Abstract

Current treatment paradigms for metabolic dysfunction-associated steatohepatitis (MASH) are based primarily on dietary restrictions and the use of existing drugs, including anti-diabetic and anti-obesity medications. Given the limited number of approved drugs specifically for MASH, recent efforts have focused on promising strategies that specifically target hepatic lipid metabolism, inflammation, fibrosis, or a combination of these processes. In this review, we examined the pathophysiology underlying the development of MASH in relation to recent advances in effective MASH therapy. Particularly, we analyzed the effects of lipogenesis inhibitors, nuclear receptor agonists, glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists, fibroblast growth factor mimetics, and combinatorial therapeutic approaches. We summarize these targets along with their preclinical and clinical candidates with the ultimate goal of optimizing the therapeutic prospects for MASH.

Introduction

Nonalcoholic fatty liver disease (NAFLD) is a major chronic liver disease characterized by excess lipid accumulation, inflammation, and hepatocyte injury [1–3]. Notably, the incidence of NAFLD is increasing, and this disease is estimated to affect approximately 25% of the adult population worldwide. This pattern is consistently associated with global increases in metabolic syndrome, obesity, and diabetes [4]. The global burden of NAFLD is projected to increase two to three times by 2030 [5, 6]. For example, in the USA, the number of NAFLD cases is expected to reach 100 million by 2030, up from 80 million in 2015 [7, 8]. Similarly, China experienced a sharp increase in NAFLD incidence in a short period from 2008 to 2018, with the highest number of patients worldwide and a prevalence of 29.2% [9]. As a result, a significant proportion of simple steatosis progresses to nonalcoholic steatohepatitis (NASH), a more severe form of NAFLD. Recently, metabolic dysfunction-associated steatotic liver disease (MASLD), which has been described as the hepatic manifestation of metabolic syndrome and a continuum from obesity to a series of metabolic disorders, was suggested as a more appropriate overarching term for NAFLD [10].

Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as NASH, is characterized by excess lipid accumulation (steatosis), inflammation, injury, and fibrosis in the liver and if left uncontrolled, can lead to cirrhosis or liver cancer [8, 11]. Steatosis accounts for the virulent nature of MASH and initiates its development. Increased fat in the liver leads to the accumulation of hepatocyte injury inducers (e.g., lipotoxicity), which triggers inflammatory responses and immune cell infiltration into the liver. Liver fibrosis progressively develops through the activation of hepatic stellate cells (HSCs), which are the major sites of fibrogenesis [12]. Notably, MASH is the fastest growing cause of hepatocellular carcinoma (HCC) in liver transplant candidates and age-adjusted liver cancer deaths worldwide [8]. Therefore, immediate action and increased awareness are needed to address the growing prevalence and risks associated with MASH.

In recent years, our understanding of the pathophysiology and management of MASLD has advanced significantly. For patients with simple steatosis, lifestyle interventions such as exercise, dietary changes, and weight loss are the main treatment strategies used to mitigate the progression of MASLD [13, 14]. However, lifestyle adjustments alone are not enough to reverse more advanced MASLD with severe inflammation and fibrosis. Therefore, the development and use of drug therapies in combination with lifestyle interventions are essential. To date, the groundbreaking approval of the thyroid hormone receptor beta (THRβ) agonist resmetirom (MGL-3196) by the United States Food and Drug Administration (FDA) in 2024 represents the first-ever selective treatment for MASH. Currently, numerous clinical trials are underway, raising the expectation of more therapeutic breakthroughs [13, 15–18]. This review summarizes MASH treatment strategies in clinical trials and introduces several novel therapies for MASH, thereby contributing to the advancement of knowledge in this critical area.

The main features and pathological processes of MASH

Insulin resistance, diabetes, obesity, and hyperlipidemia are significant contributors to the pathophysiology of MASH. The “multiple hit” hypothesis accounts for the simultaneous effects of multiple injuries on predisposed individuals and offers a more accurate understanding of the pathogenesis of MASH. These injuries contribute to the three main pathological characteristics of MASH, namely, excessive lipid accumulation (steatosis), immune cell infiltration (inflammation), and fibrosis caused by HSC activation.

Excessive lipid accumulation induces steatosis and lipotoxicity

The liver plays a unique role in lipid metabolism and maintains lipid concentrations at normal levels [19]. Fat accumulation in the liver occurs through various mechanisms, including adipose tissue lipolysis, de novo lipogenesis (DNL), and dietary fat absorption [20]. In insulin resistance, excessive activation of lipolysis in adipose tissue increases circulating levels of free fatty acids (FFAs), which ultimately promotes fat deposition in the liver and contributes to fatty liver disease [21]. In addition, excess intake of dietary carbohydrates promotes hepatic DNL in the liver [22–24]. Indeed, isotope-labeling studies have shown that liver DNL is significantly upregulated in individuals with MASLD [25]. The contribution of DNL to liver triglyceride (TG) synthesis in the fed state is approximately three to six times greater in the livers of obese MASLD patients than in those of normal individuals [25, 26].

Liver DNL is driven by increased consumption of acarbohydrate-rich diet (e.g., fructose), or in part, by dysregulated transcriptional regulation of hepatic lipogenesis under conditions of insulin resistance [23]. Two important transcription factors regulate the enzymes that catalyze lipogenesis in the liver: sterol regulatory element-binding protein 1c (SREBP1c) and carbohydrate regulatory element-binding protein (ChREBP). SREBP1c-mediated target gene transcription is activated by insulin stimulation, while ChREBP is activated by carbohydrate metabolites, which accumulate in the livers of individuals with MASLD [24]. SREBP1c and ChREBP increase the expression of several lipogenic genes, including ketohexokinase (KHK), acetyl coenzyme A (acetyl-CoA) carboxylase (ACC), fatty acid synthase (FASN), and stearoyl-CoA desaturase 1 (SCD-1). In parallel with lipogenesis, defects in the β-oxidation of mitochondrial fatty acids (FAs) in the liver also contribute to the development of hepatic steatosis and MASH progression [27, 28]. It is crucial to emphasize the important role of cholesterol and cholesterol esters as essential factors in the progression from steatosis to MASH. Excess cholesterol can significantly exacerbate cellular toxicity as well as proinflammatory and profibrotic effects in hepatocytes, immune cells, and HSCs [29]. Inhibiting cholesterol biogenesis and absorption or increasing cholesterol efflux is expected to attenuate MASH [30–33]. THRβ agonists, which facilitate FA degradation and cholesterol biosynthesis, collectively exert remarkable therapeutic effects that lead to improvements in MASH [34, 35].

Lipotoxicity may be accompanied by organelle dysfunction, cellular apoptosis, or necrosis, and is closely related to chronic inflammation. Saturated FFAs, such as palmitate (C16:0) and stearate (C18:0), exert direct cytotoxic effects. Furthermore, sphingolipid levels are increased in MASH [36, 37], and ceramide has recently been linked to the pathophysiology of this disease [38]. Hepatocyte apoptosis is significantly increased in patients and animals with MASH, is positively correlated with disease severity, and is considered one of the pathologic hallmarks of MASH [39]. Apoptotic caspase deficiency (e.g., caspase-8 and caspase-3) has been reported to protect against liver injury and fibrosis in murine models of MASH [40, 41]. Consequently, the development of drugs against lipotoxicity and cell death in MASLD has recently attracted increased attention.

Immune cell infiltration accelerates the process of MASH

Excess lipids disrupt the normal function of hepatocytes and trigger endoplasmic reticulum (ER) stress, mitochondrial dysfunction, and production of reactive oxygen species (ROS) [15]. These events activate critical signaling pathways, including the c-Jun N-terminal kinase (JNK) pathway and nuclear factor-kappa light chain enhancer of activated B cells (NF-κB) pathway, thereby regulating gene expression to induce inflammation and apoptosis, which are the main drivers of MASH progression [42, 43].

In livers, Kupffer cells (KCs) are the main source of inflammatory cytokines. KCs sense intestinal bacterial products and endogenous substances released by damaged cells via Toll-like receptors (TLR4, TLR2, and TLR9) and nuclear nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs). This recognition triggers the nuclear translocation of NF-κB and the production of various proinflammatory and profibrotic cytokines, including tumor necrosis factor-alpha (TNFα), interleukin-1beta (IL-1β), C-C motif chemokine ligand 5 (CCL5), and transforming growth factor-beta (TGF-β) [44, 45]. Neutrophil infiltration from the circulation is closely associated with MASLD/MASH, which involves abnormal inflammatory leukocytes. Furthermore, adaptive immunity also promotes MASLD/MASH development, which is characterized by increased T helper type 17 (Th17) cells and IL-17A expression [46, 47]. Additionally, the inhibition of C-C motif chemokine receptors (CCRs) is a common therapeutic approach. Based on these findings, novel therapeutics targeting alternative proinflammatory pathways, such as those involving IL-27, IL-17, IL-11, IL-1, and TNFα, have emerged as promising candidates for the treatment of MASLD/MASH [18, 48]. Targeting self-aggressive CD8+ T cells and antiplatelet therapy are also potential strategies for alleviating MASLD or preventing its progression to HCC [49].

HSC activation promotes the development of fibrosis

MASLD can progress from MASH with mild symptoms to irreversible liver fibrosis and even liver cancer. Therefore, effective intervention for liver fibrosis is the most important step to prevent the progression of MASH to cirrhosis and HCC [50, 51]. HSCs are central to liver fibrosis, and their subsets either regulate immune mechanisms through chemokines and cytokines or differentiate into matrix-producing myofibroblasts.

Among several growth factors that play key roles in the development of fibrosis induced by HSCs, TGF-β is the most potent profibrotic cytokine. TGF-β is secreted in a latent form by various hepatocyte populations and is partially activated by HSCs that express integrin αv [52]. The inhibition of TGF-β activation by targeting cell surface integrin subpopulations that contain αv (e.g., integrins αvβ1, αvβ3, αvβ5, αvβ6, and αvβ8) is considered a promising strategy [53, 54]. Another cytokine that promotes fibrosis is platelet-derived growth factor (PDGF). HSCs express high levels of PDGF receptors, and activation of HSCs can induce their proliferation and migration, thus enhancing their role in liver fibrosis. Consistent with this mechanism, multikinase inhibitors that specifically target PDGF and its isoforms have shown the ability to counteract liver fibrosis [55]. Furthermore, vascular endothelial growth factor (VEGF) produced by hepatocytes induces the activation and proliferation of HSCs, which leads to increased production of extracellular matrix (ECM) proteins and TGF-β, thereby inducing and exacerbating liver fibrosis [56]. These results suggest that inhibition of HSC activation by cytokine stimulation may be an effective approach to combat liver fibrosis.

In addition to activation of HSCs by cytokines and other factors, immune cell-mediated killing of hepatocytes also contributes to liver fibrosis. Profibrotic inflammatory chemokines and their receptors, such as CCL2, CCL21, IL-8, IL-17, IL-22, C-X-C motif chemokine ligand 9 (CXCL9), CXCL10, CXCL11, and C-X-C motif chemokine receptor 1 (CXCR1), are involved in this process [57]. As sustained activation of immune response further aggravates liver fibrosis, inhibition of specific chemokines and NF-κB can attenuate the progression of liver fibrosis [58].

Advancements in the development of innovative anti-MASH drugs

Lipid accumulation, inflammation, and fibrosis are the primary pathological features of MASH. Based on extensive research of these pathological mechanisms, the development of innovative therapeutics for MASH is promising. However, the need for pathologic biopsy analysis poses a significant challenge to clinical trials. Currently, clinical trial endpoints for MASH are mandated to meet one of the following criteria: (i) resolution of MASH without progression to fibrosis or (ii) regression of fibrosis without exacerbation of MASH. Despite the challenges in anti-MASH drug development, the FDA’s approval of the oral small-molecule resmetirom for the treatment of MASH in March 2024 has provided confidence in clinical interventions for MASH patients and has significantly strengthened the advancement of anti-MASH drug development. Given the variety of possible pharmacological therapies, therapeutic options can be comprehensively divided into metabolic, anti-inflammatory, and antifibrotic strategies. In addition, various combination therapies have been extensively explored to optimize therapeutic efficacy while minimizing potential drug-related side effects (Fig. 1; Table 1).

Table 1.

List of several clinical trial candidate drugs for MASH.

StrategyAgentTargetLatest phaseNCTLatest outcomeRef
Nuclear receptor agonists
ResmetriomTHRβ agonistApprovedNCT04951219
NCT04197479
NCT05500222
NCT03900429
↓ Hepatic steatosis
↓ Liver fibrosis
↓ Plasma LDL-C, TG
[35, 59]
VK2809THRβ agonistPhase IINCT04173065
NCT02927184
↓ Hepatic steatosis
↓ Plasma LDL-C, TG
[60]
ASC41THRβ agonistPhase IINCT05462353↓ Hepatic steatosis
↓ Plasma LDL-C, TG
TERN-501THRβ agonistPhase IINCT05415722↓ Hepatic steatosis
LanifibranorPPARα/δ/γ agonistPhase IIINCT04849728
NCT05232071
NCT03459079
↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[61]
ElafibranorPPARα/δ agonistPhase IIINCT02704403
NCT03883607
NCT01694849
↓ Liver fibrosis
↓ Inflammation
[62]
SaroglitazarPPARα/γ agonistPhase IINCT03061721
NCT03863574
↓ Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[63]
PioglitazonePPARγ agonistPhase IVNCT00994682↓Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[64, 65]
PemafibratePPARα agonistPhase IINCT05327127
NCT03350165
↓ MRE-based liver stiffness[66, 67]
Obeticholic acidFXR agonistPhase IIINCT02548351
NCT01265498
↓ Hepatic steatosis
↓ Liver fibrosis
↑ Pruritus
[68, 69]
CilofexorFXR agonistPhase IINCT02854605↓ Hepatic steatosis
↓ Serum bile acids
[70, 71]
HPG1860FXR agonistPhase IINCT05338034↓ Liver fat content
TropifexorFXR agonistPhase IINCT02855164
NCT04147195
NCT03517540
↓ Liver fat content[72]
TERN-101FXR agonistPhase IINCT04328077↓ Corrected T1 (cT1)
MET409FXR agonistPhase IINCT04702490↓ Hepatic steatosis
CS0159FXR agonistPhase IINCT05591079No results posted
GLP-1Ragonists
SemaglutideGLP-1R agonistPhase IIINCT04822181
NCT02970942
↓ Body weight
↓ Hepatic steatosis
[73]
DulaglutideGLP-1R agonistPhase IVNCT03590626↓ Hepatic steatosis[74]
LiraglutideGLP-1R agonistPhase IINCT01237119
NCT02654665
↓ Hepatic steatosis[75–77]
Lipogenesis inhibitors
GS-0976ACC inhibitorPhase IINCT02856555↓ Hepatic steatosis
↑ Plasma TG
[78]
MK-4074ACC inhibitorPhase IINCT01431521↓ Hepatic steatosis
↑ Plasma TG
[79]
DenifanstatFASN inhibitorPhase IINCT04906421↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[80]
AramcholSCD-1 inhibitorPhase IIINCT04104321↓ Hepatic steatosis
↓ Plasma ALT and AST
[81]
Bempedoic acidACLY inhibitorPhase IINCT06035874Recruiting[82]
BGT-002ACLY inhibitorPhase IICTR20230344Recruiting
Fibroblastgrowth factormimetics
EfruxiferminFGF21 analoguePhase IIINCT06215716
NCT06161571
NCT03976401
NCT04767529
↓ Hepatic steatosis
↓ Plasma ALT and AST
[83–85]
PegozaferminFGF21 analoguePhase IINCT03486912
NCT03486899
NCT03400163
NCT02413372
↓ Hepatic steatosis[86, 87]
Potential of emergingtherapeutic strategies
MSDC-0602KMPC inhibitorPhase IINCT02784444↓ Fasting glucose
↓ HbA1c
[88]
CenicrivirocCCR2/CCR5 inhibitorPhase IIINCT03028740↓ Inflammation
↓ Liver fibrosis
[89, 90]
SelonsertibASK1 inhibitorPhase IIINCT03053063
NCT03053050
↓ Inflammation
↓ Fibrosis
[91, 92]
Combinationtherapy
PF-05221304/PF-06865571ACC inhibitor/DGAT2 inhibitorPhase IINCT04321031
NCT03248882
NCT03776175
↓ Hepatic steatosis
(−) Plasma TG
[93]
Selonsertib/firsocostat/cilofexorASK1 inhibitor/ACC inhibitor/FXR agonistPhase IINCT02781584↓ Hepatic steatosis
(−) Plasma TG
[70]
Obeticholic acid/ atorvastatinFXR agonist/HMGCR inhibitorPhase IINCT02633956(−) LDL-C[83–85, 94]
Cilofexor/semaglutideFXR agonist/GLP-1R agonistPhase IINCT03987074↓ Hepatic steatosis[71, 86, 87]
StrategyAgentTargetLatest phaseNCTLatest outcomeRef
Nuclear receptor agonists
ResmetriomTHRβ agonistApprovedNCT04951219
NCT04197479
NCT05500222
NCT03900429
↓ Hepatic steatosis
↓ Liver fibrosis
↓ Plasma LDL-C, TG
[35, 59]
VK2809THRβ agonistPhase IINCT04173065
NCT02927184
↓ Hepatic steatosis
↓ Plasma LDL-C, TG
[60]
ASC41THRβ agonistPhase IINCT05462353↓ Hepatic steatosis
↓ Plasma LDL-C, TG
TERN-501THRβ agonistPhase IINCT05415722↓ Hepatic steatosis
LanifibranorPPARα/δ/γ agonistPhase IIINCT04849728
NCT05232071
NCT03459079
↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[61]
ElafibranorPPARα/δ agonistPhase IIINCT02704403
NCT03883607
NCT01694849
↓ Liver fibrosis
↓ Inflammation
[62]
SaroglitazarPPARα/γ agonistPhase IINCT03061721
NCT03863574
↓ Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[63]
PioglitazonePPARγ agonistPhase IVNCT00994682↓Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[64, 65]
PemafibratePPARα agonistPhase IINCT05327127
NCT03350165
↓ MRE-based liver stiffness[66, 67]
Obeticholic acidFXR agonistPhase IIINCT02548351
NCT01265498
↓ Hepatic steatosis
↓ Liver fibrosis
↑ Pruritus
[68, 69]
CilofexorFXR agonistPhase IINCT02854605↓ Hepatic steatosis
↓ Serum bile acids
[70, 71]
HPG1860FXR agonistPhase IINCT05338034↓ Liver fat content
TropifexorFXR agonistPhase IINCT02855164
NCT04147195
NCT03517540
↓ Liver fat content[72]
TERN-101FXR agonistPhase IINCT04328077↓ Corrected T1 (cT1)
MET409FXR agonistPhase IINCT04702490↓ Hepatic steatosis
CS0159FXR agonistPhase IINCT05591079No results posted
GLP-1Ragonists
SemaglutideGLP-1R agonistPhase IIINCT04822181
NCT02970942
↓ Body weight
↓ Hepatic steatosis
[73]
DulaglutideGLP-1R agonistPhase IVNCT03590626↓ Hepatic steatosis[74]
LiraglutideGLP-1R agonistPhase IINCT01237119
NCT02654665
↓ Hepatic steatosis[75–77]
Lipogenesis inhibitors
GS-0976ACC inhibitorPhase IINCT02856555↓ Hepatic steatosis
↑ Plasma TG
[78]
MK-4074ACC inhibitorPhase IINCT01431521↓ Hepatic steatosis
↑ Plasma TG
[79]
DenifanstatFASN inhibitorPhase IINCT04906421↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[80]
AramcholSCD-1 inhibitorPhase IIINCT04104321↓ Hepatic steatosis
↓ Plasma ALT and AST
[81]
Bempedoic acidACLY inhibitorPhase IINCT06035874Recruiting[82]
BGT-002ACLY inhibitorPhase IICTR20230344Recruiting
Fibroblastgrowth factormimetics
EfruxiferminFGF21 analoguePhase IIINCT06215716
NCT06161571
NCT03976401
NCT04767529
↓ Hepatic steatosis
↓ Plasma ALT and AST
[83–85]
PegozaferminFGF21 analoguePhase IINCT03486912
NCT03486899
NCT03400163
NCT02413372
↓ Hepatic steatosis[86, 87]
Potential of emergingtherapeutic strategies
MSDC-0602KMPC inhibitorPhase IINCT02784444↓ Fasting glucose
↓ HbA1c
[88]
CenicrivirocCCR2/CCR5 inhibitorPhase IIINCT03028740↓ Inflammation
↓ Liver fibrosis
[89, 90]
SelonsertibASK1 inhibitorPhase IIINCT03053063
NCT03053050
↓ Inflammation
↓ Fibrosis
[91, 92]
Combinationtherapy
PF-05221304/PF-06865571ACC inhibitor/DGAT2 inhibitorPhase IINCT04321031
NCT03248882
NCT03776175
↓ Hepatic steatosis
(−) Plasma TG
[93]
Selonsertib/firsocostat/cilofexorASK1 inhibitor/ACC inhibitor/FXR agonistPhase IINCT02781584↓ Hepatic steatosis
(−) Plasma TG
[70]
Obeticholic acid/ atorvastatinFXR agonist/HMGCR inhibitorPhase IINCT02633956(−) LDL-C[83–85, 94]
Cilofexor/semaglutideFXR agonist/GLP-1R agonistPhase IINCT03987074↓ Hepatic steatosis[71, 86, 87]
Table 1.

List of several clinical trial candidate drugs for MASH.

StrategyAgentTargetLatest phaseNCTLatest outcomeRef
Nuclear receptor agonists
ResmetriomTHRβ agonistApprovedNCT04951219
NCT04197479
NCT05500222
NCT03900429
↓ Hepatic steatosis
↓ Liver fibrosis
↓ Plasma LDL-C, TG
[35, 59]
VK2809THRβ agonistPhase IINCT04173065
NCT02927184
↓ Hepatic steatosis
↓ Plasma LDL-C, TG
[60]
ASC41THRβ agonistPhase IINCT05462353↓ Hepatic steatosis
↓ Plasma LDL-C, TG
TERN-501THRβ agonistPhase IINCT05415722↓ Hepatic steatosis
LanifibranorPPARα/δ/γ agonistPhase IIINCT04849728
NCT05232071
NCT03459079
↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[61]
ElafibranorPPARα/δ agonistPhase IIINCT02704403
NCT03883607
NCT01694849
↓ Liver fibrosis
↓ Inflammation
[62]
SaroglitazarPPARα/γ agonistPhase IINCT03061721
NCT03863574
↓ Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[63]
PioglitazonePPARγ agonistPhase IVNCT00994682↓Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[64, 65]
PemafibratePPARα agonistPhase IINCT05327127
NCT03350165
↓ MRE-based liver stiffness[66, 67]
Obeticholic acidFXR agonistPhase IIINCT02548351
NCT01265498
↓ Hepatic steatosis
↓ Liver fibrosis
↑ Pruritus
[68, 69]
CilofexorFXR agonistPhase IINCT02854605↓ Hepatic steatosis
↓ Serum bile acids
[70, 71]
HPG1860FXR agonistPhase IINCT05338034↓ Liver fat content
TropifexorFXR agonistPhase IINCT02855164
NCT04147195
NCT03517540
↓ Liver fat content[72]
TERN-101FXR agonistPhase IINCT04328077↓ Corrected T1 (cT1)
MET409FXR agonistPhase IINCT04702490↓ Hepatic steatosis
CS0159FXR agonistPhase IINCT05591079No results posted
GLP-1Ragonists
SemaglutideGLP-1R agonistPhase IIINCT04822181
NCT02970942
↓ Body weight
↓ Hepatic steatosis
[73]
DulaglutideGLP-1R agonistPhase IVNCT03590626↓ Hepatic steatosis[74]
LiraglutideGLP-1R agonistPhase IINCT01237119
NCT02654665
↓ Hepatic steatosis[75–77]
Lipogenesis inhibitors
GS-0976ACC inhibitorPhase IINCT02856555↓ Hepatic steatosis
↑ Plasma TG
[78]
MK-4074ACC inhibitorPhase IINCT01431521↓ Hepatic steatosis
↑ Plasma TG
[79]
DenifanstatFASN inhibitorPhase IINCT04906421↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[80]
AramcholSCD-1 inhibitorPhase IIINCT04104321↓ Hepatic steatosis
↓ Plasma ALT and AST
[81]
Bempedoic acidACLY inhibitorPhase IINCT06035874Recruiting[82]
BGT-002ACLY inhibitorPhase IICTR20230344Recruiting
Fibroblastgrowth factormimetics
EfruxiferminFGF21 analoguePhase IIINCT06215716
NCT06161571
NCT03976401
NCT04767529
↓ Hepatic steatosis
↓ Plasma ALT and AST
[83–85]
PegozaferminFGF21 analoguePhase IINCT03486912
NCT03486899
NCT03400163
NCT02413372
↓ Hepatic steatosis[86, 87]
Potential of emergingtherapeutic strategies
MSDC-0602KMPC inhibitorPhase IINCT02784444↓ Fasting glucose
↓ HbA1c
[88]
CenicrivirocCCR2/CCR5 inhibitorPhase IIINCT03028740↓ Inflammation
↓ Liver fibrosis
[89, 90]
SelonsertibASK1 inhibitorPhase IIINCT03053063
NCT03053050
↓ Inflammation
↓ Fibrosis
[91, 92]
Combinationtherapy
PF-05221304/PF-06865571ACC inhibitor/DGAT2 inhibitorPhase IINCT04321031
NCT03248882
NCT03776175
↓ Hepatic steatosis
(−) Plasma TG
[93]
Selonsertib/firsocostat/cilofexorASK1 inhibitor/ACC inhibitor/FXR agonistPhase IINCT02781584↓ Hepatic steatosis
(−) Plasma TG
[70]
Obeticholic acid/ atorvastatinFXR agonist/HMGCR inhibitorPhase IINCT02633956(−) LDL-C[83–85, 94]
Cilofexor/semaglutideFXR agonist/GLP-1R agonistPhase IINCT03987074↓ Hepatic steatosis[71, 86, 87]
StrategyAgentTargetLatest phaseNCTLatest outcomeRef
Nuclear receptor agonists
ResmetriomTHRβ agonistApprovedNCT04951219
NCT04197479
NCT05500222
NCT03900429
↓ Hepatic steatosis
↓ Liver fibrosis
↓ Plasma LDL-C, TG
[35, 59]
VK2809THRβ agonistPhase IINCT04173065
NCT02927184
↓ Hepatic steatosis
↓ Plasma LDL-C, TG
[60]
ASC41THRβ agonistPhase IINCT05462353↓ Hepatic steatosis
↓ Plasma LDL-C, TG
TERN-501THRβ agonistPhase IINCT05415722↓ Hepatic steatosis
LanifibranorPPARα/δ/γ agonistPhase IIINCT04849728
NCT05232071
NCT03459079
↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[61]
ElafibranorPPARα/δ agonistPhase IIINCT02704403
NCT03883607
NCT01694849
↓ Liver fibrosis
↓ Inflammation
[62]
SaroglitazarPPARα/γ agonistPhase IINCT03061721
NCT03863574
↓ Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[63]
PioglitazonePPARγ agonistPhase IVNCT00994682↓Insulin resistance
↓ Hepatic steatosis
↓ Liver fibrosis
[64, 65]
PemafibratePPARα agonistPhase IINCT05327127
NCT03350165
↓ MRE-based liver stiffness[66, 67]
Obeticholic acidFXR agonistPhase IIINCT02548351
NCT01265498
↓ Hepatic steatosis
↓ Liver fibrosis
↑ Pruritus
[68, 69]
CilofexorFXR agonistPhase IINCT02854605↓ Hepatic steatosis
↓ Serum bile acids
[70, 71]
HPG1860FXR agonistPhase IINCT05338034↓ Liver fat content
TropifexorFXR agonistPhase IINCT02855164
NCT04147195
NCT03517540
↓ Liver fat content[72]
TERN-101FXR agonistPhase IINCT04328077↓ Corrected T1 (cT1)
MET409FXR agonistPhase IINCT04702490↓ Hepatic steatosis
CS0159FXR agonistPhase IINCT05591079No results posted
GLP-1Ragonists
SemaglutideGLP-1R agonistPhase IIINCT04822181
NCT02970942
↓ Body weight
↓ Hepatic steatosis
[73]
DulaglutideGLP-1R agonistPhase IVNCT03590626↓ Hepatic steatosis[74]
LiraglutideGLP-1R agonistPhase IINCT01237119
NCT02654665
↓ Hepatic steatosis[75–77]
Lipogenesis inhibitors
GS-0976ACC inhibitorPhase IINCT02856555↓ Hepatic steatosis
↑ Plasma TG
[78]
MK-4074ACC inhibitorPhase IINCT01431521↓ Hepatic steatosis
↑ Plasma TG
[79]
DenifanstatFASN inhibitorPhase IINCT04906421↓ Hepatic steatosis
↓ Inflammation
↓ Liver fibrosis
[80]
AramcholSCD-1 inhibitorPhase IIINCT04104321↓ Hepatic steatosis
↓ Plasma ALT and AST
[81]
Bempedoic acidACLY inhibitorPhase IINCT06035874Recruiting[82]
BGT-002ACLY inhibitorPhase IICTR20230344Recruiting
Fibroblastgrowth factormimetics
EfruxiferminFGF21 analoguePhase IIINCT06215716
NCT06161571
NCT03976401
NCT04767529
↓ Hepatic steatosis
↓ Plasma ALT and AST
[83–85]
PegozaferminFGF21 analoguePhase IINCT03486912
NCT03486899
NCT03400163
NCT02413372
↓ Hepatic steatosis[86, 87]
Potential of emergingtherapeutic strategies
MSDC-0602KMPC inhibitorPhase IINCT02784444↓ Fasting glucose
↓ HbA1c
[88]
CenicrivirocCCR2/CCR5 inhibitorPhase IIINCT03028740↓ Inflammation
↓ Liver fibrosis
[89, 90]
SelonsertibASK1 inhibitorPhase IIINCT03053063
NCT03053050
↓ Inflammation
↓ Fibrosis
[91, 92]
Combinationtherapy
PF-05221304/PF-06865571ACC inhibitor/DGAT2 inhibitorPhase IINCT04321031
NCT03248882
NCT03776175
↓ Hepatic steatosis
(−) Plasma TG
[93]
Selonsertib/firsocostat/cilofexorASK1 inhibitor/ACC inhibitor/FXR agonistPhase IINCT02781584↓ Hepatic steatosis
(−) Plasma TG
[70]
Obeticholic acid/ atorvastatinFXR agonist/HMGCR inhibitorPhase IINCT02633956(−) LDL-C[83–85, 94]
Cilofexor/semaglutideFXR agonist/GLP-1R agonistPhase IINCT03987074↓ Hepatic steatosis[71, 86, 87]
Potential therapeutic strategies for MASH. Multiple pathways and a wide range of pathophysiological processes contribute to MASH. The three most common features of MASH are excess lipid accumulation, inflammation, and fibrosis. Modifications in lifestyle and the use of currently prescribed drugs, such as insulin resistance agents, are beneficial in the management of MASH. Novel strategies that specifically target the liver for the treatment of inflammation, fibrosis, and hepatic lipid metabolism are being investigated with promising outcomes at various stages of preclinical and clinical studies. DNL, de novo lipogenesis; FAO: fatty acid oxidation; ACLY: ATP citrate lyase; ACC: acetyl coenzyme A (acetyl-CoA) carboxylase; FASN, fatty acid synthetase; SCD-1: stearoyl-CoA desaturase-1; HMGCR: 3-hydroxy-3-methylglutaryl-CoA reductase; THRβ: thyroid hormone receptor β; PPAR: peroxisome proliferator-activated receptor; FXR: farnesoid X receptor; ASK1: apoptosis signal-regulating kinase 1; GLP-1R/GIPR: glucagon-like peptide-1 receptor/gastric inhibitory polypeptide receptor; SGLT2: sodium-glucose cotransporter-2; CCR2/5: C–C motif chemokine receptor 2/5; VLDL/LDL: very low-density lipoprotein/low-density lipoprotein.
Figure 1

Potential therapeutic strategies for MASH. Multiple pathways and a wide range of pathophysiological processes contribute to MASH. The three most common features of MASH are excess lipid accumulation, inflammation, and fibrosis. Modifications in lifestyle and the use of currently prescribed drugs, such as insulin resistance agents, are beneficial in the management of MASH. Novel strategies that specifically target the liver for the treatment of inflammation, fibrosis, and hepatic lipid metabolism are being investigated with promising outcomes at various stages of preclinical and clinical studies. DNL, de novo lipogenesis; FAO: fatty acid oxidation; ACLY: ATP citrate lyase; ACC: acetyl coenzyme A (acetyl-CoA) carboxylase; FASN, fatty acid synthetase; SCD-1: stearoyl-CoA desaturase-1; HMGCR: 3-hydroxy-3-methylglutaryl-CoA reductase; THRβ: thyroid hormone receptor β; PPAR: peroxisome proliferator-activated receptor; FXR: farnesoid X receptor; ASK1: apoptosis signal-regulating kinase 1; GLP-1R/GIPR: glucagon-like peptide-1 receptor/gastric inhibitory polypeptide receptor; SGLT2: sodium-glucose cotransporter-2; CCR2/5: C–C motif chemokine receptor 2/5; VLDL/LDL: very low-density lipoprotein/low-density lipoprotein.

Nuclear receptor agonists

THRβ agonists

Thyroid hormones (THs), particularly L-triiodothyronine (T3) and L-thyroxine (T4), serve as central regulators of numerous biological processes [95]. Their physiological effects are exerted by binding to specific nuclear receptors, known as THRα and THRβ, which are widely distributed throughout various tissues [96]. THRβ, a nuclear hormone receptor, is predominantly expressed in the liver, where it functions as an important regulator of metabolic pathways. THRβ agonists can directly modulate gene expression, thereby enhancing FA oxidation (FAO), suppressing DNL, improving insulin sensitivity, and promoting mitochondrial biogenesis [97]. Patients with hypothyroidism often present with abnormal blood lipid profiles, characterized by elevated levels of low-density lipoprotein cholesterol (LDL-C), TGs, and apolipoprotein B (ApoB) [98]. Within the euthyroid population, a decrease in TH levels increases the risk of developing MASLD. Genetic studies have also revealed that people with downregulated THRβ have an increased risk of MASH [97]. This finding validates the rationale for targeting THRβ as a promising therapeutic approach for MASLD. THs have been demonstrated to be effective at reducing liver fat content (LFC) and reversing MASLD in both rodents and humans. However, nonselective activation of THRα may lead to adverse events such as chest discomfort in some patients [99].

Currently, most THRβ agonists are modified and engineered based on the chemical molecular framework of THs. Sobetirome (GC-1), a synthetic analog of TH that is relatively selective for both the binding and activation functions of THRβ over THRα, regulates the expression of high-density lipoprotein (HDL) and LDL receptors (LDLRs) to reduce cholesterol and TG levels [100, 101]. In MASH mouse models, GC-1 has been shown to decrease fat accumulation in the liver. However, due to preclinical adverse events, the development of GC-1 was discontinued [102]. Eprotirome (KB2115) shows high selectivity for THRβ, effectively lowers serum cholesterol and TGs, and shows no cardiotoxicity [103, 104]. However, possible cartilage damage in dogs and hepatotoxicity in humans have hindered its further development [105].

To overcome the effects of THRβ agonists on extrahepatic tissues, “prodrugs” that specifically target the liver have been developed. VK2809 (MB07811) requires oxidation by the cytochrome P450 isoenzyme CYP3A (cytochrome P4503A) to form the active ingredient. In animal models of hypercholesterolemia, VK2809 significantly reduces LDL-C and TG levels with good tolerability [60]. Subsequently, a phase IIb clinical trial for the treatment of MASH was recently initiated (NCT04173065). ASC41 is metabolized by CYP3A4 in the liver and forms an active compound with increased THRβ affinity. Improvements in LDL-C and TG have been demonstrated in a phase I clinical trial, and a phase II clinical trial is currently recruiting patients (NCT05462353). Another prodrug is TERN-501, which can reduce serum cholesterol levels and ameliorate liver steatosis and fibrosis in rodents with hyperlipidemia and MASH. The results from a phase II clinical trial showed that the LFC of MASH patients treated with TERN-501 decreased by 45% at 12 weeks, and both the primary and secondary endpoints were met (NCT05415722).

Resmetirom (MGL-3196), a selective THRβ agonist, effectively targets the liver by exploiting the organic anion-transporting polypeptide 1B1 receptor expressed on hepatocytes [106]. In phase II clinical trial that included 36 weeks of consecutive liver biopsies, patients treated with resmetirom demonstrated a reduction in liver fat over 12 weeks of treatment. Furthermore, exploratory endpoints such as improvements in liver enzymes and tissue fibrosis showed favorable results, with no adverse events related to THRα activity [107]. Resmetirom is currently being tested in four phase III clinical trials, the primary objective of which is to evaluate its therapeutic effects over a period of 52 weeks in biopsy-confirmed patients with MASH. These studies have assessed safety and tolerability using liver biopsies (NCT04197479 and NCT03900429) [35, 59]. During 52 weeks, 966 patients with mild-to-moderate MASH (up to stage F3) received oral treatment with either 80 mg or 100 mg of resmetirom or placebo once daily in a randomized, controlled phase III trial. Compared with 10% of placebo recipients, 25%−30% of resmetirom recipients demonstrated MASH regression without worsening fibrosis, as determined by histologic analysis of biopsy samples. Twenty-five percent of the resmetirom recipients experienced at least one stage of improvement in fibrosis, while the placebo group experienced almost 15% less improvement in the 8-point NAFLD activity score (NAS). The FDA recently approved resmetirom for the treatment of MASH.

Peroxisome proliferator-activated receptor (PPAR) agonists

The PPAR family consists of three isotypes: PPARα (encoded by NR1C1), PPARδ (synonymous with β, encoded by NR1C2), and PPARγ (encoded by NR1C3) [108]. Upon ligand binding to the ligand-binding domain of the PPAR protein, PPAR forms a heterodimer with the retinoid X receptor (RXR). The complex binds to specific peroxisome proliferator response elements (PPREs) on DNA, resulting in the transcription of downstream target genes [109]. PPARα and PPARβ/δ-controlled gene expression is associated with mitochondrial and peroxisomal β-oxidative catabolism and ketogenesis [110]. PPARδ regulates the expression of thermogenic genes in adipose tissue and genes involved in FAO in skeletal muscle, white adipose tissue, and the liver. Furthermore, PPARγ plays a crucial role in the regulation of HSCs [111, 112]. However, weight gain is the most important side effect of treatment with PPARγ agonists, e.g., rosiglitazone, due to the increase in fat content in adipose tissue [113].

Multiple PPAR regulators have been identified and used as treatments for MASH [112, 114–116]. Pemafibrate, a novel selective PPARα modulator, has also been used to treat hyperlipidemia [66]. When combined with tofogliflozin, a highly selective sodium-glucose cotransporter-2 (SGLT2) inhibitor, pemafibrate treatment significantly prevents MASH development in rodents [117]. Pemafibrate administration leads to improvements in markers of liver inflammation and fibrosis. Remarkably, patients with lean MASLD respond better to pemafibrate therapy than patients with obese MASLD [67]. The PPARγ agonist pioglitazone reduces hepatic steatosis and inflammation, but only slightly improves fibrosis [64]. It appears that dual or pan-PPAR agonists have better clinical efficacy than single agonists. Oral administration of the dual PPARα/δ agonist elafibranor (also known as GFT-505) reduces steatotic liver inflammation and fibrosis in a mouse model, but its antifibrotic effects were limited in phase II and phase III studies (NCT01694849 and NCT02704403) [62, 118]. Treatment with the PPARα/δ/γ pan-agonist lanifibranor significantly reduces liver steatosis and inflammation in a rodent model of MASH [112, 119]. In a recent phase IIb trial (NCT03008070), lanifibranor treatment significantly resolved MASH without worsening fibrosis [61]. A phase III study evaluating the efficacy and safety of lanifibranor in adult patients with MASH is currently recruiting new patients (NCT04849728). Generally, dual- or multiple-target PPAR agonists could provide greater benefits in the treatment of multiorgan metabolic disorders in MASH. However, balancing the activation effects between the isoforms remains a challenge in the development of PPAR agonists. Based on encouraging results, numerous PPAR agonists have been developed for potential MASH treatment to overcome side effects or increase the efficacy of PPAR activity [120, 121].

Farnesoid X receptor (FXR) agonists

Although FXRs were originally found to be nuclear receptors activated by farnesol derivatives [122], subsequent studies showed that FXR is the primary sensor for bile acid (BA) [123, 124]. FXR is predominantly expressed in the liver and intestines, with lower levels in the kidneys, adipose tissue, and adrenal glands [122]. FXR forms a heterodimer with RXR to modulate the expression of target genes [125]. Clinical studies have shown that MASLD is associated with disruptions in BA homeostasis and its related signaling pathways [126]. FXR regulates the transcription of genes involved in the synthesis, absorption, uptake, and transport of BA to maintain BA homeostasis, and in glucose and lipid metabolism [127, 128]. Activation of hepatic FXR increases the expression of small heterodimer partner (SHP), and the FXR-SHP axis inhibits the expression of SREBP and important lipogenic genes [129, 130]. Moreover, FXR activation induces the expression of PPARα [131] and its target genes, as well as the expression and secretion of hepatic fibroblast growth factor 21 (FGF21) [132], both of which are involved in FA transport and oxidation. In addition, FXR controls genes involved in lipoprotein metabolism, including very low-density lipoprotein receptor (VLDLR), scavenger receptor B type 1 (SR-B1), syndecan-1, ApoCII/III, and fatty acid translocase (FAT/CD36) [125]. In summary, FXR activation reduces liver steatosis by suppressing DNL and promoting FFA oxidation. FXR activation can be anti-inflammatory and reduce liver fibrosis [133–135]. Additionally, activation of FXR has been shown to reduce hepatic gluconeogenesis, suppress glycolysis, induce glycogen synthesis, and modulate insulin signaling [127, 136–138].

Consistent with the critical role of FXR in regulating lipid and glucose metabolism in addition to BA synthesis, several FXR activators of endogenous and synthetic ligands have shown potential for clinical applications. Based on their chemical structure, FXR agonists are classified as either steroidal or nonsteroidal. Obeticholic acid (OCA or 6α-ethyl chenodeoxycholic acid; originally known as INT-747) is a semisynthetic steroidal BA derivative that acts as an FXR agonist [139]. OCA is the first FXR agonist approved by the FDA and the first drug considered to be an investigational drug for MASH. A phase III trial (NCT02548351) on OCA is fully enrolled and includes 2477 randomized MASH patients with precirrhotic liver fibrosis, including nearly 1000 who had been taking the study drug for at least four years. In two preliminary 18-month analyses, 25 mg OCA consistently demonstrated a response rate twice as high as that of placebo in reducing liver fibrosis stage, an endpoint consistent with the FDA’s draft guidance. However, the use of OCA has been reported to be associated with side effects such as pruritus and dyslipidemia [68, 69].

Most steroidal BA-like candidates exhibit adverse effects, and thus researchers have focused on developing new selective nonsteroidal FXR agonists. GW4064 was first reported as a non-BA FXR agonist and has been widely used by scientists as a tool to explore the mechanism of action and pharmacology of FXR [140]. Cilofexor/GS-9674, TREN-101, nidufexor, and tropifexor, which were developed based on GW4064, are currently in clinical trials for the treatment of MASH [72, 141, 142]. Phase II trials of HPG1860 (NCT05338034), TERN-101 (NCT04328077), MET409 (NCT04702490), MET642 (NCT04773964), and other nonsteroidal FXR agonists have demonstrated favorable safety profiles and good tolerance with no serious treatment-related adverse events. These compounds significantly reduce liver steatosis and serum BA levels, and improve liver biochemistry in patients with MASH.

FXR agonists have the potential to improve both MASH histology and pathological features of liver fibrosis, as well as excess lipid accumulation, and thus FXR is one of the most promising targets in the field of MASH. However, the side effects of FXR agonists, including hypercholesterolemia and pruritus, still need to be resolved. Exploring novel FXR agonists or multitarget modulators with minimal side effects, along with lower FXR agonist doses in combination with existing or emerging drugs targeting different mechanisms and tailored patient treatments, could reveal the promising future of FXR agonists in MASH therapy.

Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists

GLP-1R agonists are incretin-based hypoglycemic agents with therapeutic effects that include appetite reduction, weight loss, and delayed gastric emptying [143, 144]. These agonists also exert hepatoprotective effects through mechanisms such as increasing insulin sensitivity, reducing lipid accumulation, improving liver mitochondrial function, and inhibiting the stress response of the damaged ER. Interestingly, GLP-1 secretion is impaired in MASH patients, which suggests that GLP-1R is an attractive target for the treatment of MASH [145]. Several available GLP-1R agonists (e.g., liraglutide, dulaglutide, and semaglutide) have been shown to clinically alleviate the symptoms of MASH.

Liraglutide is the first once-daily human GLP-1 agonist approved for the treatment of patients with type 2 diabetes mellitus (T2DM) [146]. In the clinical trial NCT01237119, liver biopsy demonstrated resolution of definitive MASH in 39% of patients who received liraglutide for 48 weeks [75]. A Japanese study of MASH patients with glucose intolerance showed that liraglutide along with diet and exercise interventions for six months improved blood biochemistry characteristics of the disease [76]. Additionally, a clinical trial (NCT02654665) revealed that liraglutide was equally effective as a combined diet and exercise program in reducing weight, LFC, and liver damage within six months [77]. Dulaglutide, another long-acting GLP-1R agonist, has been shown to significantly reduce LFC and γ-glutamyl transpeptidase levels in patients with T2DM and MASLD [74]. Another clinical trial is currently evaluating the efficacy of dulaglutide in MASH and diabetic populations randomized to receive either 1.5 mg of dulaglutide in addition to dietary supplementation or dietary supplementation alone for 52 weeks (NCT03648554). Its primary outcome measures include improved histological characterization of MASH by liver biopsy. Semaglutide is another approved antidiabetic GLP-1R agonist and has been demonstrated to lead to the highest percentage of weight loss of any obesity drug to date [147]. In a 72-week double-blind phase II trial (NCT02970942) in patients with biopsy-confirmed MASH and fibrosis, the percentage of resolution of MASH in the semaglutide group was significantly greater than that in the placebo group [73]. A large-scale phase III trial by Novo Nordisk to evaluate the efficacy of weekly subcutaneous injections of semaglutide in patients with noncirrhotic MASH is currently underway (NCT04822181). Its primary endpoints include the resolution of steatohepatitis, improvement in liver fibrosis, and time to first liver-related clinical event.

In addition, emerging evidence suggests that dual GLP-1 and gastric inhibitory polypeptide (GIP) receptor (GIPR) agonists (e.g., tirzepatide), dual GLP-1 and glucagon receptor agonists (e.g., cotadutide), or triple GLP-1/GIP/glucagon receptor agonists (e.g., HM15211) may be more promising than each therapy alone for alleviating MASH and liver fibrosis, but further testing is needed [148]. A 26-week treatment with once-weekly tirzepatide (10 or 15 mg) significantly improved MASH-related biomarkers in patients with T2DM (NCT03131687) [149]. An ongoing phase II trial (NCT05364931) of cotadutide is currently evaluating its safety and efficacy in patients with noncirrhotic MASH with fibrosis. Moreover, another phase II trial (NCT04505436) is currently evaluating the efficacy, safety, and tolerability of HM15211 over 12 months in individuals with biopsy-diagnosed MASH.

Lipogenesis inhibitors

Excess hepatic DNL leads to TG accumulation and lipotoxicity, leading to hepatic steatosis [26]. The process of FA biosynthesis includes the following steps: glycolysis, lipogenesis, desaturation, elongation, and esterification. The key enzymes involved in the biosynthesis of FAs from carbohydrate substrates include ATP citrate synthase (ACLY), ACC, and FASN [150]. ACLY catalyzes the conversion of cytosolic citrate to acetyl-CoA, which is derived from the tricarboxylic acid (TCA) cycle. ACC induces the carboxylation of acetyl-CoA to malonyl-CoA, and then FASN converts malonyl-CoA to long-chain FAs (LCFAs). The elongation and desaturation of FAs require the elongation of the very long-chain FA protein (ELOVL) and SCD, respectively. SCD catalyzes the synthesis of monounsaturated FAs (MUFAs). In the esterification step, TGs are synthesized by enzymes including monoacylglycerol acyltransferase (MGAT) and diacylglycerol acyltransferase (DGAT). Drugs targeting ACC, FASN, SCD-1, and DGAT2 are currently being advanced into clinical trials for MASH treatment. Recently, ACLY inhibitors and MGAT2 inhibitors have been shown in preclinical studies to have potential for MASH treatment.

ACC inhibitors

ACC, a pivotal enzyme in FA metabolism, has attracted considerable attention as a promising therapeutic target for MASH. In mammals, ACC has two isoforms, ACC1 and ACC2. ACC1 is a cytoplasmic protein that catalyzes the conversion of acetyl-CoA to malonyl-CoA, which is further converted into LCFAs by FASN. ACC2 is a mitochondrial protein that regulates FA β-oxidation. Thus, ACC inhibition simultaneously inhibits hepatic DNL and increases FAO [151, 152]. A recent study also showed that ACC inhibition reduces HSC activation. Therefore, ACC inhibition can directly improve liver steatosis, inflammation, and fibrosis [153].

Available ACC enzyme inhibitors, including GS-0976 from Gilead [154], MK-4074 from Merck [79, 155], and PF-05221304 [156], significantly block hepatic lipogenesis in rodents. Furthermore, a novel arachidonic acid 12-lipoxygenase (ALOX12)-ACC1 targeting strategy (a small-molecule intermetatarsal angle 1 (IMA-1)) promotes ACC1 protein degradation and modestly regulates ACC1 activity [157, 158]. Enzyme inhibitors, but not the ACC1 degrader, significantly increase circulating TGs. The use of GS-0976 in the treatment of MASH is in various clinical trial stages. In the clinical trial NCT02876796, GS-0976 reduced hepatic DNL in overweight and obese subjects. In a randomized, double-blind, placebo-controlled phase II trial (NCT02856555), patients with hepatic steatosis (LFC > 8%) were assigned at a 2:2:1 ratio to receive a once-daily oral dosage of 20 mg of GS-0976, 5 mg of GS-0976, or placebo for 12 weeks. The results showed that GS-0976 was safe and well tolerated. Magnetic resonance imaging-based proton density fat fraction (MRI-PDFF) estimated a ≥30% decrease from baseline in 48% of patients who received the high dose and in 23% of patients who received the low dose, both of which differed significantly from the placebo (15%). However, this clinical trial also showed that patients treated with GS-0976 had an increase in plasma TGs [78]. The phase II clinical trial NCT03248882, which tested PF-05221304 (2 mg, 10 mg, 25 mg, or 50 mg), demonstrated a dose-dependent reduction in the LFC from the baseline [93]. The levels of the MASH-related biomarkers CK18-M30 and CK18-M65 were also reduced after the administration of PF-05221304. However, monotherapy with PF-05221304 also increased the serum TG level in a dose-dependent manner.

FASN inhibitors

FASN converts acetyl-CoA and malonyl-CoA from simple dietary sugars into palmitate, a saturated FA with 16-carbon atoms. Elevated hepatic palmitate production is deemed a significant initiator of MASH progression [159], thus indicating the potential use of FASN inhibitors as therapeutic agents for MASH [160, 161]. Denifanstat (TVB-2640) is a highly potent selective FASN inhibitor developed by Sagimet Biosciences [161]. In a preclinical study, denifanstat was shown to improve MASH when used as monotherapy or in combination with the GLP-1 agonist semaglutide. In the phase IIa FASCINATE-1 trial (NCT03938246), denifanstat significantly reduced LFC and improved biochemical, inflammatory, and fibrotic biomarkers of MASH after 12 weeks in a dose-dependent manner [80]. In a phase IIb clinical trial (NCT04906421), denifanstat achieved primary and multiple secondary endpoints after 52 weeks of treatment. Administration of denifanstat (50 mg) resolved MASH with a ≥2-point reduction in the NAS (36% in the denifanstat group versus 13% in the placebo group) and improved the ≥2-point reduction in the NAS (52% in the denifanstat group versus 20% in the placebo group). Denifanstat administration improved fibrosis by ≥1 stage without worsening MASH (41% of patients in the denifanstat group versus 18% of patients in the placebo group) (NCT04906421). Recently, Sagimet Biosciences announced plans to conduct a pivotal phase III program to test denifanstat in MASH patients.

SCD-1 inhibitors

SCD-1 catalyzes the conversion of saturated FAs into MUFAs. MUFAs serve as substrates for the synthesis of various types of lipids (such as TGs and phospholipids). SCD-1 inhibition reduces TG synthesis [162, 163], and thus SCD-1 inhibitors have been reported to be promising therapeutic agents for the treatment of MASH [164]. Aramchol (icomidocholic acid) is a liver-targeted SCD-1 inhibitor. Aramchol treatment reduced MASH and fibrosis in a mouse model of diet-induced MASH [165, 166]. Administration of aramchol resolved MASH in a phase IIb trial (NCT02279524) (16.7% in the aramchol group versus 5% in the placebo group). In a phase IIb study, fibrosis was reduced without worsening MASH (29.5% in the aramchol group versus 17.5% in the placebo group) [81]. Recently, a phase III trial revealed that 39% of patients showed improvement in fibrosis after at least 48 weeks of aramchol administration, as determined by the MASH Clinical Research Network scoring system (NCT04104321).

3-Hydroxy-3-methylglutaryl-CoA reductase (HMGCR) inhibitors

The elevation of free cholesterol in the liver serves as a distinct marker that differentiates MASH from non-MASH pathology [30]. Given the high occurrence of hypercholesterolemia and cardiovascular disease (CVD), the use of statins, which inhibit HMGCR, has been investigated in patients with MASLD/MASH [167–170]. Moreover, statin treatment halves CVD morbidity and mortality in statin-treated MASLD/MASH patients [171]. However, the results from these studies are difficult to interpret because of the limited patient numbers and short duration of follow-up.

ACLY and MGAT2 are possible targets in MASH

ACLY is an enzyme responsible for the formation of acetyl-CoA from citrate in FA and cholesterol synthesis [172]. In hepatocytes, ACLY inhibition reduces lipogenesis, promotes β-oxidation of FAs, and causes cholesterol efflux [173, 174]. Bempedoic acid (ETC-1002) is the first FDA-approved ACLY inhibitor for the treatment of heterozygous familial hypercholesterolemia [175–177]. ACLY silencing or ACLY inhibition alleviates diet-induced MASH and CCl4-induced fibrosis in rodents [178, 179]. The effectiveness of bempedoic acid administration for 24 weeks in lowering liver cholesterol in MASLD patients with T2DM is being investigated (NCT06035874). A randomized, double-blind, multiple-dosing, placebo-controlled, phase Ib/IIa study of the novel ACLY inhibitor BGT-002 in MASLD subjects is ongoing (CTR20230344).

MGAT2 catalyzes the production of DG from monoacylglycerol. This important enzyme is highly expressed in human small intestine and liver, and is responsible for the absorption of dietary fat and the synthesis and distribution of TGs [180]. Hepatic MGAT2 expression is increased in MASLD patients, and knockout of Mgat2 reduces fat absorption and increases the secretion of GLP-1 and peptide YY (PYY), which are gut hormones that have beneficial effects on blood glucose and appetite remodeling [181, 182]. A recent study showed that the administration of MGAT2 inhibitors reduces liver fibrosis and inflammation in murine models of MASH and reduces body weight in obese adults [183].

FGF mimetics

FGF21 analogs

FGF21 is an atypical member of the FGF superfamily [184]. FGF21 is a secreted hepatokine that circulates to its target tissues (e.g., the liver, adipose tissue, and brain) [185]. FGF21 plays an important role in the regulation of energy balance as well as glucose and lipid homeostasis [186]. Clinical evidence has indicated that FGF21 serum concentrations are elevated in MASLD patients and are positively correlated with intrahepatic TG levels, which suggests that FGF21 may be a biomarker for MASLD and may serve as a key regulator of lipid metabolism in the liver [187–189]. FGF21 has been reported to reduce intrahepatic lipid accumulation by reducing liver DNL and increasing mitochondrial β-oxidation [190].

Furthermore, FGF21 can reduce liver fibrosis, inflammation, and damage by attenuating “multi-hits”, such as oxidative stress, ER stress, and chronic inflammation, in MASLD pathogenesis [191–193]. To overcome the short half-life and poor pharmacokinetics of native FGF21, long-acting FGF21 analogs have been developed and are in various phases of preclinical and clinical research.

Efruxifermin is a differentiated Fc-FGF21 fusion protein designed to mimic the balanced biological activity of native FGF21. This FGF21 analog exhibits balanced receptor binding affinity for the metabolically relevant FGF receptor (FGFR) 1c/2c and 3c [194]. A phase IIa study (NCT03976401) showed that efruxifermin significantly reduces liver fat in patients with MASH and fibrosis stages 1−3 while maintaining a tolerable safety profile [83, 84]. Patients with MASH showed significant regression of fibrosis and resolution of steatohepatitis after 24 weeks of treatment with efruxifermin [85]. In a phase IIb study (NCT05039450), efruxifermin significantly improved the markers of liver injury and fibrosis, and insulin sensitization. An ongoing phase III trial (NCT06215716) is currently evaluating the efficacy of efruxifermin in resolving fibrosis and improving steatohepatitis in patients with precirrhotic MASH, and the SYNCHRONY Real-World trial (NCT06161571) is evaluating the safety and tolerability of efruxifermin in patients with MASH or MASLD. As the first FGF21 analog to enter a phase III trial, efruxifermin, with anti-fibrosis properties, has promising potential for the treatment of MASH.

Pegozafermin, a glycoPEGylated FGF21, is another notable FGF21 analog [86]. Over 24 weeks of treatment demonstrated significant and clinically meaningful improvements in LFC, noninvasive markers of liver fibrosis and inflammation, and lipid markers in MASH patients with stage F2−F3 fibrosis in the ENLIVEN Phase IIb trial (NCT04929483) [87]. In addition, patients with compensated cirrhosis also showed improvements in fibrosis (F4). The data collected at 48 weeks of treatment with pegozafermin showed sustained improvements in liver damage, inflammation, and fibrosis. Several additional phase III trials to further assess the safety and efficacy of pegozafermin in patients with MASH are scheduled for 2024.

FGF19 analogs

Apart from FGF21, FGF19 also appears to have beneficial effects on MASH [195]. FGF19 is a hormone that controls the synthesis of BA from cholesterol via CYP7A1. A recent study revealed that FGF19-induced FGFR activation reduces insulin-induced lipogenesis in the liver. Physiologically, FXR activation triggers the secretion of FGF19 in the ileum, which suggests that the induction of FGF19, the target gene of FXR, may be an essential factor in the action of FXR agonists. However, studies on its hepatocarcinogenic effects are limited [196, 197]. Recently, a nontumorigenic variant of FGF19, aldafermin (also called NGM282) [198], was studied in MASH patients in a placebo-controlled phase II trial. Aldafermin was found to be well tolerated after 12 weeks of treatment and >70% of patients in the aldafermin group reduced their absolute LFC by at least 5% from baseline, while only 7% of patients in the placebo group achieved the same reduction (NCT02443116) [199, 200]. However, a more recent clinical study revealed that the administration of aldafermin did not significantly improve fibrosis in at least one stage without worsening MASH (NCT03912532) [201].

Potential of emerging therapeutic strategies

Mitochondrial modulators

Mitochondrial dysfunction is closely associated with the progression of simple steatosis to MASH [202]. Targeting mitochondrial dysfunction with mitochondrial pyruvate carrier (MPC) inhibitors and mitochondrial uncouplers is a potential treatment for MASH. In rodents and nonhuman primates, the administration of mitochondrial uncouplers reverses MASLD and MASH [203]. In obese individuals, 2,4-dinitrophenol (DNP), the most well-known mitochondrial uncoupler, is used as a weight-loss drug [204]. To combat deleterious side effects, such as agranulocytosis, hyperthermia, cataracts, and death, several attempts have been made to develop DNP-based pharmacological agents, including DNP-methyl ether (DNPME), controlled-release mitochondrial protonophore (CRMP), DNP loaded LC-gel (DNP-LC gel, an injectable liquid crystal gel), and Compound (6j) [205–208]. These studies suggest that a promising therapeutic approach to improve fatty liver disease is to slightly increase uncoupling and cellular energy expenditure. In individuals with MASLD, MPC may play an important role in the intrahepatic lipid pool, despite minimal flux through this pathway [209]. Pharmacological inhibition of MPC activity improved metabolic parameters, and reduced liver injury and fibrosis [210, 211] [212]. However, in a placebo-controlled phase IIb trial, MPC inhibition by treatment with MSDC-0602K did not significantly improve MASH (NCT02784444) [88].

Galectin-3 (Gal-3) inhibitors

Gal-3 is a β-galactoside binding protein associated with many disease processes, including chronic inflammation and fibrogenesis. Gal-3 is highly profibrotic and modulates the activity of fibroblasts and macrophages in chronically inflamed organs [213, 214]. Consequently, Gal-3 inhibitors are being developed as potential treatments for MASH. GR-MD-02 (belapectin), a proposed Gal-3 inhibitor, has been reported to improve histopathological changes in MASH fibrosis in MASH models [215, 216]. However, in a phase II trial in MASH patients with portal hypertension (NCT02462967), belapectin failed to reduce the hepatic venous pressure gradient [217, 218]. Subanalysis revealed that belapectin increased the hepatic venous pressure gradient and prevented the development of esophageal varices in patients without varices at baseline endoscopy. The effectiveness of belapectin in preventing the development of esophageal varices is currently being investigated in a phase IIb/III trial (NCT04365868).

CCR2/CCR5 inhibitors

HSC activation and macrophage-mediated inflammation are key events in the development of MASH. In contrast to tissue-resident and self-renewing KCs in the liver, proinflammatory monocyte-derived macrophages (MoMFs) are derived from circulating monocytes that invade the liver upon injury [219, 220]. These cells exhibit CCR2-dependent metabolic, tolerogenic, and homeostatic properties. These MoMFs directly stimulate HSC activation and promote angiogenesis and inflammation. Both HSC and lymphocyte subsets express CCR5, which controls migration and proliferation. CCR2/5 inhibitors, which target disease-promoting liver macrophages and HSCs, have demonstrated efficacy in reducing fibrosis in animal models of MASH and fibrosis, which has led to their advancement to clinical trials. Cenicriviroc, (CVC, also TBR-652 or TAK-652), is a new and potent CCR2/5 receptor antagonist that is taken orally and has shown antifibrotic effects in animal models [221]. CVC significantly improved liver fibrosis compared with placebo in adults with MASH and stage F2 or F3 liver fibrosis in a phase IIb trial (NCT02217475) [89]. A multicenter, randomized, double-blind, placebo-controlled phase III trial (NCT03028740) evaluated the safety and efficacy of CVC. However, the primary efficacy endpoint of determining whether CVC is at least one level more effective than placebo in improving liver fibrosis and preventing the exacerbation of steatohepatitis was not met and, as a result, the study sponsor decided to discontinue the development of CVC [90].

Apoptosis signal-regulating kinase 1 (ASK1) inhibitors

ASK1, a member of the mitogen-activated protein kinase (MAPK) kinase kinase (MAP3K) family, controls the p38 MAPK and JNK signaling pathways. In response to a variety of cellular stressors, such as ROS, ER stress, and inflammatory signals, ASK1 is activated through homodimerization and subsequent autophosphorylation. The JNK and p38 MAPK signaling pathways in liver cells can then be activated by phosphorylated ASK1, which leads to apoptosis, the release of inflammatory cytokines, and the induction of fibrogenic genes [222]. Based on these recognized roles of ASK1, ASK1 inhibitors have been developed within the last decade. Selonsertib, also known as GS-4997, is an ATP-competitive ASK1 inhibitor that is currently being evaluated in the treatment of MASH. However, the findings of two phase III trials (NCT03053063 and NCT03053050) of selonsertib were negative [91, 92].

Combination therapy

Given the complexity of MASH development, combination therapy directed at multiple targets has also attracted interest, as it is expected to yield more benefits and fewer side effects than monotherapy. Theoretically, combinations of drugs with different mechanisms of action against liver steatosis, inflammation, and fibrosis should be considered [223]. A combination of medications can reduce side effects in two ways. Firstly, the combination may allow the use of lower doses to increase tolerability. Secondly, the combination can alleviate the side effects of the first drug. Such combinations should be safe and provide beneficial effects beyond the liver, such as weight loss, insulin sensitivity, and cardiovascular protection.

Combination of ACC inhibitors and DGAT2 inhibitors

Current ACC inhibitors, which have already been tested in clinical trials, also lead to a dose-dependent increase in serum TG concentration in patients. Moreover, hypertriglyceridemia significantly increases the risk of CVD in MASH patients. This complication represents a major obstacle to the clinical use of ACC inhibitors for MASH treatment. ACC inhibition results in hypertriglyceridemia, probably due to suppression of polyunsaturated FA (PUFA) synthesis, which in turn results in increased expression of SREBP-1c and glycerol-3-phosphate acyltransferase 1 (GPAT1) and increased very low-density lipoprotein cholesterol (VLDL-C) in the blood [79]. One way to overcome this deficiency is to simultaneously block TG synthesis. DGAT2 is an enzyme that catalyzes the final step in the generation of TGs. Coadministration of the DGAT2 inhibitor PF-06865571 with the ACC inhibitor PF-05221304 reduced LFC and largely eliminated hypertriglyceridemia in a 6-week clinical trial follow-up period (NCT03776175 and NCT04321031) [93]. This trial demonstrated that the coadministration of ACC inhibitors with DGAT2 inhibitors can exert anti-MASH effects while resolving the hypertriglyceridemia associated with ACC inhibition. In addition, hypertriglyceridemia caused by ACC inhibition monotherapy can be reversed by combination treatment with fenofibrate, a PPARα agonist. This combination prevents an increase in plasma TGs and improves LFC and liver biochemistry (NCT02781584) [224].

FXR agonists and combination treatments

The most common side effects of FXR agonists in MASH patients are pruritus, increased plasma LDL-C level, and decreased HDL-C level, which may increase the risk of cardiovascular events [225]. Several FXR agonists, including tropifexor, cilofexor, and OCA, are currently being investigated as combination therapies. OCA has been tested in combination with atorvastatin for the treatment of LDL-C elevation. Administration of OCA for four weeks increased plasma LDL-C level, which subsequently decreased when patients also received atorvastatin (NCT02633956) [94]. Cilofexor was tested in combination with firsocostat (an ACC inhibitor) and selonsertib (an ASK1 inhibitor) in the NCT03449446 and NCT02781584 trials [70]. Cilofexor has also been used in combination with the GLP-1 agonist semaglutide and the ACC inhibitor firsocostat. The combination treatment improved liver enzymes, fibrosis, and the NAS on histology and improved liver elastography (NCT03987074) [71].

Combination with anti-diabetic drugs

T2DM is an independent risk factor for MASLD progression [226, 227]. Combining anti-diabetic drugs with anti-MASH drugs may help to improve both diabetes-related and liver-related outcomes. The combination of vitamin E, a recommended anti-MASH drug, with pioglitazone, an anti-diabetic drug, resulted in a great response in MASH with T2DM (NCT00063622) [65]. In addition, GLP-1 agonists or SGLT2 inhibitors, which are anti-diabetic drugs intended to be used exclusively for MASH treatment, have been used in combination with anti-MASH drugs. For example, tropifexor is being investigated in combination with the SGLT1/2 inhibitor licogliflozin (NCT03205150) [228]. Cilofexor has been studied in combination with the GLP-1 agonist semaglutide (NCT03987074) [71].

Summary and perspective

NASH, a term introduced in the 1980s to describe the excessive accumulation of liver lipids in individuals without heavy alcohol consumption, triggers inflammation and cell death [229]. However, advances in pathology have shown that NASH is not only a fatty liver disease but also a metabolic disorder that is closely linked to features of metabolic syndrome such as obesity and T2DM. Therefore, after extensive discussion, the term MASLD/MASH has emerged as a replacement for NAFLD/NASH and includes patients with hepatic steatosis and the presence of at least one of five cardiometabolic risk factors (hypertension, hyperglycemia, overweight/obesity, hypertriglyceridemia, and HDL hypocholesterolemia) [230–232]. In patients with MASLD, regardless of the extent of fibrosis, primary care should focus on weight loss and treatment of cardiometabolic risk factors. This review highlights the complex pathogenesis of the MASH process and summarizes current treatments and potential therapeutic approaches. Regulators of lipid metabolism, e.g., lipogenesis inhibitors and inducers of FAO, immunomodulators, and antifibrotic candidates, as well as their combinations, have been found to be effective.

Clincal medications for MASLD/MASH management include pioglitazone, a PPARγ agonist, and vitamin E. Recently, resmetirom, a selective THRβ agonist, has received unprecedented approval for MASH treatment. Numerous promising therapeutic avenues for MASH suggest the possibility of successful treatment in the future. In recent years, pharmaceutical companies and academic institutions have devoted significant efforts to exploring lipid metabolism, inflammation, and fibrotic signaling pathways to identify novel MASH drugs. As our understanding of the biological mechanisms underlying MASH progression has deepened, we have witnessed both successful and unsuccessful therapeutic strategies, and the development of safe and effective MASH strategies has continued to advance. Despite the clinical failure of several promising drugs, such as the PPARα/δ agonist elafibranor, the FXR agonist OCA, and the ACC inhibitor GS-0976, the search for safer and more effective treatment approaches based on clinical evidence remains steadfast. For example, the pan-PPAR agonist lanifibranor balances the excitatory effects of PPARα/δ/γ and significantly improves fibrosis. Nonsteroidal FXR agonists exhibit promising therapeutic potential with fewer toxic side effects. Additionally, hyperlipidemia induced by ACC inhibitors can be mitigated, to a certain extent, through combination therapies.

The coordinated regulation of FA and cholesterol metabolism, coupled with the reprogramming of mitochondrial function in the liver, such as through THRβ agonists, could represent crucial avenues for treating MASH. The use of multitarget drugs, e.g., biguanide analogs, or drugs with extrahepatic functions (e.g., insulin resistance, adipose tissue dysfunction, and weight loss), has promoted the development of comprehensive MASH treatment. New indications for antidiabetic drugs, e.g., GLP-1R agonists and SGLT2 inhibitors, for MASH warrant further investigation. As MASH embodies extremely complex and heterogeneous mechanisms, personalized combination therapy approaches and clinical combination treatment may offer a greater chance of treating MASH as early as possible. In addition, increasingly sophisticated diagnostic techniques such as histological measurements, and the artificial intelligence (AI)-assisted drug discovery will accelerate the discovery of novel treatments for MASH.

In summary, some challenges also exist even though the FDA recently approved the first drug, and additional studies are needed to better understand the pathogenic mechanisms linking liver metabolism, response to inflammation, and injury in MASLD/MASH development.

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (92057116 to J.L.; 82170872 to Z.X.; and 82200965 to Y.L.); the Natural Science Foundation of Shanghai’s 2021 “Science and Technology Innovation Action Plan” (21ZR1475300 to Z.X.); Shanghai Science and Technology Development Funds (22YF1456900 to Y.L.).

Author contributions

Zhifu Xie (Funding acquisition [Equal], Writing—original draft [Equal], Writing—review & editing [Equal]), Yufeng Li (Funding acquisition [Equal], Writing—original draft [Equal], Writing—review & editing [Equal]), Long Cheng (Resources [Supporting], Writing—original draft [Supporting]), Yidan Huang (Resources [Supporting], Writing—original draft [Supporting]), Wanglin Rao (Resources [Supporting], Writing—original draft [Supporting]), Honglu Shi (Resources [Supporting], Writing—original draft [Supporting]), and Jingya Li (Funding acquisition [Equal], Resources [Lead], Supervision [Lead], Writing—review & editing [Lead]).

Conflict of interest

The authors declare that no conflict of interest exists.

References

1.

Loomba
R
,
Friedman
SL
,
Shulman
GI.
Mechanisms and disease consequences of nonalcoholic fatty liver disease
.
Cell
2021
;
184
:
2537
64
.

2.

Diehl
AM
,
Day
C.
Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis
.
N Engl J Med
2017
;
377
:
2063
72
.

3.

Powell
EE
,
Wong
VW
,
Rinella
M.
Non-alcoholic fatty liver disease
.
Lancet
2021
;
397
:
2212
24
.

4.

Riazi
K
,
Azhari
H
,
Charette
JH
et al. .
The prevalence and incidence of NAFLD worldwide: a systematic review and meta-analysis
.
Lancet Gastroenterol Hepatol
2022
;
7
:
851
61
.

5.

Estes
C
,
Anstee
QM
,
Arias-Loste
MT
et al. .
Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016−2030
.
J Hepatol
2018
;
69
:
896
904
.

6.

Mitra
S
,
De
A
,
Chowdhury
A.
Epidemiology of non-alcoholic and alcoholic fatty liver diseases
.
Transl Gastroenterol Hepatol
2020
;
5
:
16
.

7.

Harrison
SA
,
Gawrieh
S
,
Roberts
K
et al. .
Prospective evaluation of the prevalence of non-alcoholic fatty liver disease and steatohepatitis in a large middle-aged US cohort
.
J Hepatol
2021
;
75
:
284
91
.

8.

Huang
DQ
,
Singal
AG
,
Kono
Y
et al. .
Changing global epidemiology of liver cancer from 2010 to 2019: NASH is the fastest growing cause of liver cancer
.
Cell Metab
2022
;
34
:
969
77.e2
.

9.

Zhou
F
,
Zhou
J
,
Wang
W
et al. .
Unexpected rapid increase in the burden of NAFLD in China from 2008 to 2018: a systematic review and meta-analysis
.
Hepatology
2019
;
70
:
1119
33
.

10.

Eslam
M
,
Sanyal
AJ
,
George
J
.
MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease
.
Gastroenterology
2020
;
158
:
1999
2014.e1
.

11.

Buzzetti
E
,
Pinzani
M
,
Tsochatzis
EA.
The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD)
.
Metabolism
2016
;
65
:
1038
48
.

12.

Schwabe
RF
,
Tabas
I
,
Pajvani
UB.
Mechanisms of fibrosis development in nonalcoholic steatohepatitis
.
Gastroenterology
2020
;
158
:
1913
28
.

13.

Vuppalanchi
R
,
Noureddin
M
,
Alkhouri
N
et al. .
Therapeutic pipeline in nonalcoholic steatohepatitis
.
Nature Rev Gastroenterol Hepatol
2021
;
18
:
373
92
.

14.

Vilar-Gomez
E
,
Martinez-Perez
Y
,
Calzadilla-Bertot
L
et al. .
Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis
.
Gastroenterology
2015
;
149
:
367
78.e5
.

15.

Xu
X
,
Poulsen
KL
,
Wu
L
et al. .
Targeted therapeutics and novel signaling pathways in non-alcohol-associated fatty liver/steatohepatitis (NAFL/NASH)
.
Signal Transduct Target Ther
2022
;
7
:
287
.

16.

Friedman
SL
,
Neuschwander-Tetri
BA
,
Rinella
M
et al. .
Mechanisms of NAFLD development and therapeutic strategies
.
Nat Med
2018
;
24
:
908
22
.

17.

Yang
YY
,
Xie
L
,
Zhang
NP
et al. .
Updates on novel pharmacotherapeutics for the treatment of nonalcoholic steatohepatitis
.
Acta Pharmacol Sin
2022
;
43
:
1180
90
.

18.

Vuppalanchi
R
,
Noureddin
M
,
Alkhouri
N
et al. .
Therapeutic pipeline in nonalcoholic steatohepatitis
.
Nat Rev Gastroenterol Hepatol
2021
;
18
:
373
92
.

19.

Rui
L.
Energy metabolism in the liver
.
Compr Physiol
2014
;
4
:
177
97
.

20.

Hodson
L
,
Gunn
PJ.
The regulation of hepatic fatty acid synthesis and partitioning: the effect of nutritional state
.
Nat Rev Endocrinol
2019
;
15
:
689
700
.

21.

Korenblat
KM
,
Fabbrini
E
,
Mohammed
BS
et al. .
Liver, muscle, and adipose tissue insulin action is directly related to intrahepatic triglyceride content in obese subjects
.
Gastroenterology
2008
;
134
:
1369
75
.

22.

Marques-Lopes
I
,
Ansorena
D
,
Astiasaran
I
et al. .
Postprandial de novo lipogenesis and metabolic changes induced by a high-carbohydrate, low-fat meal in lean and overweight men
.
Am J Clin Nutr
2001
;
73
:
253
61
.

23.

Todoric
J
,
Di Caro
G
,
Reibe
S
et al. .
Fructose stimulated de novo lipogenesis is promoted by inflammation
.
Nat Metab
2020
;
2
:
1034
45
.

24.

Zhao
S
,
Jang
C
,
Liu
J
et al. .
Dietary fructose feeds hepatic lipogenesis via microbiota-derived acetate
.
Nature
2020
;
579
:
586
91
.

25.

Smith
GI
,
Shankaran
M
,
Yoshino
M
et al. .
Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease
.
J Clin Invest
2020
;
130
:
1453
60
.

26.

Lambert
JE
,
Ramos-Roman
MA
,
Browning
JD
et al. .
Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease
.
Gastroenterology
2014
;
146
:
726
35
.

27.

Moore
MP
,
Cunningham
RP
,
Meers
GM
et al. .
Compromised hepatic mitochondrial fatty acid oxidation and reduced markers of mitochondrial turnover in human NAFLD
.
Hepatology
2022
;
76
:
1452
65
.

28.

Li
Y
,
Xu
J
,
Lu
Y
et al. .
DRAK2 aggravates nonalcoholic fatty liver disease progression through SRSF6-associated RNA alternative splicing
.
Cell Metab
2021
;
33
:
2004
20.e9
.

29.

Horn
CL
,
Morales
AL
,
Savard
C
et al. .
Role of cholesterol-associated steatohepatitis in the development of NASH
.
Hepatology Commun
2022
;
6
:
12
35
.

30.

Ioannou
GN.
The role of cholesterol in the pathogenesis of NASH
.
Trends Endocrinol Metabol: TEM
2016
;
27
:
84
95
.

31.

Wang
XB
,
Cai
BS
,
Yang
XM
et al. .
Cholesterol stabilizes TAZ in hepatocytes to promote experimental non-alcoholic steatohepatitis
.
Cell Metab
2020
;
31
:
969
86
.

32.

Jiang
SY
,
Yang
XL
,
Yang
ZM
et al. .
Discovery of an insulin-induced gene binding compound that ameliorates nonalcoholic steatohepatitis by inhibiting sterol regulatory element-binding protein-mediated lipogenesis
.
Hepatology
2022
;
76
:
1466
81
.

33.

Van Rooyen
DM
,
Gan
LT
,
Yeh
MM
et al. .
Pharmacological cholesterol lowering reverses fibrotic NASH in obese, diabetic mice with metabolic syndrome
.
J Hepatol
2013
;
59
:
144
52
.

34.

Taub
R
,
Chiang
E
,
Chabot-Blanchet
M
et al. .
Lipid lowering in healthy volunteers treated with multiple doses of MGL-3196, a liver-targeted thyroid hormone receptor-β agonist
.
Atherosclerosis
2013
;
230
:
373
80
.

35.

Harrison
SA
,
Taub
R
,
Neff
GW
et al. .
Resmetirom for nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled phase 3 trial
.
Nat Med
2023
;
29
:
2919
28
.

36.

Turpin
SM
,
Nicholls
HT
,
Willmes
DM
et al. .
Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance
.
Cell Metab
2014
;
20
:
678
86
.

37.

Hajduch
E
,
Lachkar
F
,
Ferré
P
et al. .
Roles of ceramides in non-alcoholic fatty liver disease
.
J Clin Med
2021
;
10
:
792
.

38.

Raichur
S
,
Wang
ST
,
Chan
PW
et al. .
CerS2 haploinsufficiency inhibits β-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance
.
Cell Metab
2014
;
20
:
687
95
.

39.

Tamimi
TI
,
Elgouhari
HM
,
Alkhouri
N
et al. .
An apoptosis panel for nonalcoholic steatohepatitis diagnosis
.
J Hepatol
2011
;
54
:
1224
9
.

40.

Hatting
M
,
Zhao
G
,
Schumacher
F
et al. .
Hepatocyte caspase-8 is an essential modulator of steatohepatitis in rodents
.
Hepatology
2013
;
57
:
2189
201
.

41.

Thapaliya
S
,
Wree
A
,
Povero
D
et al. .
Caspase 3 inactivation protects against hepatic cell death and ameliorates fibrogenesis in a diet-induced NASH Model
.
Dig Dis Sci
2014
;
59
:
1197
206
.

42.

Peiseler
M
,
Schwabe
R
,
Hampe
J
et al. .
Immune mechanisms linking metabolic injury to inflammation and fibrosis in fatty liver disease — novel insights into cellular communication circuits
.
J Hepatol
2022
;
77
:
1136
60
.

43.

Albhaisi
S
,
Noureddin
M.
Current and potential therapies targeting inflammation in NASH
.
Front Endocrinol (Lausanne)
2021
;
12
:
767314
.

44.

Roh
YS
,
Seki
E.
Toll-like receptors in alcoholic liver disease, non-alcoholic steatohepatitis and carcinogenesis
.
J Gastroenterol Hepatol
2013
;
28
:
38
42
.

45.

Wen
Y
,
Lambrecht
J
,
Ju
C
et al. .
Hepatic macrophages in liver homeostasis and diseases-diversity, plasticity and therapeutic opportunities
.
Cell Mol Immunol
2021
;
18
:
45
56
.

46.

Gomes
AL
,
Teijeiro
A
,
Buren
S
et al. .
Metabolic inflammation-associated IL-17A causes non-alcoholic steatohepatitis and hepatocellular carcinoma
.
Cancer Cell
2016
;
30
:
161
75
.

47.

Rau
M
,
Schilling
AK
,
Meertens
J
et al. .
Progression from nonalcoholic fatty liver to nonalcoholic steatohepatitis is marked by a higher frequency of Th17 cells in the liver and an increased Th17/resting regulatory T cell ratio in peripheral blood and in the liver
.
J Immunol
2016
;
196
:
97
105
.

48.

Dibra
D
,
Xia
X
,
Mitra
A
et al. .
Mutant p53 in concert with an IL27 receptor α deficiency causes spontaneous liver inflammation, fibrosis, and steatosis in mice
.
Hepatology
2016
;
63
:
1000
12
.

49.

Dudek
M
,
Pfister
D
,
Donakonda
S
et al. .
Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH
.
Nature
2021
;
592
:
444
9
.

50.

Mederacke
I
,
Hsu
CC
,
Troeger
JS
et al. .
Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology
.
Nat Commun
2013
;
4
:
2823
.

51.

Feng
G
,
Valenti
L
,
Wong
VW
et al. .
Recompensation in cirrhosis: unravelling the evolving natural history of nonalcoholic fatty liver disease
.
Nat Rev Gastroenterol Hepatol
2024
;
21
:
46
56
.

52.

Hammerich
L
,
Tacke
F.
Hepatic inflammatory responses in liver fibrosis
.
Nat Rev Gastroenterol Hepatol
2023
;
20
:
633
46
.

53.

Henderson
NC
,
Arnold
TD
,
Katamura
Y
et al. .
Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs
.
Nat Med
2013
;
19
:
1617
24
.

54.

Henderson
NC
,
Rieder
F
,
Wynn
TA.
Fibrosis: from mechanisms to medicines
.
Nature
2020
;
587
:
555
66
.

55.

Borkham-Kamphorst
E
,
Weiskirchen
R.
The PDGF system and its antagonists in liver fibrosis
.
Cytokine Growth Factor Rev
2016
;
28
:
53
61
.

56.

Ying
HZ
,
Chen
Q
,
Zhang
WY
et al. .
PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review)
.
Mol Med Rep
2017
;
16
:
7879
89
.

57.

Heymann
F
,
Tacke
F.
Immunology in the liver — from homeostasis to disease
.
Nat Rev Gastroenterol Hepatol
2016
;
13
:
88
110
.

58.

Shen
H
,
Sheng
L
,
Chen
Z
et al. .
Mouse hepatocyte overexpression of NF-κB-inducing kinase (NIK) triggers fatal macrophage-dependent liver injury and fibrosis
.
Hepatology
2014
;
60
:
2065
76
.

59.

Harrison
SA
,
Bedossa
P
,
Guy
CD
et al. .
A phase 3, randomized, controlled trial of resmetirom in NASH with liver fibrosis
.
N Engl J Med
2024
;
390
:
497
509
.

60.

Erion
MD
,
Cable
EE
,
Ito
BR
et al. .
Targeting thyroid hormone receptor-β agonists to the liver reduces cholesterol and triglycerides and improves the therapeutic index
.
Proc Natl Acad Sci U S A
2007
;
104
:
15490
5
.

61.

Francque
SM
,
Bedossa
P
,
Ratziu
V
et al. .
A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH
.
N Engl J Med
2021
;
385
:
1547
58
.

62.

Ratziu
V
,
Harrison
SA
,
Francque
S
et al. .
Elafibranor, an agonist of the peroxisome proliferator-activated receptor-α and -δ, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening
.
Gastroenterology
2016
;
150
:
1147
59.e5
.

63.

Gawrieh
S
,
Noureddin
M
,
Loo
N
et al. .
Saroglitazar, a PPAR-α/γ agonist, for treatment of NAFLD: a randomized controlled double-blind phase 2 trial
.
Hepatology
2021
;
74
:
1809
24
.

64.

Sanyal
AJ
,
Chalasani
N
,
Kowdley
KV
et al. .
Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis
.
N Engl J Med
2010
;
362
:
1675
85
.

65.

Armstrong
MJ
,
Houlihan
DD
,
Rowe
IA.
Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis
.
N Engl J Med
2010
;
363
:
1185
6
.

66.

Das Pradhan
A
,
Glynn
RJ
,
Fruchart
JC
et al.
Triglyceride lowering with pemafibrate to reduce cardiovascular risk
.
N Engl J Med
2022
;
387
:
1923
34
.

67.

Shinozaki
S
,
Tahara
T
,
Miura
K
et al. .
Pemafibrate therapy for non-alcoholic fatty liver disease is more effective in lean patients than obese patients
.
Clin Exp Hepatol
2022
;
8
:
278
83
.

68.

Younossi
ZM
,
Ratziu
V
,
Loomba
R
et al.  
Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial
.
Lancet
2019
;
394
:
2184
96
.

69.

Rinella
ME
,
Dufour
JF
,
Anstee
QM
et al. .
Non-invasive evaluation of response to obeticholic acid in patients with NASH: results from the REGENERATE study
.
J Hepatol
2022
;
76
:
536
48
.

70.

Loomba
R
,
Noureddin
M
,
Kowdley
KV
et al.
Combination therapies including cilofexor and firsocostat for bridging fibrosis and cirrhosis attributable to NASH
.
Hepatology
2021
;
73
:
625
43
.

71.

Alkhouri
N
,
Herring
R
,
Kabler
H
et al. .
Safety and efficacy of combination therapy with semaglutide, cilofexor and firsocostat in patients with non-alcoholic steatohepatitis: a randomised, open-label phase II trial
.
J Hepatol
2022
;
77
:
607
18
.

72.

Sanyal
AJ
,
Lopez
P
,
Lawitz
EJ
et al. .
Tropifexor for nonalcoholic steatohepatitis: an adaptive, randomized, placebo-controlled phase 2a/b trial
.
Nat Med
2023
;
29
:
392
400
.

73.

Newsome
PN
,
Buchholtz
K
,
Cusi
K
et al.
A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis
.
N Engl J Med
2021
;
384
:
1113
24
.

74.

Kuchay
MS
,
Krishan
S
,
Mishra
SK
et al. .
Effect of dulaglutide on liver fat in patients with type 2 diabetes and NAFLD: randomised controlled trial (D-LIFT trial)
.
Diabetologia
2020
;
63
:
2434
45
.

75.

Armstrong
MJ
,
Gaunt
P
,
Aithal
GP
et al.
Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study
.
Lancet
2016
;
387
:
679
90
.

76.

Eguchi
Y
,
Kitajima
Y
,
Hyogo
H
et al.
Pilot study of liraglutide effects in non-alcoholic steatohepatitis and non-alcoholic fatty liver disease with glucose intolerance in Japanese patients (LEAN-J)
.
Hepatol Res
2015
;
45
:
269
78
.

77.

Khoo
J
,
Hsiang
JC
,
Taneja
R
et al. .
Randomized trial comparing effects of weight loss by liraglutide with lifestyle modification in non-alcoholic fatty liver disease
.
Liver Int
2019
;
39
:
941
9
.

78.

Loomba
R
,
Kayali
Z
,
Noureddin
M
et al. .
GS-0976 reduces hepatic steatosis and fibrosis markers in patients with nonalcoholic fatty liver disease
.
Gastroenterology
2018
;
155
:
1463
73.e6
.

79.

Kim
CW
,
Addy
C
,
Kusunoki
J
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
:
576
.

80.

Loomba
R
,
Mohseni
R
,
Lucas
KJ
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
86
.

81.

Ratziu
V
,
de Guevara
L
,
Safadi
R
et al.
Aramchol in patients with nonalcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase 2b trial
.
Nat Med
2021
;
27
:
1825
35
.

82.

Morrow
MR
,
Batchuluun
B
,
Wu
JH
et al. .
Inhibition of ATP-citrate lyase improves NASH, liver fibrosis, and dyslipidemia
.
Cell Metab
2022
;
34
:
919
36.e8
.

83.

Harrison
SA
,
Ruane
PJ
,
Freilich
B
et al. .
A randomized, double-blind, placebo-controlled phase IIa trial of efruxifermin for patients with compensated NASH cirrhosis
.
JHEP Rep
2023
;
5
:
100563
.

84.

Harrison
SA
,
Ruane
PJ
,
Freilich
BL
et al. .
Efruxifermin in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a trial
.
Nat Med
2021
;
27
:
1262
71
.

85.

Harrison
SA
,
Frias
JP
,
Neff
G
et al.
Safety and efficacy of once-weekly efruxifermin versus placebo in non-alcoholic steatohepatitis (HARMONY): a multicentre, randomised, double-blind, placebo-controlled, phase 2b trial
.
Lancet Gastroenterol Hepatol
2023
;
8
:
1080
93
.

86.

Loomba
R
,
Lawitz
EJ
,
Frias
JP
et al. .
Safety, pharmacokinetics, and pharmacodynamics of pegozafermin in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 1b/2a multiple-ascending-dose study
.
Lancet Gastroenterol Hepatol
2023
;
8
:
120
32
.

87.

Loomba
R
,
Sanyal
AJ
,
Kowdley
KV
et al. .
Randomized, controlled trial of the FGF21 analogue pegozafermin in NASH
.
N Engl J Med
2023
;
389
:
998
1008
.

88.

Harrison
SA
,
Alkhouri
N
,
Davison
BA
et al. .
Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase IIb study
.
J Hepatol
2020
;
72
:
613
26
.

89.

Friedman
S
,
Sanyal
A
,
Goodman
Z
et al. .
Efficacy and safety study of cenicriviroc for the treatment of non-alcoholic steatohepatitis in adult subjects with liver fibrosis: CENTAUR phase 2b study design
.
Contemp Clin Trials
2016
;
47
:
356
65
.

90.

Anstee
QM
,
Neuschwander-Tetri
BA
,
Wai-Sun Wong
V
et al. .
Cenicriviroc lacked efficacy to treat liver fibrosis in nonalcoholic steatohepatitis: AURORA phase III randomized study
.
Clin Gastroenterol Hepatol
2024
;
22
:
124
34.e1
.

91.

Loomba
R
,
Lawitz
E
,
Mantry
PS
et al.
The ASK1 inhibitor selonsertib in patients with nonalcoholic steatohepatitis: a randomized, phase 2 trial
.
Hepatology
2018
;
67
:
549
59
.

92.

Harrison
SA
,
Wong
VW
,
Okanoue
T
et al.
Selonsertib for patients with bridging fibrosis or compensated cirrhosis due to NASH: results from randomized phase III STELLAR trials
.
J Hepatol
2020
;
73
:
26
39
.

93.

Calle
RA
,
Amin
NB
,
Carvajal-Gonzalez
S
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
48
.

94.

Pockros
PJ
,
Fuchs
M
,
Freilich
B
et al. .
CONTROL: a randomized phase 2 study of obeticholic acid and atorvastatin on lipoproteins in nonalcoholic steatohepatitis patients
.
Liver Int
2019
;
39
:
2082
93
.

95.

Zucchi
R.
Thyroid hormone analogues: an update
.
Thyroid
2020
;
30
:
1099
105
.

96.

Jakobsson
T
,
Vedin
LL
,
Parini
P.
Potential role of thyroid receptor β agonists in the treatment of hyperlipidemia
.
Drugs
2017
;
77
:
1613
21
.

97.

Vidal-Cevallos
P
,
Murua-Beltran Gall
S
,
Uribe
M
et al. .
Understanding the relationship between nonalcoholic fatty liver disease and thyroid disease
.
Int J Mol Sci
2023
;
24
:
14605
.

98.

Ritter
MJ
,
Amano
I
,
Hollenberg
AN.
Thyroid hormone signaling and the liver
.
Hepatology
2020
;
72
:
742
52
.

99.

Bruinstroop
E
,
Dalan
R
,
Cao
Y
et al. .
Low-dose levothyroxine reduces intrahepatic lipid content in patients with type 2 diabetes mellitus and NAFLD
.
J Clin Endocrinol Metab
2018
;
103
:
2698
706
.

100.

Trost
SU
,
Swanson
E
,
Gloss
B
et al. .
The thyroid hormone receptor-β-selective agonist GC-1 differentially affects plasma lipids and cardiac activity
.
Endocrinology
2000
;
141
:
3057
64
.

101.

Villicev
CM
,
Freitas
FRS
,
Aoki
MS
et al. .
Thyroid hormone receptor β-specific agonist GC-1 increases energy expenditure and prevents fat-mass accumulation in rats
.
J Endocrinol
2007
;
193
:
21
9
.

102.

Saponaro
F
,
Sestito
S
,
Runfola
M
et al. .
Selective thyroid hormone receptor-beta (TRβ) agonists: new perspectives for the treatment of metabolic and neurodegenerative disorders
.
Front Med (Lausanne)
2020
;
7
:
331
.

103.

Berkenstam
A
,
Kristensen
J
,
Mellstrom
K
et al. .
The thyroid hormone mimetic compound KB2115 lowers plasma LDL cholesterol and stimulates bile acid synthesis without cardiac effects in humans
.
Proc Natl Acad Sci U S A
2008
;
105
:
663
7
.

104.

Baxter
JD
,
Webb
P.
Thyroid hormone mimetics: potential applications in atherosclerosis, obesity and type 2 diabetes
.
Nat Rev Drug Discov
2009
;
8
:
308
20
.

105.

Sjouke
B
,
Langslet
G
,
Ceska
R
et al. .
Eprotirome in patients with familial hypercholesterolaemia (the AKKA trial): a randomised, double-blind, placebo-controlled phase 3 study
.
Lancet Diabetes Endocrinol
2014
;
2
:
455
63
.

106.

Hones
GS
,
Sivakumar
RG
,
Hoppe
C
et al. .
Cell-specific transport and thyroid hormone receptor isoform selectivity account for hepatocyte-targeted thyromimetic action of MGL-3196
.
Int J Mol Sci
2022
;
23
:
13714
.

107.

Karim
G
,
Bansal
MB.
Resmetirom: an orally administered, smallmolecule, liver-directed, β-selective THR agonist for the treatment of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis
.
touchREV Endocrinol
2023
;
19
:
60
70
.

108.

Tanaka
N
,
Aoyama
T
,
Kimura
S
et al. .
Targeting nuclear receptors for the treatment of fatty liver disease
.
Pharmacol Ther
2017
;
179
:
142
57
.

109.

Poulsen
L
,
Siersbaek
M
,
Mandrup
S.
PPARs: fatty acid sensors controlling metabolism
.
Semin Cell Dev Biol
2012
;
23
:
631
9
.

110.

Tong
L
,
Wang
L
,
Yao
S
et al. .
PPARδ attenuates hepatic steatosis through autophagy-mediated fatty acid oxidation
.
Cell Death Dis
2019
;
10
:
197
.

111.

Tao
L
,
Wu
L
,
Zhang
W
et al. .
Peroxisome proliferator-activated receptor γ inhibits hepatic stellate cell activation regulated by miR-942 in chronic hepatitis B liver fibrosis
.
Life Sci
2020
;
253
:
117572
.

112.

Lefere
S
,
Puengel
T
,
Hundertmark
J
et al. .
Differential effects of selective- and pan-PPAR agonists on experimental steatohepatitis and hepatic macrophages
.
J Hepatol
2020
;
73
:
757
70
.

113.

Ratziu
V
,
Giral
P
,
Jacqueminet
S
et al.
Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) Trial
.
Gastroenterology
2008
;
135
:
100
10
.

114.

Gross
B
,
Pawlak
M
,
Lefebvre
P
et al. .
PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD
.
Nat Rev Endocrinol
2017
;
13
:
36
49
.

115.

Francque
S
,
Szabo
G
,
Abdelmalek
MF
et al. .
Nonalcoholic steatohepatitis: the role of peroxisome proliferator-activated receptors
.
Nat Rev Gastroenterol Hepatol
2021
;
18
:
24
39
.

116.

Gong
L
,
Wei
F
,
Gonzalez
FJ
et al. .
Hepatic fibrosis: targeting peroxisome proliferator-activated receptor alpha from mechanism to medicines
.
Hepatology
2023
;
78
:
1625
53
.

117.

Murakami
K
,
Sasaki
Y
,
Asahiyama
M
et al. .
Selective PPARα modulator pemafibrate and sodium-glucose cotransporter 2 inhibitor tofogliflozin combination treatment improved histopathology in experimental mice model of non-alcoholic steatohepatitis
.
Cells
2022
;
11
:
720
.

118.

Cariou
B
,
Zair
Y
,
Staels
B
et al. .
Effects of the new dual PPAR α/δ agonist GFT505 on lipid and glucose homeostasis in abdominally obese patients with combined dyslipidemia or impaired glucose metabolism
.
Diabetes Care
2011
;
34
:
2008
14
.

119.

Wettstein
G
,
Luccarini
JM
,
Poekes
L
et al. .
The new-generation pan-peroxisome proliferator-activated receptor agonist IVA337 protects the liver from metabolic disorders and fibrosis
.
Hepatol Commun
2017
;
1
:
524
37
.

120.

Feng
Z
,
Xiang
J
,
Liu
H
et al. .
Design, synthesis, and biological evaluation of triazolone derivatives as potent PPARα/δ dual agonists for the treatment of nonalcoholic steatohepatitis
.
J Med Chem
2022
;
65
:
2571
92
.

121.

Xiang
C
,
Chen
X
,
Yao
J
et al. .
Design, synthesis and anti-NASH effect evaluation of novel GFT505 derivatives in vitro and in vivo
.
Eur J Med Chem
2023
;
257
:
115510
.

122.

Forman
BM
,
Goode
E
,
Chen
J
et al. .
Identification of a nuclear receptor that is activated by farnesol metabolites
.
Cell
1995
;
81
:
687
93
.

123.

Makishima
M
,
Okamoto
AY
,
Repa
JJ
et al. .
Identification of a nuclear receptor for bile acids
.
Science
1999
;
284
:
1362
5
.

124.

Parks
DJ
,
Blanchard
SG
,
Bledsoe
RK
et al. .
Bile acids: natural ligands for an orphan nuclear receptor
.
Science
1999
;
284
:
1365
8
.

125.

Calkin
AC
,
Tontonoz
P.
Transcriptional integration of metabolism by the nuclear sterol-activated receptors LXR and FXR
.
Nat Rev Mol Cell Biol
2012
;
13
:
213
24
.

126.

Puri
P
,
Daita
K
,
Joyce
A
et al. .
The presence and severity of nonalcoholic steatohepatitis is associated with specific changes in circulating bile acids
.
Hepatology
2018
;
67
:
534
48
.

127.

Ma
K
,
Saha
PK
,
Chan
L
et al. .
Farnesoid X receptor is essential for normal glucose homeostasis
.
J Clin Invest
2006
;
116
:
1102
9
.

128.

Sinal
CJ
,
Tohkin
M
,
Miyata
M
et al. .
Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis
.
Cell
2000
;
102
:
731
44
.

129.

Watanabe
M
,
Houten
SM
,
Wang
L
et al. .
Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c
.
J Clin Invest
2004
;
113
:
1408
18
.

130.

Sirvent
A
,
Claudel
T
,
Martin
G
et al. .
The farnesoid X receptor induces very low density lipoprotein receptor gene expression
.
FEBS Lett
2004
;
566
:
173
7
.

131.

Pineda Torra
I
,
Claudel
T
,
Duval
C
et al. .
Bile acids induce the expression of the human peroxisome proliferator-activated receptor α gene via activation of the farnesoid X receptor
.
Mol Endocrinol
2003
;
17
:
259
72
.

132.

Cyphert
HA
,
Ge
X
,
Kohan
AB
et al. .
Activation of the farnesoid X receptor induces hepatic expression and secretion of fibroblast growth factor 21
.
J Biol Chem
2012
;
287
:
25123
38
.

133.

Schumacher
JD
,
Guo
GL.
Pharmacologic modulation of bile acid-FXR-FGF15/FGF19 pathway for the treatment of nonalcoholic steatohepatitis
.
Handb Exp Pharmacol
2019
;
256
:
325
57
.

134.

Mencarelli
A
,
Renga
B
,
Migliorati
M
et al. .
The bile acid sensor farnesoid X receptor is a modulator of liver immunity in a rodent model of acute hepatitis
.
J Immunol
2009
;
183
:
6657
66
.

135.

Fiorucci
S
,
Antonelli
E
,
Rizzo
G
et al. .
The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis
.
Gastroenterology
2004
;
127
:
1497
512
.

136.

Yamagata
K
,
Daitoku
H
,
Shimamoto
Y
et al. .
Bile acids regulate gluconeogenic gene expression via small heterodimer partner-mediated repression of hepatocyte nuclear factor 4 and Foxo1
.
J Biol Chem
2004
;
279
:
23158
65
.

137.

Caron
S
,
Huaman Samanez
C
,
Dehondt
H
et al. .
Farnesoid X receptor inhibits the transcriptional activity of carbohydrate response element binding protein in human hepatocytes
.
Mol Cell Biol
2013
;
33
:
2202
11
.

138.

Cariou
B
,
van Harmelen
K
,
Duran-Sandoval
D
et al. .
The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice
.
J Biol Chem
2006
;
281
:
11039
49
.

139.

Pellicciari
R
,
Fiorucci
S
,
Camaioni
E
et al. .
6α-ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity
.
J Med Chem
2002
;
45
:
3569
72
.

140.

Maloney
PR
,
Parks
DJ
,
Haffner
CD
et al. .
Identification of a chemical tool for the orphan nuclear receptor FXR
.
J Med Chem
2000
;
43
:
2971
4
.

141.

Li
J
,
Liu
M
,
Li
Y
et al. .
Discovery and optimization of non-bile acid FXR agonists as preclinical candidates for the treatment of nonalcoholic steatohepatitis
.
J Med Chem
2020
;
63
:
12748
72
.

142.

Fang
Y
,
Hegazy
L
,
Finck
BN
et al. .
Recent advances in the medicinal chemistry of farnesoid X receptor
.
J Med Chem
2021
;
64
:
17545
71
.

143.

Jinnouchi
H
,
Sugiyama
S
,
Yoshida
A
et al. .
Liraglutide, a glucagon-like peptide-1 analog, increased insulin sensitivity assessed by hyperinsulinemic-euglycemic clamp examination in patients with uncontrolled type 2 diabetes mellitus
.
J Diabetes Res
2015
;
2015
:
706416
.

144.

Frias
JP
,
Nauck
MA
,
Van
J
et al. .
Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial
.
Lancet
2018
;
392
:
2180
93
.

145.

Mahapatra
MK
,
Karuppasamy
M
,
Sahoo
BM.
Therapeutic potential of semaglutide, a newer GLP-1 receptor agonist, in abating obesity, non-alcoholic steatohepatitis and neurodegenerative diseases: a narrative review
.
Pharm Res
2022
;
39
:
1233
48
.

146.

Wang
Z
,
Ye
M
,
Zhang
XJ
et al. .
Impact of NAFLD and its pharmacotherapy on lipid profile and CVD
.
Atherosclerosis
2022
;
355
:
30
44
.

147.

Chao
AM
,
Tronieri
JS
,
Amaro
A
et al. .
Semaglutide for the treatment of obesity
.
Trends Cardiovasc Med
2023
;
33
:
159
66
.

148.

Targher
G
,
Mantovani
A
,
Byrne
CD.
Mechanisms and possible hepatoprotective effects of glucagon-like peptide-1 receptor agonists and other incretin receptor agonists in non-alcoholic fatty liver disease
.
Lancet Gastroenterol Hepatol
2023
;
8
:
179
91
.

149.

Hartman
ML
,
Sanyal
AJ
,
Loomba
R
et al. .
Effects of novel dual GIP and GLP-1 receptor agonist tirzepatide on biomarkers of nonalcoholic steatohepatitis in patients with type 2 diabetes
.
Diabetes Care
2020
;
43
:
1352
5
.

150.

Jeon
YG
,
Kim
YY
,
Lee
G
et al. .
Physiological and pathological roles of lipogenesis
.
Nat Metab
2023
;
5
:
735
59
.

151.

Tong
L.
Acetyl-coenzyme A carboxylase: crucial metabolic enzyme and attractive target for drug discovery
.
Cell Mol Life Sci
2005
;
62
:
1784
803
.

152.

Bian
H
,
Liu
YM
,
Chen
ZN.
New avenues for NASH therapy by targeting ACC
.
Cell Metab
2022
;
34
:
191
3
.

153.

Bates
J
,
Vijayakumar
A
,
Ghoshal
S
et al. .
Acetyl-CoA carboxylase inhibition disrupts metabolic reprogramming during hepatic stellate cell activation
.
J Hepatol
2020
;
73
:
896
905
.

154.

Harriman
G
,
Greenwood
J
,
Bhat
S
et al. .
Acetyl-CoA carboxylase inhibition by ND-630 reduces hepatic steatosis, improves insulin sensitivity, and modulates dyslipidemia in rats
.
Proc Natl Acad Sci U S A
2016
;
113
:
E1796
805
.

155.

Zhang
J
,
Muise
ES
,
Han
S
et al. .
Molecular profiling reveals a common metabolic signature of tissue fibrosis
.
Cell Rep Med
2020
;
1
:
100056
.

156.

Ross
TT
,
Crowley
C
,
Kelly
KL
et al. .
Acetyl-CoA carboxylase inhibition improves multiple dimensions of NASH pathogenesis in model systems
.
Cell Mol Gastroenterol Hepatol
2020
;
10
:
829
51
.

157.

Zhang
XJ
,
Ji
YX
,
Cheng
X
et al. .
A small molecule targeting ALOX12-ACC1 ameliorates nonalcoholic steatohepatitis in mice and macaques
.
Sci Transl Med
2021
;
13
:
eabg8116
.

158.

Zhang
XJ
,
She
ZG
,
Wang
J
et al. .
Multiple omics study identifies an interspecies conserved driver for nonalcoholic steatohepatitis
.
Sci Transl Med
2021
;
13
:
eabg8117
.

159.

Ogawa
Y
,
Imajo
K
,
Honda
Y
et al. .
Palmitate-induced lipotoxicity is crucial for the pathogenesis of nonalcoholic fatty liver disease in cooperation with gut-derived endotoxin
.
Sci Rep
2018
;
8
:
11365
.

160.

Wang
H
,
Zhou
Y
,
Xu
H
et al. .
Therapeutic efficacy of FASN inhibition in preclinical models of HCC
.
Hepatology
2022
;
76
:
951
66
.

161.

O’Farrell
M
,
Duke
G
,
Crowley
R
et al. .
FASN inhibition targets multiple drivers of NASH by reducing steatosis, inflammation and fibrosis in preclinical models
.
Sci Rep
2022
;
12
:
15661
.

162.

Miyazaki
M
,
Sampath
H
,
Liu
X
et al. .
Stearoyl-CoA desaturase-1 deficiency attenuates obesity and insulin resistance in leptin-resistant obese mice
.
Biochem Biophys Res Commun
2009
;
380
:
818
22
.

163.

Miyazaki
M
,
Flowers
MT
,
Sampath
H
et al. .
Hepatic stearoyl-CoA desaturase-1 deficiency protects mice from carbohydrate-induced adiposity and hepatic steatosis
.
Cell Metab
2007
;
6
:
484
96
.

164.

Issandou
M
,
Bouillot
A
,
Brusq
JM
et al. .
Pharmacological inhibition of stearoyl-CoA desaturase 1 improves insulin sensitivity in insulin-resistant rat models
.
Eur J Pharmacol
2009
;
618
:
28
36
.

165.

Iruarrizaga-Lejarreta
M
,
Varela-Rey
M
,
Fernandez-Ramos
D
et al. .
Role of aramchol in steatohepatitis and fibrosis in mice
.
Hepatol Commun
2017
;
1
:
911
27
.

166.

Bhattacharya
D
,
Basta
B
,
Mato
JM
et al. .
Aramchol downregulates stearoyl CoA-desaturase 1 in hepatic stellate cells to attenuate cellular fibrogenesis
.
JHEP Rep
2021
;
3
:
100237
.

167.

Dongiovanni
P
,
Petta
S
,
Mannisto
V
et al. .
Statin use and non-alcoholic steatohepatitis in at risk individuals
.
J Hepatol
2015
;
63
:
705
12
.

168.

Fatima
K
,
Moeed
A
,
Waqar
E
et al. .
Efficacy of statins in treatment and development of non-alcoholic fatty liver disease and steatohepatitis: a systematic review and meta-analysis
.
Clin Res Hepatol Gastroenterol
2022
;
46
:
101816
.

169.

Braun
LR
,
Feldpausch
MN
,
Czerwonka
N
et al. .
Effects of pitavastatin on insulin sensitivity and liver fat: a randomized clinical trial
.
J Clin Endocrinol Metab
2018
;
103
:
4176
86
.

170.

Kimura
Y
,
Hyogo
H
,
Yamagishi
S
et al. .
Atorvastatin decreases serum levels of advanced glycation endproducts (AGEs) in nonalcoholic steatohepatitis (NASH) patients with dyslipidemia: clinical usefulness of AGEs as a biomarker for the attenuation of NASH
.
J Gastroenterol
2010
;
45
:
750
7
.

171.

Athyros
VG
,
Boutari
C
,
Stavropoulos
K
et al. .
Statins: an under-appreciated asset for the prevention and the treatment of NAFLD or NASH and the related cardiovascular risk
.
Curr Vasc Pharmacol
2018
;
16
:
246
53
.

172.

Wei
J
,
Leit
S
,
Kuai
J
et al. .
An allosteric mechanism for potent inhibition of human ATP-citrate lyase
.
Nature
2019
;
568
:
566
70
.

173.

Xie
Z
,
Zhang
M
,
Song
Q
et al. .
Development of the novel ACLY inhibitor 326E as a promising treatment for hypercholesterolemia
.
Acta Pharm Sin B
2023
;
13
:
739
53
.

174.

Wang
Q
,
Jiang
L
,
Wang
J
et al. .
Abrogation of hepatic ATP-citrate lyase protects against fatty liver and ameliorates hyperglycemia in leptin receptor-deficient mice
.
Hepatology
2009
;
49
:
1166
75
.

175.

Ray
KK
,
Bays
HE
,
Catapano
AL
et al. .
Safety and efficacy of bempedoic acid to reduce LDL cholesterol
.
N Engl J Med
2019
;
380
:
1022
32
.

176.

Pinkosky
SL
,
Newton
RS
,
Day
EA
et al. .
Liver-specific ATP-citrate lyase inhibition by bempedoic acid decreases LDL-C and attenuates atherosclerosis
.
Nat Commun
2016
;
7
:
13457
.

177.

Nissen
SE
,
Lincoff
AM
,
Brennan
D
et al.
Bempedoic acid and cardiovascular outcomes in statin-intolerant patients
.
N Engl J Med
2023
;
388
:
1353
64
.

178.

Morrow
MR
,
Batchuluun
B
,
Wu
J
et al. .
Inhibition of ATP-citrate lyase improves NASH, liver fibrosis, and dyslipidemia
.
Cell Metab
2022
;
34
:
919
36.e8
.

179.

Desjardins
EM
,
Wu
J
,
Lavoie
DCT
et al. .
Combination of an ACLY inhibitor with a GLP-1R agonist exerts additive benefits on nonalcoholic steatohepatitis and hepatic fibrosis in mice
.
Cell Rep Med
2023
;
4
:
101193
.

180.

Cao
J
,
Lockwood
J
,
Burn
P
et al. .
Cloning and functional characterization of a mouse intestinal acyl-CoA:monoacylglycerol acyltransferase, MGAT2
.
J Biol Chem
2003
;
278
:
13860
6
.

181.

Yen
CL
,
Cheong
ML
,
Grueter
C
et al. .
Deficiency of the intestinal enzyme acyl CoA:monoacylglycerol acyltransferase-2 protects mice from metabolic disorders induced by high-fat feeding
.
Nat Med
2009
;
15
:
442
6
.

182.

Hall
AM
,
Kou
K
,
Chen
Z
et al. .
Evidence for regulated monoacylglycerol acyltransferase expression and activity in human liver
.
J Lipid Res
2012
;
53
:
990
9
.

183.

Cheng
D
,
Zinker
BA
,
Luo
Y
et al. .
MGAT2 inhibitor decreases liver fibrosis and inflammation in murine NASH models and reduces body weight in human adults with obesity
.
Cell Metab
2022
;
34
:
1732
48.e5
.

184.

Nishimura
T
,
Nakatake
Y
,
Konishi
M
et al. .
Identification of a novel FGF, FGF-21, preferentially expressed in the liver
.
Biochim Biophys Acta
2000
;
1492
:
203
6
.

185.

Keuper
M
,
Haring
HU
,
Staiger
H.
Circulating FGF21 levels in human health and metabolic disease
.
Exp Clin Endocrinol Diabetes
2020
;
128
:
752
70
.

186.

Lewis
JE
,
Ebling
FJP
,
Samms
RJ
et al. .
Going back to the biology of FGF21: new insights
.
Trends Endocrinol Metab
2019
;
30
:
491
504
.

187.

Li
H
,
Fang
Q
,
Gao
F
et al. .
Fibroblast growth factor 21 levels are increased in nonalcoholic fatty liver disease patients and are correlated with hepatic triglyceride
.
J Hepatol
2010
;
53
:
934
40
.

188.

Dushay
J
,
Chui
PC
,
Gopalakrishnan
GS
et al. .
Increased fibroblast growth factor 21 in obesity and nonalcoholic fatty liver disease
.
Gastroenterology
2010
;
139
:
456
63
.

189.

Morris-Stiff
G
,
Feldstein
AE.
Fibroblast growth factor 21 as a biomarker for NAFLD: integrating pathobiology into clinical practice
.
J Hepatol
2010
;
53
:
795
6
.

190.

Tian
H
,
Zhang
S
,
Liu
Y
et al. .
Fibroblast growth factors for nonalcoholic fatty liver disease: opportunities and challenges
.
Int J Mol Sci
2023
;
24
:
4583
.

191.

Fisher
FM
,
Chui
PC
,
Nasser
IA
et al. .
Fibroblast growth factor 21 limits lipotoxicity by promoting hepatic fatty acid activation in mice on methionine and choline-deficient diets
.
Gastroenterology
2014
;
147
:
1073
83.e6
.

192.

Xu
P
,
Zhang
Y
,
Liu
Y
et al. .
Fibroblast growth factor 21 attenuates hepatic fibrogenesis through TGF-β/smad2/3 and NF-κB signaling pathways
.
Toxicol Appl Pharmacol
2016
;
290
:
43
53
.

193.

Feingold
KR
,
Grunfeld
C
,
Heuer
JG
et al. .
FGF21 is increased by inflammatory stimuli and protects leptin-deficient ob/ob mice from the toxicity of sepsis
.
Endocrinology
2012
;
153
:
2689
700
.

194.

Puengel
T
,
Tacke
F.
Efruxifermin, an investigational treatment for fibrotic or cirrhotic nonalcoholic steatohepatitis (NASH)
.
Expert Opin Investig Drugs
2023
;
32
:
451
61
.

195.

Degirolamo
C
,
Sabba
C
,
Moschetta
A.
Therapeutic potential of the endocrine fibroblast growth factors FGF19, FGF21 and FGF23
.
Nat Rev Drug Discov
2016
;
15
:
51
69
.

196.

Nicholes
K
,
Guillet
S
,
Tomlinson
E
et al. .
A mouse model of hepatocellular carcinoma: ectopic expression of fibroblast growth factor 19 in skeletal muscle of transgenic mice
.
Am J Pathol
2002
;
160
:
2295
307
.

197.

Ursic-Bedoya
J
,
Desandre
G
,
Chavey
C
et al. .
FGF19 and its analog aldafermin cooperate with MYC to induce aggressive hepatocarcinogenesis
.
EMBO Mol Med
2024
;
16
:
238
50
.

198.

Zhou
M
,
Learned
RM
,
Rossi
SJ
et al. .
Engineered FGF19 eliminates bile acid toxicity and lipotoxicity leading to resolution of steatohepatitis and fibrosis in mice
.
Hepatol Commun
2017
;
1
:
1024
42
.

199.

Harrison
SA
,
Rinella
ME
,
Abdelmalek
MF
et al. .
NGM282 for treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial
.
Lancet
2018
;
391
:
1174
85
.

200.

Harrison
SA
,
Neff
G
,
Guy
CD
et al. .
Efficacy and safety of aldafermin, an engineered FGF19 analog, in a randomized, double-blind, placebo-controlled trial of patients with nonalcoholic steatohepatitis
.
Gastroenterology
2021
;
160
:
219
31.e1
.

201.

Harrison
SA
,
Abdelmalek
MF
,
Neff
G
et al. .
Aldafermin in patients with non-alcoholic steatohepatitis (ALPINE 2/3): a randomised, double-blind, placebo-controlled, phase 2b trial
.
Lancet Gastroenterol Hepatol
2022
;
7
:
603
16
.

202.

Li
YF
,
Xie
ZF
,
Song
Q
et al. .
Mitochondria homeostasis: biology and involvement in hepatic steatosis to NASH
.
Acta Pharmacol Sin
2022
;
43
:
1141
55
.

203.

Goedeke
L
,
Shulman
GI.
Therapeutic potential of mitochondrial uncouplers for the treatment of metabolic associated fatty liver disease and NASH
.
Mol Metab
2021
;
46
:
101178
.

204.

Grundlingh
J
,
Dargan
PI
,
El-Zanfaly
M
et al. .
2,4-dinitrophenol (DNP): a weight loss agent with significant acute toxicity and risk of death
.
J Med Toxicol
2011
;
7
:
205
12
.

205.

Wei
G
,
Song
X
,
Fu
Y
et al. .
Sustained-release mitochondrial protonophore reverses nonalcoholic fatty liver disease in rats
.
Int J Pharm
2017
;
530
:
230
8
.

206.

Perry
RJ
,
Kim
T
,
Zhang
XM
et al. .
Reversal of hypertriglyceridemia, fatty liver disease, and insulin resistance by a liver-targeted mitochondrial uncoupler
.
Cell Metab
2013
;
18
:
740
8
.

207.

Jiang
H
,
Jin
J
,
Duan
Y
et al. .
Mitochondrial uncoupling coordinated with PDH activation safely ameliorates hyperglycemia via promoting glucose oxidation
.
Diabetes
2019
;
68
:
2197
209
.

208.

Goedeke
L
,
Peng
L
,
Montalvo-Romeral
V
et al. .
Controlled-release mitochondrial protonophore (CRMP) reverses dyslipidemia and hepatic steatosis in dysmetabolic nonhuman primates
.
Sci Transl Med
2019
;
11
:
eaay0284
.

209.

Tavoulari
S
,
Sichrovsky
M
,
Kunji
ERS.
Fifty years of the mitochondrial pyruvate carrier: new insights into its structure, function, and inhibition
.
Acta Physiol (Oxf)
2023
;
238
:
e14016
.

210.

McCommis
KS
,
Hodges
WT
,
Brunt
EM
et al. .
Targeting the mitochondrial pyruvate carrier attenuates fibrosis in a mouse model of nonalcoholic steatohepatitis
.
Hepatology
2017
;
65
:
1543
56
.

211.

Kamm
DR
,
Pyles
KD
,
Sharpe
MC
et al. .
Novel insulin sensitizer MSDC-0602K improves insulinemia and fatty liver disease in mice, alone and in combination with liraglutide
.
J Biol Chem
2021
;
296
:
100807
.

212.

Hodges
WT
Jarasvaraparn
C
Ferguson
D
et al. .
Mitochondrial pyruvate carrier inhibitors improve metabolic parameters in diet-induced obese mice
.
J Biol Chem
2022
;
298
:
101554
.

213.

Henderson
NC
,
Mackinnon
AC
,
Farnworth
SL
et al. .
Galectin-3 regulates myofibroblast activation and hepatic fibrosis
.
Proc Natl Acad Sci U S A
2006
;
103
:
5060
5
.

214.

Iacobini
C
,
Menini
S
,
Ricci
C
et al. .
Galectin-3 ablation protects mice from diet-induced NASH: a major scavenging role for galectin-3 in liver
.
J Hepatol
2011
;
54
:
975
83
.

215.

Traber
PG
,
Zomer
E.
Therapy of experimental NASH and fibrosis with galectin inhibitors
.
PLoS One
2013
;
8
:
e83481
.

216.

Traber
PG
,
Chou
H
,
Zomer
E
et al. .
Regression of fibrosis and reversal of cirrhosis in rats by galectin inhibitors in thioacetamide-induced liver disease
.
PLoS One
2013
;
8
:
e75361
.

217.

Harrison
SA
,
Marri
SR
,
Chalasani
N
et al. .
Randomised clinical study: GR-MD-02, a galectin-3 inhibitor, vs. placebo in patients having non-alcoholic steatohepatitis with advanced fibrosis
.
Aliment Pharmacol Ther
2016
;
44
:
1183
98
.

218.

Chalasani
N
,
Abdelmalek
MF
,
Garcia-Tsao
G
et al.
Effects of belapectin, an inhibitor of galectin-3, in patients with nonalcoholic steatohepatitis with cirrhosis and portal hypertension
.
Gastroenterology
2020
;
158
:
1334
45.e5
.

219.

Roh
YS
,
Seki
E.
Chemokines and chemokine receptors in the development of NAFLD
.
Adv Exp Med Biol
2018
;
1061
:
45
53
.

220.

Krenkel
O
,
Puengel
T
,
Govaere
O
et al. .
Therapeutic inhibition of inflammatory monocyte recruitment reduces steatohepatitis and liver fibrosis
.
Hepatology
2018
;
67
:
1270
83
.

221.

Kruger
AJ
,
Fuchs
BC
,
Masia
R
et al. .
Prolonged cenicriviroc therapy reduces hepatic fibrosis despite steatohepatitis in a diet-induced mouse model of nonalcoholic steatohepatitis
.
Hepatol Commun
2018
;
2
:
529
45
.

222.

Schuster
S
,
Feldstein
AE.
NASH: novel therapeutic strategies targeting ASK1 in NASH
.
Nat Rev Gastroenterol Hepatol
2017
;
14
:
329
30
.

223.

Dufour
JF
,
Caussy
C
,
Loomba
R.
Combination therapy for non-alcoholic steatohepatitis: rationale, opportunities and challenges
.
Gut
2020
;
69
:
1877
84
.

224.

Lawitz
EJ
,
Bhandari
BR
,
Ruane
PJ
et al. .
Fenofibrate mitigates hypertriglyceridemia in nonalcoholic steatohepatitis patients treated with cilofexor/firsocostat
.
Clin Gastroenterol Hepatol
2023
;
21
:
143
52.e3
.

225.

Kremoser
C.
FXR agonists for NASH: how are they different and what difference do they make
?
J Hepatol
2021
;
75
:
12
5
.

226.

Targher
G
,
Corey
KE
,
Byrne
CD
et al. .
The complex link between NAFLD and type 2 diabetes mellitus — mechanisms and treatments
.
Nat Rev Gastroenterol Hepatol
2021
;
18
:
599
612
.

227.

Leite
NC
,
Salles
GF
,
Araujo
AL
et al. .
Prevalence and associated factors of non-alcoholic fatty liver disease in patients with type-2 diabetes mellitus
.
Liver Int
2009
;
29
:
113
9
.

228.

Harrison
SA
,
Manghi
FP
,
Smith
WB
et al. .
Licogliflozin for nonalcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a study
.
Nat Med
2022
;
28
:
1432
8
.

229.

Ludwig
J
,
Viggiano
TR
,
McGill
DB
et al. .
Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease
.
Mayo Clin Proc
1980
;
55
:
434
8
.

230.

Eslam
M
,
Newsome
PN
,
Sarin
SK
et al. .
A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement
.
J Hepatol
2020
;
73
:
202
9
.

231.

Fan
X
,
Song
Y
,
Zhao
J.
Evolving liver disease insights from NAFLD to MASLD
.
Trends Endocrinol Metab
2024
. Doi: 10.1016/j.tem.2024.02.012.

232.

Fouad
Y
,
Waked
I
,
Bollipo
S
et al. .
What’s in a name? Renaming ‘NAFLD’ to ‘MAFLD’
.
Liver Int
2020
;
40
:
1254
61
.

Author notes

Zhifu Xie and Yufeng Li contributed equally to this work.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.