap, S.; Mechanick, J.I.; Mouzaki, M.; Nadolsky, K.; et al. American Association of Clinical Endocrinology Clinical Practice Guideline for the Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Primary Care and Endocrinology Clinical Settings. Endocr. Pract. 2022, 28, 528–562. [Google Scholar] [CrossRef] [PubMed]
  17. Keam, S.J. Resmetirom: First Approval. Drugs 2024, 84, 729–735. [Google Scholar] [CrossRef]
  18. Belfort-DeAguiar, R.; Lomonaco, R.; Cusi, K. Approach to the Patient With Nonalcoholic Fatty Liver Disease. J. Clin. Endocrinol. Metab. 2023, 108, 483–495. [Google Scholar] [CrossRef]
  19. Dansinger, M.L.; Gleason, J.A.; Griffith, J.L.; Selker, H.P.; Schaefer, E.J. Comparison of the Atkins, Ornish, Weight Watchers, and Zone Diets for Weight Loss and Heart Disease Risk Reduction. JAMA 2005, 293, 43. [Google Scholar] [CrossRef]
  20. Sumithran, P.; Prendergast, L.A.; Delbridge, E.; Purcell, K.; Shulkes, A.; Kriketos, A.; Proietto, J. Long-Term Persistence of Hormonal Adaptations to Weight Loss. N. Engl. J. Med. 2011, 365, 1597–1604. [Google Scholar] [CrossRef]
  21. Rajewski, P.; Cieściński, J.; Rajewski, P.; Suwała, S.; Rajewska, A.; Potasz, M. Dietary Interventions and Physical Activity as Crucial Factors in the Prevention and Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease. Biomedicines 2025, 13, 217. [Google Scholar] [CrossRef]
  22. Plauth, M.; Bernal, W.; Dasarathy, S.; Merli, M.; Plank, L.D.; Schütz, T.; Bischoff, S.C. ESPEN Guideline on Clinical Nutrition in Liver Disease. Clin. Nutr. 2019, 38, 485–521. [Google Scholar] [CrossRef]
  23. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The Diagnosis and Management of Nonalcoholic Fatty Liver Disease: Practice Guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [PubMed]
  24. 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 for the Management of Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2016, 64, 1388–1402. [Google Scholar] [CrossRef] [PubMed]
  25. Abenavoli, L.; Milanović, M.; Milić, N.; Luzza, F.; Giuffrè, A.M. Olive Oil Antioxidants and Non-Alcoholic Fatty Liver Disease. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 739–749. [Google Scholar] [CrossRef] [PubMed]
  26. Anania, C.; Perla, F.M.; Olivero, F.; Pacifico, L.; Chiesa, C. Mediterranean Diet and Nonalcoholic Fatty Liver Disease. World J. Gastroenterol. 2018, 24, 2083–2094. [Google Scholar] [CrossRef]
  27. Xu, C.; Markova, M.; Seebeck, N.; Loft, A.; Hornemann, S.; Gantert, T.; Kabisch, S.; Herz, K.; Loske, J.; Ost, M.; et al. High-protein Diet More Effectively Reduces Hepatic Fat than Low-protein Diet despite Lower Autophagy and FGF21 Levels. Liver Int. 2020, 40, 2982–2997. [Google Scholar] [CrossRef]
  28. Choi, Y.J.; Jeon, S.-M.; Shin, S. Impact of a Ketogenic Diet on Metabolic Parameters in Patients with Obesity or Overweight and with or without Type 2 Diabetes: A Meta-Analysis of Randomized Controlled Trials. Nutrients 2020, 12, 2005. [Google Scholar] [CrossRef]
  29. Belopolsky, Y.; Khan, M.Q.; Sonnenberg, A.; Davidson, D.J.; Fimmel, C.J. Ketogenic, Hypocaloric Diet Improves Nonalcoholic Steatohepatitis. J. Transl. Intern. Med. 2020, 8, 26–31. [Google Scholar] [CrossRef]
  30. Holmer, M.; Lindqvist, C.; Petersson, S.; Moshtaghi-Svensson, J.; Tillander, V.; Brismar, T.B.; Hagström, H.; Stål, P. Treatment of NAFLD with Intermittent Calorie Restriction or Low-Carb High-Fat Diet–A Randomised Controlled Trial. JHEP Rep. 2021, 3, 100256. [Google Scholar] [CrossRef]
  31. Johari, M.I.; Yusoff, K.; Haron, J.; Nadarajan, C.; Ibrahim, K.N.; Wong, M.S.; Hafidz, M.I.A.; Chua, B.E.; Hamid, N.; Arifin, W.N.; et al. A Randomised Controlled Trial on the Effectiveness and Adherence of Modified Alternate-Day Calorie Restriction in Improving Activity of Non-Alcoholic Fatty Liver Disease. Sci. Rep. 2019, 9, 11232. [Google Scholar] [CrossRef] [PubMed]
  32. Trepanowski, J.F.; Kroeger, C.M.; Barnosky, A.; Klempel, M.; Bhutani, S.; Hoddy, K.K.; Rood, J.; Ravussin, E.; Varady, K.A. Effects of Alternate-Day Fasting or Daily Calorie Restriction on Body Composition, Fat Distribution, and Circulating Adipokines: Secondary Analysis of a Randomized Controlled Trial. Clin. Nutr. 2018, 37, 1871–1878. [Google Scholar] [CrossRef] [PubMed]
  33. Lavallee, C.M.; Bruno, A.; Ma, C.; Raman, M. The Role of Intermittent Fasting in the Management of Nonalcoholic Fatty Liver Disease: A Narrative Review. Nutrients 2022, 14, 4655. [Google Scholar] [CrossRef] [PubMed]
  34. Machado, M.V. Aerobic Exercise in the Management of Metabolic Dysfunction Associated Fatty Liver Disease. Diabetes Metab. Syndr. Obes. 2021, 14, 3627–3645. [Google Scholar] [CrossRef]
  35. Bae, J.C.; Suh, S.; Park, S.E.; Rhee, E.J.; Park, C.Y.; Oh, K.W.; Park, S.W.; Kim, S.W.; Hur, K.Y.; Kim, J.H.; et al. Regular Exercise Is Associated with a Reduction in the Risk of NAFLD and Decreased Liver Enzymes in Individuals with NAFLD Independent of Obesity in Korean Adults. PLoS ONE 2012, 7, e46819. [Google Scholar] [CrossRef]
  36. Ryu, S.; Chang, Y.; Jung, H.-S.; Yun, K.E.; Kwon, M.-J.; Choi, Y.; Kim, C.-W.; Cho, J.; Suh, B.-S.; Cho, Y.K.; et al. Relationship of Sitting Time and Physical Activity with Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2015, 63, 1229–1237. [Google Scholar] [CrossRef]
  37. Byambasukh, O.; Zelle, D.; Corpeleijn, E. Physical Activity, Fatty Liver, and Glucose Metabolism Over the Life Course: The Lifelines Cohort. Am. J. Gastroenterol. 2019, 114, 907–915. [Google Scholar] [CrossRef]
  38. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef]
  39. Cusi, K.; Orsak, B.; Bril, F.; Lomonaco, R.; Hecht, J.; Ortiz-Lopez, C.; Tio, F.; Hardies, J.; Darland, C.; Musi, N.; et al. Long-Term Pioglitazone Treatment for Patients With Nonalcoholic Steatohepatitis and Prediabetes or Type 2 Diabetes Mellitus. Ann. Intern. Med. 2016, 165, 305–315. [Google Scholar] [CrossRef]
  40. Musso, G.; Cassader, M.; Paschetta, E.; Gambino, R. Thiazolidinediones and Advanced Liver Fibrosis in Nonalcoholic Steatohepatitis. JAMA Intern. Med. 2017, 177, 633. [Google Scholar] [CrossRef]
  41. Wen, H.; Deng, H.; Yang, L.; Li, L.; Lin, J.; Zheng, P.; Bjelakovic, M.; Ji, G. Vitamin E for People with Non-Alcoholic Fatty Liver Disease. Cochrane Database Syst. Rev. 2024, 2024, CD015033. [Google Scholar] [CrossRef]
  42. Chee, N.M.; Sinnanaidu, R.P.; Chan, W. Vitamin E Improves Serum Markers and Histology in Adults with Metabolic Dysfunction-associated Steatotic Liver Disease: Systematic Review and Meta-analysis. J. Gastroenterol. Hepatol. 2024, 39, 2545–2554. [Google Scholar] [CrossRef] [PubMed]
  43. Schölmerich, J.; Becher, M.-S.; Schmidt, K.; Schubert, R.; Kremer, B.; Feldhaus, S.; Gerok, W. Influence of Hydroxylation and Conjugation of Bile Salts on Their Membrane-Damaging Properties-Studies on Isolated Hepatocytes and Lipid Membrane Vesicles. Hepatology 1984, 4, 661–666. [Google Scholar] [CrossRef]
  44. Lindor, K.D.; Kowdley, K.V.; Luketic, V.A.C.; Harrison, E.M.; McCashland, T.; Befeler, A.S.; Harnois, D.; Jorgensen, R.; Petz, J.; Keach, J.; et al. High-Dose Ursodeoxycholic Acid for the Treatment of Primary Sclerosing Cholangitis. Hepatology 2009, 50, 808–814. [Google Scholar] [CrossRef] [PubMed]
  45. Xiang, Z.; Chen, Y.; Ma, K.; Ye, Y.; Zheng, L.; Yang, Y.; Li, Y.; Jin, X. The Role of Ursodeoxycholic Acid in Non-Alcoholic Steatohepatitis: A Systematic Review. BMC Gastroenterol. 2013, 13, 140. [Google Scholar] [CrossRef]
  46. Lindor, K.D.; Kowdley, K.V.; Heathcote, J.E.; Harrison, E.M.; Jorgensen, R.; Angulo, P.; Lymp, J.F.; Burgart, L.; Colin, P. Ursodeoxycholic Acid for Treatment of Nonalcoholic Steatohepatitis: Results of a Randomized Trial. Hepatology 2004, 39, 770–778. [Google Scholar] [CrossRef]
  47. Laurin, J.; Lindor, K.D.; Crippin, J.S.; Gossard, A.; Gores, G.J.; Ludwig, J.; Rakela, J.; McGill, D.B. Ursodeoxycholic Acid or Clofibrate in the Treatment of Non–Alcohol–Induced Steatohepatitis: A Pilot Study. Hepatology 1996, 23, 1464–1467. [Google Scholar] [CrossRef]
  48. Zhang, W.; Tang, Y.; Huang, J.; Hu, H. Efficacy of Ursodeoxycholic Acid in Nonalcoholic Fatty Liver Disease: An Updated Meta-Analysis of Randomized Controlled Trials. Asia Pac. J. Clin. Nutr. 2020, 29, 696–705. [Google Scholar]
  49. Kim, C.-W.; Addy, C.; Kusunoki, J.; Anderson, N.N.; Deja, S.; Fu, X.; Burgess, S.C.; Li, C.; Ruddy, M.; Chakravarthy, M.; et al. Acetyl CoA Carboxylase Inhibition Reduces Hepatic Steatosis but Elevates Plasma Triglycerides in Mice and Humans: A Bedside to Bench Investigation. Cell Metab. 2017, 26, 394–406.e6. [Google Scholar] [CrossRef]
  50. Calle, R.A.; Amin, N.B.; Carvajal-Gonzalez, S.; Ross, T.T.; Bergman, A.; Aggarwal, S.; Crowley, C.; Rinaldi, A.; Mancuso, J.; Aggarwal, N.; et al. ACC Inhibitor Alone or Co-Administered with a DGAT2 Inhibitor in Patients with Non-Alcoholic Fatty Liver Disease: Two Parallel, Placebo-Controlled, Randomized Phase 2a Trials. Nat. Med. 2021, 27, 1836–1848. [Google Scholar] [CrossRef]
  51. Sivakumar, P.; Saul, M.; Robinson, D.; King, L.E.; Amin, N.B. SomaLogic Proteomics Reveals New Biomarkers and Provides Mechanistic, Clinical Insights into Acetyl CoA Carboxylase (ACC) Inhibition in Non-Alcoholic Steatohepatitis (NASH). Sci. Rep. 2024, 14, 17072. [Google Scholar] [CrossRef] [PubMed]
  52. Amin, N.B.; Darekar, A.; Anstee, Q.M.; Wong, V.W.-S.; Tacke, F.; Vourvahis, M.; Lee, D.S.; Charlton, M.; Alkhouri, N.; Nakajima, A.; et al. Efficacy and Safety of an Orally Administered DGAT2 Inhibitor Alone or Coadministered with a Liver-Targeted ACC Inhibitor in Adults with Non-Alcoholic Steatohepatitis (NASH): Rationale and Design of the Phase II, Dose-Ranging, Dose-Finding, Randomised, Placebo-Controlled MIRNA (Metabolic Interventions to Resolve NASH with Fibrosis) Study. BMJ Open 2022, 12, e056159. [Google Scholar] [CrossRef]
  53. Harrison, S.A.; Baum, S.J.; Gunn, N.T.; Younes, Z.H.; Kohli, A.; Patil, R.; Koziel, M.J.; Chera, H.; Zhao, J.; Chakravarthy, M.V. Safety, Tolerability, and Biologic Activity of AXA1125 and AXA1957 in Subjects With Nonalcoholic Fatty Liver Disease. Am. J. Gastroenterol. 2021, 116, 2399–2409. [Google Scholar] [CrossRef] [PubMed]
  54. Cusi, K.; Alkhouri, N.; Harrison, S.A.; Fouqueray, P.; Moller, D.E.; Hallakou-Bozec, S.; Bolze, S.; Grouin, J.-M.; Jeannin Megnien, S.; Dubourg, J.; et al. Efficacy and Safety of PXL770, a Direct AMP Kinase Activator, for the Treatment of Non-Alcoholic Fatty Liver Disease (STAMP-NAFLD): A Randomised, Double-Blind, Placebo-Controlled, Phase 2a Study. Lancet Gastroenterol. Hepatol. 2021, 6, 889–902. [Google Scholar] [CrossRef] [PubMed]
  55. Loomba, R.; Lutchman, G.; Kleiner, D.E.; Ricks, M.; Feld, J.J.; Borg, B.B.; Modi, A.; Nagabhyru, P.; Sumner, A.E.; Liang, T.J.; et al. Clinical Trial: Pilot Study of Metformin for the Treatment of Non-alcoholic Steatohepatitis. Aliment. Pharmacol. Ther. 2009, 29, 172–182. [Google Scholar] [CrossRef]
  56. Ratziu, V.; de Guevara, L.; Safadi, R.; Poordad, F.; Fuster, F.; Flores-Figueroa, J.; Arrese, M.; Fracanzani, A.L.; Ben Bashat, D.; Lackner, K.; et al. Aramchol in Patients with Nonalcoholic Steatohepatitis: A Randomized, Double-Blind, Placebo-Controlled Phase 2b Trial. Nat. Med. 2021, 27, 1825–1835. [Google Scholar] [CrossRef]
  57. Loomba, R.; Mohseni, R.; Lucas, K.J.; Gutierrez, J.A.; Perry, R.G.; Trotter, J.F.; Rahimi, R.S.; Harrison, S.A.; Ajmera, V.; Wayne, J.D.; et al. TVB-2640 (FASN Inhibitor) for the Treatment of Nonalcoholic Steatohepatitis: FASCINATE-1, a Randomized, Placebo-Controlled Phase 2a Trial. Gastroenterology 2021, 161, 1475–1486. [Google Scholar] [CrossRef]
  58. Harrison, S.A.; Alkhouri, N.; Ortiz-Lasanta, G.; Rudraraju, M.; Tai, D.; Wack, K.; Shah, A.; Besuyen, R.; Steineger, H.H.; Fraser, D.A.; et al. A Phase IIb Randomised-Controlled Trial of the FFAR1/FFAR4 Agonist Icosabutate in MASH. J. Hepatol. 2025, in press. [Google Scholar] [CrossRef]
  59. Harrison, S.A.; Ruane, P.J.; Freilich, B.L.; Neff, G.; Patil, R.; Behling, C.A.; Hu, C.; Fong, E.; de Temple, B.; Tillman, E.J.; et al. Efruxifermin in Non-Alcoholic Steatohepatitis: A Randomized, Double-Blind, Placebo-Controlled, Phase 2a Trial. Nat. Med. 2021, 27, 1262–1271. [Google Scholar] [CrossRef]
  60. Harrison, S.A.; Ruane, P.J.; Freilich, B.; Neff, G.; Patil, R.; Behling, C.; Hu, C.; Shringarpure, R.; de Temple, B.; Fong, E.; et al. A Randomized, Double-Blind, Placebo-Controlled Phase IIa Trial of Efruxifermin for Patients with Compensated NASH Cirrhosis. JHEP Rep. 2023, 5, 100563. [Google Scholar] [CrossRef]
  61. Alkhouri, N.; Beyer, C.; Shumbayawonda, E.; Andersson, A.; Yale, K.; Rolph, T.; Chung, R.T.; Vuppalanchi, R.; Cusi, K.; Loomba, R.; et al. Decreases in CT1 and Liver Fat Content Reflect Treatment-Induced Histological Improvements in MASH. J. Hepatol. 2025, 82, 438–445. [Google Scholar] [CrossRef] [PubMed]
  62. Luo, Y.; Decato, B.E.; Charles, E.D.; Shevell, D.E.; McNaney, C.; Shipkova, P.; Apfel, A.; Tirucherai, G.S.; Sanyal, A.J. Pegbelfermin Selectively Reduces Secondary Bile Acid Concentrations in Patients with Non-Alcoholic Steatohepatitis. JHEP Rep. 2022, 4, 100392. [Google Scholar] [CrossRef] [PubMed]
  63. Sanyal, A.; Charles, E.D.; Neuschwander-Tetri, B.A.; Loomba, R.; Harrison, S.A.; Abdelmalek, M.F.; Lawitz, E.J.; Halegoua-DeMarzio, D.; Kundu, S.; Noviello, S.; et al. Pegbelfermin (BMS-986036), a PEGylated Fibroblast Growth Factor 21 Analogue, in Patients with Non-Alcoholic Steatohepatitis: A Randomised, Double-Blind, Placebo-Controlled, Phase 2a Trial. Lancet 2018, 392, 2705–2717. [Google Scholar] [CrossRef] [PubMed]
  64. Raji, A.; Gantz, I.; Crutchlow, M.; Flynn, H.; Xu, L.; Rodgers, A.J.; Krishnan, R.; Rizk, M.L.; Hu, S.; Kaufman, K.D.; et al. Clinical Trial: A Phase 2b Study to Evaluate the Efficacy and Safety of MK-3655 in Individuals With Pre-Cirrhotic MASH. Aliment. Pharmacol. Ther. 2025, 61, 1152–1162. [Google Scholar] [CrossRef]
  65. Hameed, B.; Terrault, N.A.; Gill, R.M.; Loomba, R.; Chalasani, N.; Hoofnagle, J.H.; Van Natta, M.L. Clinical and Metabolic Effects Associated with Weight Changes and Obeticholic Acid in Non-alcoholic Steatohepatitis. Aliment. Pharmacol. Ther. 2018, 47, 645–656. [Google Scholar] [CrossRef]
  66. Siddiqui, M.S.; Van Natta, M.L.; Connelly, M.A.; Vuppalanchi, R.; Neuschwander-Tetri, B.A.; Tonascia, J.; Guy, C.; Loomba, R.; Dasarathy, S.; Wattacheril, J.; et al. Impact of Obeticholic Acid on the Lipoprotein Profile in Patients with Non-Alcoholic Steatohepatitis. J. Hepatol. 2020, 72, 25–33. [Google Scholar] [CrossRef]
  67. Shen, W.; Middleton, M.S.; Cunha, G.M.; Delgado, T.I.; Wolfson, T.; Gamst, A.; Fowler, K.J.; Alazraki, A.; Trout, A.T.; Ohliger, M.A.; et al. Changes in Abdominal Adipose Tissue Depots Assessed by MRI Correlate with Hepatic Histologic Improvement in Non-Alcoholic Steatohepatitis. J. Hepatol. 2023, 78, 238–246. [Google Scholar] [CrossRef]
  68. Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Van Natta, M.L.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. Farnesoid X Nuclear Receptor Ligand Obeticholic Acid for Non-Cirrhotic, Non-Alcoholic Steatohepatitis (FLINT): A Multicentre, Randomised, Placebo-Controlled Trial. Lancet 2015, 385, 956–965. [Google Scholar] [CrossRef]
  69. Mudaliar, S.; Henry, R.R.; Sanyal, A.J.; Morrow, L.; Marschall, H.; Kipnes, M.; Adorini, L.; Sciacca, C.I.; Clopton, P.; Castelloe, E.; et al. Efficacy and Safety of the Farnesoid X Receptor Agonist Obeticholic Acid in Patients With Type 2 Diabetes and Nonalcoholic Fatty Liver Disease. Gastroenterology 2013, 145, 574–582.e1. [Google Scholar] [CrossRef]
  70. Ratziu, V.; Harrison, S.A.; Loustaud-Ratti, V.; Bureau, C.; Lawitz, E.; Abdelmalek, M.; Alkhouri, N.; Francque, S.; Girma, H.; Darteil, R.; et al. Hepatic and Renal Improvements with FXR Agonist Vonafexor in Individuals with Suspected Fibrotic NASH. J. Hepatol. 2023, 78, 479–492. [Google Scholar] [CrossRef]
  71. Ratziu, V.; Rinella, M.E.; Neuschwander-Tetri, B.A.; Lawitz, E.; Denham, D.; Kayali, Z.; Sheikh, A.; Kowdley, K.V.; Desta, T.; Elkhashab, M.; et al. EDP-305 in Patients with NASH: A Phase II Double-Blind Placebo-Controlled Dose-Ranging Study. J. Hepatol. 2022, 76, 506–517. [Google Scholar] [CrossRef]
  72. Loomba, R.; Abdelmalek, M.F.; Armstrong, M.J.; Jara, M.; Kjær, M.S.; Krarup, N.; Lawitz, E.; Ratziu, V.; Sanyal, A.J.; Schattenberg, J.M.; et al. Semaglutide 2·4 Mg Once Weekly in Patients with Non-Alcoholic Steatohepatitis-Related Cirrhosis: A Randomised, Placebo-Controlled Phase 2 Trial. Lancet Gastroenterol. Hepatol. 2023, 8, 511–522. [Google Scholar] [CrossRef]
  73. Armstrong, M.J.; Okanoue, T.; Sundby Palle, M.; Sejling, A.; Tawfik, M.; Roden, M. Similar Weight Loss with Semaglutide Regardless of Diabetes and Cardiometabolic Risk Parameters in Individuals with Metabolic Dysfunction-associated Steatotic Liver Disease: Post Hoc Analysis of Three Randomised Controlled Trials. Diabetes Obes. Metab. 2025, 27, 710–718. [Google Scholar] [CrossRef] [PubMed]
  74. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.-S.; Harrison, S.A. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124. [Google Scholar] [CrossRef]
  75. Ditzenberger, G.L.; Lake, J.E.; Kitch, D.W.; Kantor, A.; Muthupillai, R.; Moser, C.; Belaunzaran-Zamudio, P.F.; Brown, T.T.; Corey, K.; Landay, A.L.; et al. Effects of Semaglutide on Muscle Structure and Function in the SLIM LIVER Study. Clin. Infect. Dis. 2025, 80, 389–396. [Google Scholar] [CrossRef] [PubMed]
  76. Romero-Gómez, M.; Lawitz, E.; Shankar, R.R.; Chaudhri, E.; Liu, J.; Lam, R.L.H.; Kaufman, K.D.; Engel, S.S.; Bruzone, S.O.; Coronel, M.J.; et al. A Phase IIa Active-Comparator-Controlled Study to Evaluate the Efficacy and Safety of Efinopegdutide in Patients with Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2023, 79, 888–897. [Google Scholar] [CrossRef] [PubMed]
  77. Loomba, R.; Hartman, M.L.; Lawitz, E.J.; Vuppalanchi, R.; Boursier, J.; Bugianesi, E.; Yoneda, M.; Behling, C.; Cummings, O.W.; Tang, Y.; et al. Tirzepatide for Metabolic Dysfunction–Associated Steatohepatitis with Liver Fibrosis. N. Engl. J. Med. 2024, 391, 299–310. [Google Scholar] [CrossRef]
  78. Sanyal, A.J.; Bedossa, P.; Fraessdorf, M.; Neff, G.W.; Lawitz, E.; Bugianesi, E.; Anstee, Q.M.; Hussain, S.A.; Newsome, P.N.; Ratziu, V.; et al. A Phase 2 Randomized Trial of Survodutide in MASH and Fibrosis. N. Engl. J. Med. 2024, 391, 311–319. [Google Scholar] [CrossRef]
  79. Shankar, S.S.; Daniels, S.J.; Robertson, D.; Sarv, J.; Sánchez, J.; Carter, D.; Jermutus, L.; Challis, B.; Sanyal, A.J. Safety and Efficacy of Novel Incretin Co-Agonist Cotadutide in Biopsy-Proven Noncirrhotic MASH With Fibrosis. Clin. Gastroenterol. Hepatol. 2024, 22, 1847–1857.e11. [Google Scholar] [CrossRef]
  80. Kazierad, D.J.; Chidsey, K.; Somayaji, V.R.; Bergman, A.J.; Birnbaum, M.J.; Calle, R.A. Inhibition of Ketohexokinase in Adults with NAFLD Reduces Liver Fat and Inflammatory Markers: A Randomized Phase 2 Trial. Med 2021, 2, 800–813.e3. [Google Scholar] [CrossRef]
  81. Saxena, A.R.; Lyle, S.; Khavandi, K.; Qiu, R.; Whitlock, M.; Esler, W.P.; Kim, A.M. A Phase 2a, Randomized, Double-blind, Placebo-controlled, Three-arm, Parallel-group Study to Assess the Efficacy, Safety, Tolerability and Pharmacodynamics of PF-06835919 in Patients with Non-alcoholic Fatty Liver Disease and Type 2 Diabetes. Diabetes Obes. Metab. 2023, 25, 992–1001. [Google Scholar] [CrossRef] [PubMed]
  82. Noureddin, M.; Khan, S.; Portell, F.; Jorkasky, D.; Dennis, J.; Khan, O.; Johansson, L.; Johansson, E.; Sanyal, A.J. Safety and Efficacy of Once-Daily HU6 versus Placebo in People with Non-Alcoholic Fatty Liver Disease and High BMI: A Randomised, Double-Blind, Placebo-Controlled, Phase 2a Trial. Lancet Gastroenterol. Hepatol. 2023, 8, 1094–1105. [Google Scholar] [CrossRef] [PubMed]
  83. Simon, T.G.; Wilechansky, R.M.; Stoyanova, S.; Grossman, A.; Dichtel, L.E.; Lauer, G.M.; Miller, K.K.; Hoshida, Y.; Corey, K.E.; Loomba, R.; et al. Aspirin for Metabolic Dysfunction–Associated Steatotic Liver Disease Without Cirrhosis. JAMA 2024, 331, 920. [Google Scholar] [CrossRef]
  84. Harrison, S.A.; Thang, C.; Bolze, S.; Dewitt, S.; Hallakou-Bozec, S.; Dubourg, J.; Bedossa, P.; Cusi, K.; Ratziu, V.; Grouin, J.-M.; 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. [Google Scholar] [CrossRef]
  85. Monternier, P.; Singh, J.; Parasar, P.; Theurey, P.; DeWitt, S.; Jacques, V.; Klett, E.; Kaur, N.; Nagaraja, T.N.; Moller, D.E.; et al. Therapeutic Potential of Deuterium-stabilized (R)-pioglitazone—PXL065—For X-linked Adrenoleukodystrophy. J. Inherit. Metab. Dis. 2022, 45, 832–847. [Google Scholar] [CrossRef] [PubMed]
  86. Gawrieh, S.; Noureddin, M.; Loo, N.; Mohseni, R.; Awasty, V.; Cusi, K.; Kowdley, K.V.; Lai, M.; Schiff, E.; Parmar, D.; et al. Saroglitazar, a PPAR-α/γ Agonist, for Treatment of NAFLD: A Randomized Controlled Double-Blind Phase 2 Trial. Hepatology 2021, 74, 1809–1824. [Google Scholar] [CrossRef]
  87. Siddiqui, M.S.; Parmar, D.; Sheikh, F.; Sarin, S.K.; Cisneros, L.; Gawrieh, S.; Momin, T.; Duseja, A.; Sanyal, A.J. Saroglitazar, a Dual PPAR α/γ Agonist, Improves Atherogenic Dyslipidemia in Patients With Non-Cirrhotic Nonalcoholic Fatty Liver Disease: A Pooled Analysis. Clin. Gastroenterol. Hepatol. 2023, 21, 2597–2605.e2. [Google Scholar] [CrossRef]
  88. Francque, S.M.; Bedossa, P.; Ratziu, V.; Anstee, Q.M.; Bugianesi, E.; Sanyal, A.J.; Loomba, R.; Harrison, S.A.; Balabanska, R.; Mateva, L.; et al. A Randomized, Controlled Trial of the Pan-PPAR Agonist Lanifibranor in NASH. N. Engl. J. Med. 2021, 385, 1547–1558. [Google Scholar] [CrossRef]
  89. Cooreman, M.P.; Butler, J.; Giugliano, R.P.; Zannad, F.; Dzen, L.; Huot-Marchand, P.; Baudin, M.; Beard, D.R.; Junien, J.-L.; Broqua, P.; et al. The Pan-PPAR Agonist Lanifibranor Improves Cardiometabolic Health in Patients with Metabolic Dysfunction-Associated Steatohepatitis. Nat. Commun. 2024, 15, 3962. [Google Scholar] [CrossRef]
  90. Pierre, B.; Elisabetta, B.; Vlad, R.; Philippe, H.M.; Bruno, S.; Jean-Louis, J.; Pierre, B.; Jean-Louis, A. A Randomised, Double-Blind, Placebo-Controlled, Multi-Centre, Dose-Range, Proof-of-Concept, 24-Week Treatment Study of Lanifibranor in Adult Subjects with Non-Alcoholic Steatohepatitis: Design of the NATIVE Study. Contemp. Clin. Trials 2020, 98, 106170. [Google Scholar] [CrossRef]
  91. Barb, D.; Kalavalapalli, S.; Godinez Leiva, E.; Bril, F.; Huot-Marchand, P.; Dzen, L.; Rosenberg, J.T.; Junien, J.-L.; Broqua, P.; Rocha, A.O.; et al. Pan-PPAR Agonist Lanifibranor Improves Insulin Resistance and Hepatic Steatosis in Patients with T2D and MASLD. J. Hepatol. 2025, 82, 979–991. [Google Scholar] [CrossRef] [PubMed]
  92. Harrison, S.A.; Manghi, F.P.; Smith, W.B.; Alpenidze, D.; Aizenberg, D.; Klarenbeek, N.; Chen, C.-Y.; Zuckerman, E.; Ravussin, E.; Charatcharoenwitthaya, P.; et al. Licogliflozin for Nonalcoholic Steatohepatitis: A Randomized, Double-Blind, Placebo-Controlled, Phase 2a Study. Nat. Med. 2022, 28, 1432–1438. [Google Scholar] [CrossRef] [PubMed]
  93. Albhaisi, S.; Kim, K.; Baker, J.; Chidambaram, N.; Patel, M.V.; Charlton, M.; Sanyal, A.J. LPCN 1144 Resolves NAFLD in Hypogonadal Males. Hepatol. Commun. 2020, 4, 1430–1440. [Google Scholar] [CrossRef]
  94. Mateo-Marín, M.A.; Alves-Bezerra, M. Targeting Acetyl-CoA Carboxylases for the Treatment of MASLD. J. Lipid Res. 2024, 65, 100676. [Google Scholar] [CrossRef] [PubMed]
  95. Zalewska, A.; Szarmach, I.; Żendzian-Piotrowska, M.; Maciejczyk, M. The Effect of N-Acetylcysteine on Respiratory Enzymes, ADP/ATP Ratio, Glutathione Metabolism, and Nitrosative Stress in the Salivary Gland Mitochondria of Insulin Resistant Rats. Nutrients 2020, 12, 458. [Google Scholar] [CrossRef]
  96. Zalewska, A.; Zięba, S.; Kostecka-Sochoń, P.; Kossakowska, A.; Żendzian-Piotrowska, M.; Matczuk, J.; Maciejczyk, M. NAC Supplementation of Hyperglycemic Rats Prevents the Development of Insulin Resistance and Improves Antioxidant Status but Only Alleviates General and Salivary Gland Oxidative Stress. Oxidative Med. Cell. Longev. 2020, 2020, 8831855. [Google Scholar] [CrossRef]
  97. Liu, A.; Cai, Y.; Yuan, Y.; Liu, M.; Zhang, Z.; Xu, Y.; Jiao, P. Efficacy and Safety of Carnitine Supplementation on NAFLD: A Systematic Review and Meta-Analysis. Syst. Rev. 2023, 12, 74. [Google Scholar] [CrossRef]
  98. Savic, D.; Hodson, L.; Neubauer, S.; Pavlides, M. The Importance of the Fatty Acid Transporter L-Carnitine in Non-Alcoholic Fatty Liver Disease (NAFLD). Nutrients 2020, 12, 2178. [Google Scholar] [CrossRef]
  99. Op den Kamp-Bruls, Y.M.H.; Op den Kamp, Y.J.M.; Veeraiah, P.; Zapata Perez, R.; Phielix, E.; Havekes, B.; Schaart, G.; Kornips, E.; Berendsen, B.R.B.; Virmani, A.; et al. Carnitine Supplementation Improves Insulin Sensitivity and Skeletal Muscle Acetylcarnitine Formation in Patients with Type 2 Diabetes. Diabetes Obes. Metab. 2025, 27, 2864–2877. [Google Scholar] [CrossRef]
  100. Yin, H.; Yuan, F.; Jiao, F.; Niu, Y.; Jiang, X.; Deng, J.; Guo, Y.; Chen, S.; Zhai, Q.; Hu, C.; et al. Intermittent Leucine Deprivation Produces Long-Lasting Improvement in Insulin Sensitivity by Increasing Hepatic Gcn2 Expression. Diabetes 2022, 71, 206–218. [Google Scholar] [CrossRef]
  101. De Bandt, J.-P.; Coumoul, X.; Barouki, R. Branched-Chain Amino Acids and Insulin Resistance, from Protein Supply to Diet-Induced Obesity. Nutrients 2022, 15, 68. [Google Scholar] [CrossRef]
  102. Viollet, B. Targeting the AMPK Pathway for the Treatment of Type 2 Diabetes. Front. Biosci. 2009, 14, 3380. [Google Scholar] [CrossRef] [PubMed]
  103. Viollet, B.; Horman, S.; Leclerc, J.; Lantier, L.; Foretz, M.; Billaud, M.; Giri, S.; Andreelli, F. AMPK Inhibition in Health and Disease. Crit. Rev. Biochem. Mol. Biol. 2010, 45, 276–295. [Google Scholar] [CrossRef] [PubMed]
  104. Simbrunner, B.; Trauner, M.; Reiberger, T. Review Article: Therapeutic Aspects of Bile Acid Signalling in the Gut-liver Axis. Aliment. Pharmacol. Ther. 2021, 54, 1243–1262. [Google Scholar] [CrossRef] [PubMed]
  105. Iruarrizaga-Lejarreta, M.; Varela-Rey, M.; Fernández-Ramos, D.; Martínez-Arranz, I.; Delgado, T.C.; Simon, J.; Gutiérrez-de Juan, V.; delaCruz-Villar, L.; Azkargorta, M.; Lavin, J.L.; et al. Role of Aramchol in Steatohepatitis and Fibrosis in Mice. Hepatol. Commun. 2017, 1, 911–927. [Google Scholar] [CrossRef] [PubMed]
  106. Bhattacharya, D.; Basta, B.; Mato, J.M.; Craig, A.; Fernández-Ramos, D.; Lopitz-Otsoa, F.; Tsvirkun, D.; Hayardeny, L.; Chandar, V.; Schwartz, R.E.; et al. Aramchol Downregulates Stearoyl CoA-Desaturase 1 in Hepatic Stellate Cells to Attenuate Cellular Fibrogenesis. JHEP Rep. 2021, 3, 100237. [Google Scholar] [CrossRef]
  107. Fernández-Ramos, D.; Lopitz-Otsoa, F.; Delacruz-Villar, L.; Bilbao, J.; Pagano, M.; Mosca, L.; Bizkarguenaga, M.; Serrano-Macia, M.; Azkargorta, M.; Iruarrizaga-Lejarreta, M.; et al. Arachidyl Amido Cholanoic Acid Improves Liver Glucose and Lipid Homeostasis in Nonalcoholic Steatohepatitis via AMPK and MTOR Regulation. World J. Gastroenterol. 2020, 26, 5101–5117. [Google Scholar] [CrossRef]
  108. Wang, W.; Gao, X.; Niu, W.; Yin, J.; He, K. Targeting Metabolism: Innovative Therapies for MASLD Unveiled. Int. J. Mol. Sci. 2025, 26, 4077. [Google Scholar] [CrossRef] [PubMed]
  109. Aseem, S.O.; Way, G.; Wang, J.; Zhao, D.; Tai, Y.; Gurley, E.; Zeng, J.; Wang, X.; Hylemon, P.B.; Huebert, R.C.; et al. Aramchol Attenuates Fibrosis in Mouse Models of Biliary Fibrosis and Blocks the TGFβ-Induced Fibroinflammatory Mediators in Cholangiocytes. bioRxiv 2024, bioRxiv:2024.11.06.621880. [Google Scholar]
  110. Meyers, C.D.; Serrano-Wu, M.; Amer, A.; Chen, J.; Erik, R.; Commerford, R.; Hubbard, B.; Brousseau, M.; Li, L.; Meihui, P.; et al. The DGAT1 Inhibitor Pradigastat Decreases Chylomicron Secretion and Prevents Postprandial Triglyceride Elevation in Humans. J. Clin. Lipidol. 2013, 7, 285. [Google Scholar] [CrossRef]
  111. Meyers, C.D.; Tremblay, K.; Amer, A.; Chen, J.; Jiang, L.; Gaudet, D. Effect of the DGAT1 Inhibitor Pradigastat on Triglyceride and ApoB48 Levels in Patients with Familial Chylomicronemia Syndrome. Lipids Health Dis. 2015, 14, 8. [Google Scholar] [CrossRef] [PubMed]
  112. Buhman, K.K.; Smith, S.J.; Stone, S.J.; Repa, J.J.; Wong, J.S.; Knapp, F.F.; Burri, B.J.; Hamilton, R.L.; Abumrad, N.A.; Farese, R.V. DGAT1 Is Not Essential for Intestinal Triacylglycerol Absorption or Chylomicron Synthesis. J. Biol. Chem. 2002, 277, 25474–25479. [Google Scholar] [CrossRef]
  113. O’Farrell, M.; Duke, G.; Crowley, R.; Buckley, D.; Martins, E.B.; Bhattacharya, D.; Friedman, S.L.; Kemble, G. FASN Inhibition Targets Multiple Drivers of NASH by Reducing Steatosis, Inflammation and Fibrosis in Preclinical Models. Sci. Rep. 2022, 12, 15661. [Google Scholar] [CrossRef] [PubMed]
  114. Matsukawa, T.; Yagi, T.; Uchida, T.; Sakai, M.; Mitsushima, M.; Naganuma, T.; Yano, H.; Inaba, Y.; Inoue, H.; Yanagida, K.; et al. Hepatic FASN Deficiency Differentially Affects Nonalcoholic Fatty Liver Disease and Diabetes in Mouse Obesity Models. JCI Insight 2023, 8, e161282. [Google Scholar] [CrossRef]
  115. Kiepura, A.; Stachyra, K.; Olszanecki, R. Anti-Atherosclerotic Potential of Free Fatty Acid Receptor 4 (FFAR4). Biomedicines 2021, 9, 467. [Google Scholar] [CrossRef]
  116. Szukiewicz, D. Potential Therapeutic Exploitation of G Protein-Coupled Receptor 120 (GPR120/FFAR4) Signaling in Obesity-Related Metabolic Disorders. Int. J. Mol. Sci. 2025, 26, 2501. [Google Scholar] [CrossRef] [PubMed]
  117. Kim, K.H.; Jeong, Y.T.; Oh, H.; Kim, S.H.; Cho, J.M.; Kim, Y.-N.; Kim, S.S.; Kim, D.H.; Hur, K.Y.; Kim, H.K.; et al. Autophagy Deficiency Leads to Protection from Obesity and Insulin Resistance by Inducing Fgf21 as a Mitokine. Nat. Med. 2013, 19, 83–92. [Google Scholar] [CrossRef]
  118. Kim, M.; Doridot, L.; Cunniff, J.C.; Parker, T.S.; Levine, D.M.; Hellerstein, M.K.; Hudgins, L.C.; Maratos-Flier, E.; Herman, M.A. A Critical Role for ChREBP-Mediated FGF21 Secretion in Hepatic Fructose Metabolism. Mol. Metab. 2017, 6, 14–21. [Google Scholar] [CrossRef]
  119. Lee, H.A.; Kim, H.Y. Therapeutic Mechanisms and Clinical Effects of Glucagon-like Peptide 1 Receptor Agonists in Nonalcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2023, 24, 9324. [Google Scholar] [CrossRef]
  120. Yabut, J.M.; Drucker, D.J. Glucagon-like Peptide-1 Receptor-Based Therapeutics for Metabolic Liver Disease. Endocr. Rev. 2023, 44, 14–32. [Google Scholar] [CrossRef]
  121. Perry, R.J.; Zhang, D.; Guerra, M.T.; Brill, A.L.; Goedeke, L.; Nasiri, A.R.; Rabin-Court, A.; Wang, Y.; Peng, L.; Dufour, S.; et al. Glucagon Stimulates Gluconeogenesis by INSP3R1-Mediated Hepatic Lipolysis. Nature 2020, 579, 279–283. [Google Scholar] [CrossRef]
  122. Kim, T.; Nason, S.; Holleman, C.; Pepin, M.; Wilson, L.; Berryhill, T.F.; Wende, A.R.; Steele, C.; Young, M.E.; Barnes, S.; et al. Glucagon Receptor Signaling Regulates Energy Metabolism via Hepatic Farnesoid X Receptor and Fibroblast Growth Factor 21. Diabetes 2018, 67, 1773–1782. [Google Scholar] [CrossRef] [PubMed]
  123. Lv, S.; Qiu, X.; Li, J.; Liang, J.; Li, W.; Zhang, C.; Zhang, Z.-N.; Luan, B. Glucagon-Induced Extracellular CAMP Regulates Hepatic Lipid Metabolism. J. Endocrinol. 2017, 234, 73–87. [Google Scholar] [CrossRef]
  124. Gutierrez, J.A.; Liu, W.; Perez, S.; Xing, G.; Sonnenberg, G.; Kou, K.; Blatnik, M.; Allen, R.; Weng, Y.; Vera, N.B.; et al. Pharmacologic Inhibition of Ketohexokinase Prevents Fructose-Induced Metabolic Dysfunction. Mol. Metab. 2021, 48, 101196. [Google Scholar] [CrossRef] [PubMed]
  125. Helsley, R.N.; Park, S.H.; Vekaria, H.J.; Sullivan, P.G.; Conroy, L.R.; Sun, R.C.; del Mar Romero, M.; Herrero, L.; Bons, J.; King, C.D.; et al. Ketohexokinase-C Regulates Global Protein Acetylation to Decrease Carnitine Palmitoyltransferase 1a-Mediated Fatty Acid Oxidation. J. Hepatol. 2023, 79, 25–42. [Google Scholar] [CrossRef]
  126. Futatsugi, K.; Smith, A.C.; Tu, M.; Raymer, B.; Ahn, K.; Coffey, S.B.; Dowling, M.S.; Fernando, D.P.; Gutierrez, J.A.; Huard, K.; et al. Discovery of PF-06835919: A Potent Inhibitor of Ketohexokinase (KHK) for the Treatment of Metabolic Disorders Driven by the Overconsumption of Fructose. J. Med. Chem. 2020, 63, 13546–13560. [Google Scholar] [CrossRef] [PubMed]
  127. Chouchani, E.T.; Kazak, L.; Spiegelman, B.M. New Advances in Adaptive Thermogenesis: UCP1 and Beyond. Cell Metab. 2019, 29, 27–37. [Google Scholar] [CrossRef]
  128. Brand, M.D.; Nicholls, D.G. Assessing Mitochondrial Dysfunction in Cells. Biochem. J. 2011, 435, 297–312. [Google Scholar] [CrossRef]
  129. Perry, R.J.; Zhang, D.; Zhang, X.-M.; Boyer, J.L.; Shulman, G.I. Controlled-Release Mitochondrial Protonophore Reverses Diabetes and Steatohepatitis in Rats. Science 2015, 347, 1253–1256. [Google Scholar] [CrossRef]
  130. Vlasova, K.Y.; Ostroverkhov, P.; Vedenyapina, D.; Yakimova, T.; Trusova, A.; Lomakina, G.Y.; Vodopyanov, S.S.; Grin, M.; Klyachko, N.; Chekhonin, V.; et al. Liposomal Form of 2,4-Dinitrophenol Lipophilic Derivatives as a Promising Therapeutic Agent for ATP Synthesis Inhibition. Nanomaterials 2022, 12, 2162. [Google Scholar] [CrossRef]
  131. Yoshida, S.; Ikenaga, N.; Liu, S.B.; Peng, Z.-W.; Chung, J.; Sverdlov, D.Y.; Miyamoto, M.; Kim, Y.O.; Ogawa, S.; Arch, R.H.; et al. Extrahepatic Platelet-Derived Growth Factor-β, Delivered by Platelets, Promotes Activation of Hepatic Stellate Cells and Biliary Fibrosis in Mice. Gastroenterology 2014, 147, 1378–1392. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, H.; Cai, W.; Chu, E.S.H.; Tang, J.; Wong, C.-C.; Wong, S.H.; Sun, W.; Liang, Q.; Fang, J.; Sun, Z.; et al. Hepatic Cyclooxygenase-2 Overexpression Induced Spontaneous Hepatocellular Carcinoma Formation in Mice. Oncogene 2017, 36, 4415–4426. [Google Scholar] [CrossRef]
  133. Paik, Y.-H.; Kim, J.K.; Lee, J.I.; Kang, S.H.; Kim, D.Y.; An, S.H.; Lee, S.J.; Lee, D.K.; Han, K.-H.; Chon, C.Y.; et al. Celecoxib Induces Hepatic Stellate Cell Apoptosis through Inhibition of Akt Activation and Suppresses Hepatic Fibrosis in Rats. Gut 2009, 58, 1517–1527. [Google Scholar] [CrossRef] [PubMed]
  134. Malehmir, M.; Pfister, D.; Gallage, S.; Szydlowska, M.; Inverso, D.; Kotsiliti, E.; Leone, V.; Peiseler, M.; Surewaard, B.G.J.; Rath, D.; et al. Platelet GPIbα Is a Mediator and Potential Interventional Target for NASH and Subsequent Liver Cancer. Nat. Med. 2019, 25, 641–655. [Google Scholar] [CrossRef]
  135. Raj, H.; Durgia, H.; Palui, R.; Kamalanathan, S.; Selvarajan, S.; Kar, S.S.; Sahoo, J. SGLT-2 Inhibitors in Non-Alcoholic Fatty Liver Disease Patients with Type 2 Diabetes Mellitus: A Systematic Review. World J. Diabetes 2019, 10, 114–132. [Google Scholar] [CrossRef]
  136. Hsiang, J.C.; Wong, V.W.-S. SGLT2 Inhibitors in Liver Patients. Clin. Gastroenterol. Hepatol. 2020, 18, 2168–2172.e2. [Google Scholar] [CrossRef] [PubMed]
  137. Mammi, C.; Calanchini, M.; Antelmi, A.; Cinti, F.; Rosano, G.M.C.; Lenzi, A.; Caprio, M.; Fabbri, A. Androgens and Adipose Tissue in Males: A Complex and Reciprocal Interplay. Int. J. Endocrinol. 2012, 2012, 789653. [Google Scholar] [CrossRef]
  138. Alemany, M. Steroid Hormones Interrelationships in the Metabolic Syndrome: An Introduction to the Ponderostat Hypothesis. Hormones 2012, 11, 272–289. [Google Scholar] [CrossRef]
  139. Shen, M.; Shi, H. Sex Hormones and Their Receptors Regulate Liver Energy Homeostasis. Int. J. Endocrinol. 2015, 2015, 294278. [Google Scholar] [CrossRef]
  140. Sarkar, M.; Yates, K.; Suzuki, A.; Lavine, J.; Gill, R.; Ziegler, T.; Terrault, N.; Dhindsa, S. Low Testosterone Is Associated With Nonalcoholic Steatohepatitis and Fibrosis Severity in Men. Clin. Gastroenterol. Hepatol. 2021, 19, 400–402.e2. [Google Scholar] [CrossRef]
  141. Karim, G.; Bansal, M.B. Resmetirom: An Orally Administered, Small-Molecule, Liver-Directed, β-Selective THR Agonist for the Treatment of Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Eur. Endocrinol. 2023, 19, 60. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Progression of MAFLD according to the “multiple-hit” theory.
Figure 1. Progression of MAFLD according to the “multiple-hit” theory.
Ijms 26 06518 g001
Figure 2. Non-invasive evaluation of liver fibrosis risk.
Figure 2. Non-invasive evaluation of liver fibrosis risk.
Ijms 26 06518 g002
Figure 3. Main molecular effects of new, experimental drugs in MASLD.
Figure 3. Main molecular effects of new, experimental drugs in MASLD.
Ijms 26 06518 g003
Table 1. Contemporary pharmacological treatment of MASLD.
Table 1. Contemporary pharmacological treatment of MASLD.
DrugDoseMechanism of ActionHistological EffectNotable Side Effects
Pioglitazone [13,15,38,39,40]30–45 mg/dayPPAR-γ agonist; improves insulin sensitivity, reduces hepatic lipotoxicityImproves steatosis, inflammation, and hepatocyte ballooningWeight gain, fluid retention, possible increased risk of bladder cancer
Vitamin E [15,38,41,42]800 IU/dayAntioxidant; neutralizes ROS, reduces oxidative stress and hepatocyte injuryImproves steatohepatitis, no effect on fibrosisPossible increased risk of prostate cancer
Ursodeoxycholic acid [43,44,45,46,47,48]13–15 mg/kg/dayHydrophilic bile acid; cytoprotective, anti-inflammatory, reduces bile acid toxicityUncertain; a small number of studies show biochemical but not histological improvement Diarrhea
Table 2. A summary of experimental drugs used in treatment of MASLD.
Table 2. A summary of experimental drugs used in treatment of MASLD.
Name ReferenceNCTMechanism of ActionDuration/PhaseResults PostedEffect Population
MK-4074[49]NCT01431521Dual acetyl-CoA carboxylase (ACC1 and -2) inhibitor4 weeks, 12016 steatosis; no effect; AST, ALTObese, NAFLD
Clesacostat PF-05221304[50]NCT03248882Dual acetyl-CoA carboxylase (ACC1 and -2) inhibitor16 weeks, 2a2020 steatosis, ALTOverweight/obese, NAFLD, or NASH
Clesacostat PF-05221304[51]NCT03776175Dual acetyl-CoA carboxylase (ACC1 and -2) inhibitor6 weeks, 2a2020 steatosisOverweight/obese NAFLD, T2DM, or other metabolic syndrome comorbidities
Clesacostat PF-05221304[52]NCT04399538Dual acetyl-CoA carboxylase (ACC1 and -2) inhibitor6 weeks, 2a2023 steatosis; TGOverweight/obese NAFLD
AXA1125 & AXA1957[53]NCT04073368Amino acids and NAC16 weeks, N/A2021 steatosis, ALT; AE; no effect: AST, HOMA-IRNASH
PXL770[54]NCT03763877AMPK activator12 weeks, 22021 steatosis, ALT, AST, HbA1c, Fib4; no effect: lipid profile, weightOverweight/obese NAFLD
Metformin[55]NCT00063232AMPK activator48 weeks, 22011 inflammation, ALT, HOMA-IRNASH
Aramchol[56]NCT02279524Bile salt fatty acid conjugate52 weeks, 2b2021 steatosis, fibrosis, ALT, AST, HbA1cOverweight/obese NASH
Elobixibat NCT04006145Inhibitor of the ileal bile acid transporter16 weeks, 22021 LDLNAFLD/NASH
Pradigastat LCQ908 NCT01811472DGAT1 Inhibitor24 weeks, 22016 steatosis, ALT, weight, WC; no effect: AST, GGTP, TGNAFLD
Ervogastat PF-06865571[51]NCT03776175DGAT2 inhibitor6 weeks, 2a2020 steatosisOverweight/obese NAFLD, T2DM, or other metabolic syndrome comorbidities
Ervogastat PF-06865571[52]NCT04399538DGAT2 inhibitor6 weeks, 2a2023 steatosis; TGOverweight/obese NAFLD
Denifanstat TVB-2640[57]NCT03938246FAS inhibitor12 weeks, 22024 steatosis, ALTOverweight/obese NAFLD with stage 1-3 fibrosis
Icosabutate[58]NCT04052516Free fatty acid receptor 4 (FFAR4) agonist52 weeks, 2b2025 ALT, GGTP, AST, HOMA-IR, bilirubin, hsCRP, pro-C3; no effect: steatosisNASH
Efruxifermin[59,60,61]NCT03976401FGF21 analog12 weeks, 2a2022 steatosis, ALT, liver stiffness, pro-C3Overweight/obese, NAFLD
Pegbelfermin BMS-986036[62,63]NCT02413372Pegylated FGF2116 weeks, 22020 steatosisOverweight/obese, NASH
MK-3655[64]NCT04583423Humanized mAb that binds KLB FGFR1c/β-klotho activator16 weeks, 2 2024No effectOverweight/obese, NASH
Obeticholic acid INT-747[65,66,67,68]NCT01265498FXR agonist72 weeks, 22015 histological steatosis, ALT, AST, GGTP, total bilirubin, HDL, body weight, BMI; alkaline phosphatase, HOMA-IR, total cholesterol, LDL; no effect: TG, HbA1c, WC, blood pressureNASH
Obeticholic acid INT-747[69]NCT00501592FXR agonist6 weeks, 22012 ALT, GGTPT2DM, NAFLD
TERN-101 NCT04328077FXR agonist12 weeks, 22022 ALTOverweight/obese, NASH
Vonafexor[70]NCT03812029FXR Agonist12 weeks, 2a2023 steatosis, ALT, GGTP, body weight, WCF2/F3 fibrosis, NASH
EDP-305[71]NCT03421431FXR agonist12 weeks, 22021 steatosis, ALT, increase ApoA1; no effect: lipid profile, AboB, ApoC3, HOMA-IR, HbA1cNASH
Semaglutide[72,73]NCT03987451GLP-1 agonist48 weeks, 22024 steatosis, ALT, AST, GGTP, HbA1c, HOMA-IR, body weight, BMIOverweight/obese, NASH
Semaglutide[74]NCT02970942GLP-1 agonist72 weeks, 22021 steatosis, ALT, AST, GGTP, HbA1c, body weightNASH
Semaglutide[75]NCT04216589GLP-1 agonist24 weeks, 22024 steatosis, HbA1c, HOMA-IR, body weight, BMI, WCNAFLD, HIV infection
Semaglutide[76]NCT04944992GLP-1 agonist24 weeks, 22023 steatosis, weight, tot. chol. LDL, TG, apoB; HDLOverweight/obese, NAFLD
Tirzepatide[77]NCT04166773Dual GLP-1/GIP agonist56 weeks, 22025 steatosis, fibrosis, body weightOverweight/obese, NASH
Efinopegdutide[76]NCT04944992Dual GLP-1/glucagon receptor agonist24 weeks, 22023 steatosis, weight, tot. chol. LDL, TG, apoB, HDLOverweight/obese, NAFLD
Survodutide BI456906[78]NCT04771273Dual GLP-1/glucagon receptor agonist48 weeks, 22024 steatosis, fibrosisNASH or NAFLD with fibrosis stages F1-F3
Cotadutide MEDI0382[79]NCT04019561Dual GLP-1/glucagon receptor agonist23 days, 22022 steatosis, ALT, AST, body weight, BMIOverweight/obese, NAFLD/NASH
PF-06835919[80,81]NCT03969719Ketohexokinase inhibitor16 weeks, 2a2022 steatosis, ALT, HbA1c; no change: HOMA-IRNAFLD, T2DM
HU6[82]NCT04874233Mitochondrial uncoupler, prodrug of 2,4-dinitrophenol (DNP) 61 days, 2a2024 steatosisOverweight/obese, NAFLD
Aspirin[83]NCT04031729Nonsteroidal anti-inflammatory drug (NSAID)6 months,1/22024 steatosisNAFLD
dR-pioglitazone PXL065[84,85]NCT04321343Deuterium-stabilized R-stereoisomer of pioglitazone (PPARγ agonist)36 weeks, 22023 steatosis, fibrosis, ALT, AST, GGTP, HbA1c, HOMA-IRNAFLD and fibrosis score F1, F2, or F3
Saroglitazar[86]NCT03061721Dual PPAR α/γ agonist16 weeks, 22024 steatosis, fibrosis, ALTOverweight/obese, NAFLD
Saroglitazar[87]NCT03863574Dual PPAR α/γ agonist24 weeks, 22024 ALT, GGTP, AST, TGOverweight/obese, NASH
Lanifibranor IVA337[88,89,90,91]NCT03008070Pan-PPAR agonist24 weeks, 22021 fibrosis, ALT, AST, GGTP, hsCRP, HOMA, HbA1c, TG, ApoA1; no effect: total cholesterol and LDL; HDL, adiponectine NASH
Lanifibranor IVA337[88,89,90,91]NCT03459079Pan-PPAR agonist24 weeks, 22024 steatosis, fibrosis, HbA1c, hepatic IR, fibrosis, HDLNAFLD
Licogliflozin LIK066[92]NCT03205150SGLT1 and SGLT2 inhibitor12 weeks, 22021 steatosis, ALT, AST, body weightNASH
Lipocine LPCN 1144[93]NCT04134091Prodrug of testosterone 36 weeks, 22023 steatosis, fibrosis, ALT, AST, ALP, GGTPNASH
TERN-501 NCT05415722THR-β agonist12 weeks, 22025 steatosisOverweight/obese, NASH
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Drygalski, K. Pharmacological Treatment of MASLD: Contemporary Treatment and Future Perspectives. Int. J. Mol. Sci. 2025, 26, 6518. https://doi.org/10.3390/ijms26136518

AMA Style

Drygalski K. Pharmacological Treatment of MASLD: Contemporary Treatment and Future Perspectives. International Journal of Molecular Sciences. 2025; 26(13):6518. https://doi.org/10.3390/ijms26136518

Chicago/Turabian Style

Drygalski, Krzysztof. 2025. "Pharmacological Treatment of MASLD: Contemporary Treatment and Future Perspectives" International Journal of Molecular Sciences 26, no. 13: 6518. https://doi.org/10.3390/ijms26136518

APA Style

Drygalski, K. (2025). Pharmacological Treatment of MASLD: Contemporary Treatment and Future Perspectives. International Journal of Molecular Sciences, 26(13), 6518. https://doi.org/10.3390/ijms26136518

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop