Next Article in Journal
Hidden Dangers: Herbal and Dietary Supplement Induced Hepatotoxicity
Previous Article in Journal
The Evolution of Circulating Biomarkers for Use in Acetaminophen/Paracetamol-Induced Liver Injury in Humans: A Scoping Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutics for Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD)

by
Kamlesh K. Bhopale
* and
Mukund P. Srinivasan
Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
*
Author to whom correspondence should be addressed.
Livers 2023, 3(4), 597-617; https://doi.org/10.3390/livers3040040
Submission received: 14 September 2023 / Revised: 11 October 2023 / Accepted: 19 October 2023 / Published: 28 October 2023

Abstract

:
Metabolic dysfunction associated fatty liver disease (MAFLD) has been recently recognized as a new global chronic liver disease entity with non-alcoholic fatty liver disease (NAFLD) associated with overweight/obesity or type 2 diabetes mellitus (T2DM) and evidence of metabolic dysregulation. Due to the rising rates of obesity and diabetes, MAFLD is considered a rapidly emerging chronic liver disease globally. Nearly 25–30% of the global population poses health issues due to MAFLD with a substantial economic burden to societies. Disease progression depends on the persistence of risk factors and etiological agents, from simple steatosis, hepatitis, fibrosis, to cirrhosis, and if untreated, leads to hepatocellular carcinoma. In this review article we summarize various risk and etiological factors, diagnostic techniques, and therapeutic evaluation of pharmacological agents developed for MAFLD. Effective pharmaceutical agents for the treatment of MAFLD (and NAFLD) are lacking, and research is ongoing to search for effective medications in this direction. Currently, pioglitazone is advised for MAFLD patients, whereas Vitamin E is advised for non-diabetic MAFLD patients with ≥F2 non-cirrhosis. Current approaches to disease management emphasize diet control, lifestyle changes, and weight loss. In this review, we summarized the pharmacological agents currently being developed and their current status to treat patients with MAFLD.

1. Introduction

The term ‘metabolic dysfunction-associated fatty liver disease’ (MAFLD) refers to the most common form of chronic liver disease recognized previously as non-alcoholic fatty liver disease (NAFLD) and has been now linked to evidence of metabolic dysregulation, such as obesity and/or diabetes. Non-alcoholic fatty liver disease (NAFLD), which is characterized by the accumulation of fat in more than 5% of hepatocytes (detected by imaging techniques or histology), occurs in the absence of excessive alcohol consumption and other secondary causes, such as viral hepatitis. A persistent NAFLD may further progress to non-alcoholic steatohepatitis (NASH), liver fibrosis, liver cirrhosis, or hepatocellular carcinoma (HCC) [1]. This term “non-alcoholic fatty liver disease” was first used by Schaffner and Thaler in 1986 to describe the liver condition of excessive fat accumulation in the patient’s liver without a secondary cause of steatosis [2]. NAFLD is intimately correlated with metabolic syndrome, which comprises abdominal obesity, hyperglycemia, hypertension, and dyslipidemia [3]. NAFLD by definition has limitations or is confined to only steatosis. The diagnostic criteria define hepatic steatosis of ≥5% without concurrent liver disease. With the advancement in understanding of disease and close association of NAFLD with obesity, metabolic syndrome, and type 2 diabetes mellitus (2DM), recently, a consensus international specialist advocated changing the term of NAFLD to metabolic dysfunction-associated fatty liver disease (MAFLD), on the criterion evidence of hepatic steatosis and one of the following three conditions: overweight/obesity, type 2 diabetes mellitus (T2DM), or signs of metabolic dysregulation [4,5]. The term “MAFLD,” was originally proposed in 2020 [5], to identify fatty liver disease conditions linked to metabolic abnormalities. The updates of NAFLD to new definition of MAFLD is considered most appropriate nomenclature and of great value to clinicians in the diagnosis of high-risk patients who are benefited with target management and medical treatment. Moreover, increased understanding in the healthcare community and wider general public awareness have been found to be of great value. Therefore, prevalence of MAFLD may significantly increase when obesity, diabetes, dyslipidemia, and hypertension are taken combinedly into the consideration [6,7]. MAFLD has an enhanced risk of mortality and a similar risk of cause-specific mortality in NAFLD [8]. MAFLD is also associated with hepatic cardiovascular and oncological sequences, resulting in an economic burden on society [9]. A considerable proportion of MAFLD patients are non-obese, and the spectrum of MAFLD varies from steatosis to concomitant inflammation, which can lead to fibrosis, cirrhosis, and hepatocellular carcinoma (HCC), with only a few patients progressing to more advanced disease stages [10,11] Research work in search of new molecules to effectively treat NAFLD/MAFLD is ongoing.
MAFLD is rapidly emerging as a major contributor to chronic liver disease in the United States because of the rising prevalence of type 2 diabetes and obesity. Nearly 25% of the world’s population is affected by MAFLD, which has a serious fiscal impact in many countries.
MAFLD has become a global health issue, accounting for 25–30% in Asia Pacific region and 25% in Western countries [12]. The estimated global prevalence of MAFLD in overweight or obese adults as reported by Liu et al. (2022) from around the general population was reported in North America as around 34.0%, Central America 56.3%, South America 70.9%, Northern Europe 31.4%, Western Europe 49.9%, Eastern Europe 81.5%, Southern Europe 52.3%, Eastern Asia 52.1% Southeastern Asia 42.3%, and Southern Asia 47.0% [13]. Such an astonishingly high prevalence rate invites attention from the medical community health policy makers and the general public. MAFLD puts a huge economic burden on society. The national economic burden of NAFLD in terms of direct annual medical costs has been estimated at USD 103 billion in the US and in the UK GBP 5.24 billion/year, France, Italy, and Germany combined EUR 27.7 billion/year spent in the management of MAFLD and related conditions. The economic burden of MAFLD and related conditions in the 10 years time is predicted to increase USD 908 billion in the US and to EUR 302 billion in Europe [12,13,14,15].
Most people between the age group of 40 and 60 years are generally affected by MAFLD although it can also occur in children above the age of 10 years. The prevalence of disease is more in males than in females in the younger age group, whereas in the older age group above 65 years, it is reversed.
An unhealthy, high-carbohydrate, and high-fat diet leads to NAFLD and metabolic diseases such as diabetes. Gut microbial dysbiosis and choline deficiency also play key roles in disease development. The disease mostly goes unnoticed in the initial stages. Medical history, abdominal ultrasonography, and blood/serum biomarkers are recommended as primary diagnostic tools for detecting hepatic steatosis in MAFLD. Although liver biopsy is considered the standard for the diagnosis of fatty liver disease, its invasiveness and unavoidable complications limit its clinical application.
Pathophysiology of MAFLD is not well understood. Accumulation of aberrant lipid metabolism, sedentary life style and unhealthy dietary patterns, insulin resistance, and diabetes are several interlinked pathological factors involved in the pathogenesis of the disease. Hepatic steatosis is a characteristic feature of NAFLD and MAFLD, based on serum biomarkers, imaging methods, or histology, coupled with no alcohol consumption and no other liver disease or causes of hepatic steatosis. In addition, at least one of the criteria, such as being overweight or having type 2 diabetes, is linked with MAFLD. Evidence of metabolic dysregulation, as seen in the form of increased waist circumference, hypertension low serum HDL-cholesterol levels, hypertriglyceridemia, impaired fasting plasma glucose, elevated HOMA-IR (approximates insulin resistance), increased C-Reactive protein (chronic), is also strongly associated with MAFLD. The differential features of NAFLD and MAFLD are presented in Figure 1.
Research work in search of new molecules to effectively treat NAFLD/MAFLD is ongoing [7] since no suitable, safe, and effective pharmaceutical agent is available for the treatment covering the entire spectrum of the disease.
In this review, we describe the different pharmacological agents or targets developed recently for MAFLD as effective medications that can be recommended as per clinical practice guidelines (CPGs) issued by the Liver International Society in the US, which may help to improve the health of MAFLD patients since at present no suitable and potent treatment is available.

2. Materials and Methods

A systematic literature search was conducted to source articles from the following databases: PubMed Central, Ovid-Medline search, and Google scholar. Specific keywords included “Non-Alcoholic Fatty Liver Disease”, “NAFLD” or “Metabolic dysfunction-associated fatty liver disease”, “MAFLD”, combined with any of the terms: “therapeutic agents”, “pharmacological agents”, metabolic disorder, metabolic dysfunction, etiology, diagnosis. Boolean operators, specifically “AND” and “OR”, were also taken into consideration when searching for articles. The articles published within 19 years, i.e., from January 2004 to June 2023 were included and those articles published more than 19 years ago, i.e., before January 2004 were excluded. Thereafter, descriptive characteristics of the only relevant studies were extracted as follows: first author with the year of publication, journal title, the aim of the study, study design, and conclusion of the study.

3. Aetiology

The infiltration of lipids in the liver and their deposition involves various genetic and acquired factors [16]. The primary underlying cause of hepatic steatosis cannot be explained, and alcohol is not a real etiological cause; the disease is defined as non-alcoholic fatty liver disease. Secondary factors such as viral hepatitis are not considered etiological factors for NAFLD.
Metabolic diseases and conditions such as type 2 diabetes, insulin resistance, abdominal obesity, blood pressure, hyperglycemia, and hyperlipidemia are strongly associated with NAFLD. Furthermore, NAFLD is associated with hormonal disorders, persistently elevated transaminase levels, increasing age, and hypoxia.

3.1. Dietary Factors

Dietary factors such as unhealthy diet composition, quantity, and habits leading to obesity are major factors in NAFLD/MAFLD. Further, overnutrition in lean NAFLD and NASH is a major factor of disease according to the Asia-Pacific Working Group (APWG) [17]. Lipid deposition in hepatocytes, oxidative stress, mitochondrial malfunction, and dysregulation of several cytokines are predicted to cause cellular injury, apoptosis, and the onset of NASH [18,19,20]. De novo lipogenesis and inflammation are relevant mechanisms involved in the pathogenesis of NAFLD and NASH [21,22].
The progression of NAFLD to NASH and fibrosis is significantly influenced by diet quality and quantity, particularly omega 6 fatty acids [23,24]. NAFLD onset can be attributed to choline insufficiency [25]. Diets containing high sugar levels, such as fructose or sucrose, increase the risk of NAFLD [26]. The term “glucotoxicity” refers to the harmful effects of high blood glucose levels on cells. T2DM and the evolution of NAFLD are caused by a persistent glucotoxicity with hyperglycemia [27]. Insulin resistance (IR) is caused by low-grade grade glucotoxicity [28]. In addition, glucotoxicity-related inflammation can be explained by increased oxidative stress in hepatocytes [29]. The progression of hepatic steatosis and T2DM is collectively influenced by overall glucotoxicity, insulin resistance, and chronic inflammation, which supports the newly defined terminology MAFLD.
High-fat diet feeding in both humans and rodents develops hepatic steatosis within a few days, as well as a diet enriched in saturated fat or elevated intrahepatic triglyceride content in overweight males [30,31,32]. These results confirm that lipotoxicity plays a significant role in NAFLD. In obese individuals, nearly 60% of hepatic triglycerides are derived from free fatty acids (FFAs) [33]. Increasing visceral adipose tissue in obese people may lead to insulin resistance and hyperinsulinemia, which promotes lipolysis of adipose tissue [33]. Two classes of lipids, diacylglycerol (DAG) and ceramide, play essential roles in causing hepatic IR [34]. Lipid metabolites function as intermediates in the causal link between lipotoxicity and IR and both contribute to the development of NAFLD/MAFLD.

3.2. Metabolic Conditions

The risk factors for developing NAFLD and NASH are metabolic disorders such as diabetes and obesity, in addition to age, sex, and race/ethnicity [35]. Diabetic patients with insulin resistance and obesity have enhanced lipolysis and increased free fatty acid transport to hepatocytes [36]. The majority of NAFLD patients with normal weights, referred to as “Lean NAFLD,” are sedentary and have poor insulin sensitivity, making them more prone to cardiovascular disease. These are the result of an increase in hepatic de novo lipogenesis and a decrease in the ability of adipose tissue to store fat [10,17]. Secondary causes of NAFLD include inborn errors of metabolism, iatrogenic causes (diseases caused by medical treatment or examination), viral hepatitis, and nutritional disorders. These factors are less well recognized but are nevertheless significant. Many etiological factors, including alcohol abuse, obesity, and diabetes mellitus, contribute to fatty liver changes.

3.3. Gut Microbes

Furthermore, gut microorganisms contribute to the development of NAFLD [37]. Dysbiosis of the gut microbiota affects the risk of NAFLD in various ways [38]. Owing to the increased permeability of the intestine, the liver may be exposed to harmful substances, such as translocated bacterial toxins and inflammatory chemical signals that promote hepatic inflammation. These alterations enhance nutrient and calorie absorption and alter metabolism [39,40,41]. Excessive consumption of micronutrients disturbs gut homeostasis and causes gut inflammation [42]. The various etiological factors responsible for the development of NAFLD and MAFLD are depicted in Figure 2.

4. Diagnosis

The diagnosis of the disease is based on the medical history of the patient, including dietary habits. The selection of a diagnostic test depends on cost and sensitivity. The suggested MAFLD diagnostic criteria are predicated on the presence of hepatic steatosis, which can be identified using imaging methods, blood biomarkers, or liver histology [43].

4.1. Imaging Techniques

NAFLD is an asymptomatic disease that is frequently detected using abdominal ultrasound, CT scans, or magnetic resonance imaging (MRI) and can show hepatic fatty infiltration [44]. The limitation of this technique is that its sensitivity decreases when the steatosis level is lower than 30% [45,46].
Owing to its accessibility and low cost, ultrasound is favored as the first-line examination [47]. The limitations of the test are as follows: (1) it is unable to distinguish between steatosis and fibrosis, and (2) it is not sensitive if the liver’s fat content is less than 20%. Alternatively, the controlled attenuation parameter (CAP) is more sensitive, and 1H-MRS is also an acceptable quantitative marker of steatosis. Laboratory biochemical testing is also appropriate for high-risk populations [47]. For all patients with metabolic risk factors, the EASL and AASLD recommend performing an ultrasound examination and measuring liver enzymes.
In clinical practice, ultrasonography is widely used because of its convenience, cost-effectiveness, and sensitivity when the fat content is >30% in the liver [48,49].

4.2. Computerized Tomography (CT)

This diagnostic technique is more precise than ultrasonography (US), but it is limited to patients with mild steatosis and radiation exposure [50]. CT is more sensitive than US in assessing hepatic fat content, but substances such as iron may act as confounders and affect the diagnosis [51]. The CAP Technique is an ultrasound-based method for the quantification of steatosis (>10% steatosis). Although the test does not seem to be very dependable [52], the Asia-Pacific guidelines still recommend CAP as a useful tool in NAFLD patients.
Although magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) can detect hepatic steatosis and fibrosis, their use in clinical practice is constrained by high cost [53,54]. Because of the test’s operational complexity and complex algorithm, MRS use in routine clinical practice is limited despite its ability to identify 5.56% of the liver fat content, which makes it the gold standard for steatosis [54].
More recently, transient elastography (TE FibroScan), performed using an ultrasound procedure, has gained popularity because it provides quick and convenient measurements of liver stiffness, which are closely linked to the stage of liver fibrosis [55]. The sensitivity and specificity of CK18-based NAFLD diagnosis are 66% and 82%, respectively [14].

4.3. Blood and Serum Biomarkers

Biochemical parameters of NAFLD in patients with metabolic syndrome including alanine aminotransferase (ALT), aspartate aminotransferase (AST), hemoglobin A1C, homeostatic model assessment (HOMA)-IR, triglycerides, BMI, and gamma-glutamyl transferase (GGT) could provide good prediction of disease condition [56,57,58].
The insulin-like growth factor1 (GF-1) and cytokeratin18 fragment (CK18F) have also demonstrated encouraging results in routine NAFLD screening [59]. Another diagnostic blood miRNA test is highly effective in detecting NASH in obese individuals [60]. It is crucial to identify the disease at an early stage using non-invasive diagnostic methods so that the proper course of action can be taken to prevent the disease from progressing from an early stage of simple steatosis to steatohepatitis.

4.4. Histological Biopsy

Liver biopsy remains the gold standard for determining the presence of NAFLD and, more specifically, for determining the presence of NASH at an advanced stage [61]. Liver biopsy is an invasive procedure that carries a 0.05% risk of mortality due to test-related complications [43]. Additionally, the test is expensive and requires trained pathologists and skilled operators [62]. Furthermore, this technique has limitations for mass screening (European Assoc 2015). The sampling error that occurs during biopsy can result in an erroneous prognosis. Non-invasive methods may be preferred as the first strategy to identify NAFLD/MAFLD because inter-observer variability can result in over- or underestimation of the hepatic fibrosis stage [63]. If NASH and liver fibrosis cannot be determined using non-invasive methods, additional liver biopsy is only performed on individuals in whom the etiology of the liver needs to be clarified [64].

5. Pharmacological Therapeutics Evaluation

Pharmacological therapeutics have been developed for MAFLD based on the mechanisms of action and adverse events. The Liver International Society in the Americas (AASLD) [9,14,15] made recommendations according to the clinical practice guidelines used by these agents.
Medications based on potential benefits include vitamin E, which is recommended for non-diabetic, non-cirrhotic patients, and pioglitazone with or without diabetes, while no clear benefit was observed using statins for cardiovascular disease; similarly, metformin, n-3polyunsaturated fatty acids, ursodeoxycholic acid, and pentoxifylline showed no clear benefit in MAFLD.
Currently, drugs with potential benefits include thiazolidinediones (pioglitazone) and Vitamin E’ in MAFLD. Pioglitazone has been proven to be effective in diabetic and MAFLD patients with substantial fibrosis (>F2), while vitamin E is advised for nondiabetic MAFLD patients with >F2 non-cirrhosis [65]. There have been studies on new pharmacological agents targeting various MAFLD patients [66]. Eight new drug classes with different targets are described here, owing to the disputed pathophysiology of MAFLD. They are first- and second-generation farnesoid X receptors, peroxisome proliferator activated receptor (PPAR), C-c chemokine receptor), the GLP-1 (glucagon-like peptide-1) receptor, and thyroid hormone receptor (TSH).

5.1. Farnesoid X Receptor Agonist

The first generation include obeticholic acid (OCA)(INT-747), whereas the second generation included Cilofexor (GS-9674) and Tropifexor. New pharmacological therapeutics, such as FXR, are targets for metabolic dysregulation caused by obesity, including type 2 diabetes, NAFLD, and atherosclerosis. The nuclear receptor family, which includes the farnesoid X receptor (FXR), is activated by bile acids and expressed in the liver. The effective absorption of fat and fat-soluble vitamins is facilitated by bile acids, which also aid in digestion and play a significant role in controlling inflammation and the cholesterol and triglyceride homeostasis. Bile acids regulate hepatic lipid and glucose metabolism [67]. Inhibiting lipogenesis, gluconeogenesis, and the regulation of insulin sensitivity are inhibited by the FXR receptor, which is activated by bile acids [68,69].
OCA is a semi-synthetic bile acid analogue. The chemical structure of OCA is 6-alpha-ethyl-chenodeoxycholin acid, which has been developed and commercialized under the brand name Ocaliva, in combination with ursodeoxycholic acid used to treat primary biliary cholangitis. Intercept Pharmaceuticals Inc. holds the right to develop this drug outside of Japan and China. OCA was the first FXR agonist used in human drug studies. USFDA approved it in October 2016 for the treatment of primary biliary cholangitis. OCA was proposed to treat NASH [70] and reduced markers of liver inflammation, fibrosis, and increased insulin sensitivity [71]. The long-term safety issues of increased cholesterol and adverse cardiovascular events may warrant the concomitant use of statins in OCA treatment. Furthermore, the Intercept Pharma Govt Scientist FDA label updated in 2018 warns of liver injury from improper Ocaliva dosing.
The most frequent adverse effect, mild to moderate pruritus, was recorded, which led to discontinuation of OCA. Currently, OCA is not approved by the US Food and Drug Administration for the treatment of MAFLD.

5.2. Second Generation: Cilofexor (GS-9674), Tropifexor (LJN 452)

Cilofexor, a non-steroid FXR agonist, primarily activates intestinal FXR without affecting enterohepatic circulation. Cilofexor decreased portal hypertension and reduced liver fibrosis in NASH rats [72]. In a recent study, hepatic steatosis, as determined by magnetic resonance imaging, protein density fat fraction, and blood gamma-glutamyltransferase levels were significantly reduced in NASH patients. However, there were no discernible alterations in the lipid profiles. Side effects such as mild-to-severe pruritus have also been observed [73]. Cilofexor is being developed by Gilead Sciences and showed improvement in NAFLD and NASH as seen by reduction in fibrosis and steatosis, improved cholestasis, and reduced markers of liver injury [73,74].
Another second-generation potent oral FXR agonist, tropifexor, which is structurally unrelated to bile acids, has shown significant efficacy in reducing hepatic steatosis. This investigational drug was discovered by researchers from Novartis and the Genomix Institute of the Novartis Research Foundation. The interim analysis of Phase 2-part C revealed a drop in ALT as well as a decrease in body weight and hepatic fat. Significant side effects of pruritus and elevated blood LDL-cholesterol are still the most frequent adverse events leading to tropifexor discontinuation [75]. A combination therapy of tropifexor and cenicriviroc (an antiretroviral agent) showed improvement in liver fibrosis in NASH patients [76,77].

5.3. Peroxisome Proliferator-Activated Receptor (PPAR) Agonists

PPAR agonists act on peroxisome proliferator-activated receptors. PPAR agonists are classified into three groups: (1) PPAR alpha/or gamma elafibranor (GET 505), (2) Pan- PPAR agonist (PPAR-alpha, PPAR-beta/gamma, and PPAR-gamma) lanifibranor (IVA 337), and (3) dual agonist of PPAR-alpha/gamma saroglitazar. PPAR transcription factors, particularly those that control lipid and glucose metabolism are involved in controlling energy balance as well [78]. PPAR-gamma activation enhances insulin sensitivity and plays a role in fat storage, whereas PPAR-alpha activation lowers plasma triglyceride levels. By suppressing the inflammatory macrophage phenotype, the activation of PPAR-beta/gamma improves fatty acid metabolism and exhibits anti-inflammatory effects [79].
Elafibranor (GFT505) is a dual PPAR alpha-PPAR delta agonist investigational drug developed by Genfit for the treatment of cardiometabolic diseases such as diabetes, insulin resistance, dyslipidemia, and non-alcoholic fatty liver disease [80]. Elafibranor (GFT 505) showed lower plasma triglyceride levels and increased HDL cholesterol in prediabetic individuals. Currently, there are no indications of PPARy-related adverse effects, which is a comforting safety profile. Elafibranor is a top contender for the treatment of NAFLD because of its effects on insulin sensitization and hepatoprotection. Elafibranor failed to sufficiently enhance histological NASH endpoints in a phase 3 trial (NCT 02704403). However, several individuals exhibited renal impairment and elevated serum creatinine levels during treatment, raising concerns about its safety.
Lanifibranor (IVA 337) is a pan-PPAR agonist of PPAR-alpha, PPAR-beta/delta, and PPAR-gamma that has both antifibrotic and positive metabolic effects [81]. In terms of lowering liver fibrosis and inflammatory gene expression, the pan-PPAR agonist lanifibranor is more effective than single or dual PPAR agonists [81]. In NASH patients, lanifibranor was well tolerated and reduced steatosis and fibrosis by at least two points without worsening fibrosis. Based on these encouraging findings, the FDA designated lanifibranor as a “breakthrough treatment” for a phase 3 trial targeting NASH patients. Lanifibranor is the first drug candidate to achieve statistically significant results at the main endpoints, and the USA FDA and European Medicine Agency (EMA) authorized lanifibranor for a phase III trial in the future.
Another PPAR alpha/gamma dual agonist compound, saroglitazar, is marketed under the trade name Lipaglyn for the treatment of type 2 diabetes mellitus, dyslipidemia, and hypertriglyceridemia associated with type 2 diabetes mellitus, which is not controlled by statin therapy. In clinical research, saroglitazar has been demonstrated to decrease triglycerides, LDL, VLDL, non-HDL, and increase HDL cholesterol. Additionally, it has demonstrated anti-diabetic effects by lowering fasting blood sugar and HB A1c in diabetic patients and might be useful in diabetic nephropathy [82]. Saroglitazar exhibits positive effects by reducing hepatic steatosis in NASH patients and by enhancing lipid and glycemic profiles in diabetic people with dyslipidemia [83]. However, no significant difference in liver stiffness measurements was reported [84,85]. No major serious adverse events have been reported; however, long-term cardiovascular safety has not been established [82]. Saroglitazar (Lipaglyn), a dual PPAR alpha/gamma agonist, has been approved for the treatment of NASH. This is the first medication used to treat NASH worldwide and has received approval from the Drug Controller General of India (DCGI). Saroglitazar lowers the hepatic fat content and ALT levels in obese patients with NASH [85].
Thiazolidinedione (TZD) is a peroxisome proliferator-activated receptor gamma agonist. Insulin sensitizers are also known in the management of type 2 diabetes mellitus (T2DM). Thiazolidinediones mostly promote adipocyte differentiation, lower plasma FFA, decrease hepatic fat deposition, and improve insulin resistance [86]. Pioglitazone significantly reduced hepatic fat content in T2DM patients with NASH; however, vitamin E was effective in NASH patients without diabetes [87].

5.4. Glucagaon-like Peptide-I (GLP-1) Agonists

Current diabetes and obesity treatments include GLP-1 analogs because of their beneficial metabolic effects, particularly on glucose metabolism. MAFLD development is intimately correlated with the GLP-1 signaling pathway [88]. GLP-1 receptor agonist (GLP-1-RA) is effective in improving cardiovascular and T2DM outcomes [89].
This class contained three subclasses: (1) Liraglutide and semaglutide, which function as GLP1 receptor agonists; (2) Tirazapatide, a dual agnostic GLP-1 receptor and glucose-dependent insulinotropic polypeptide (GIP) receptor (LY3298176); (3) dual glucagon and GLP-1 receptor agonist Cotadutide (MEDI0382). The incretin hormone glucagon-like peptide (GLP-1) is released after food intake and stimulates insulin secretion along with hyperglycemia affecting weight loss [90].
Liraglutide (a-GLP-1 agonist), is marketed under the trade name Victoza and is used to treat type 2 diabetes, obesity, and chronic weight management. Victoza is recommended for type 2 diabetes, in addition to diet and exercise, to enhance glycemic control and lower the risk of serious adverse cardiovascular events. Long-term use involves the risk of thyroid C-cell tumors, pancreatitis, hypoglycemia, renal impairment, hypersensitivity reactions, and acute gallbladder disease. Common side effects include low blood sugar, nausea, dizziness, abdominal pain, pain at the site of injection, gastrointestinal side effects, medullary thyroid cancer, angioedema, pancreatitis, gallbladder disease, and kidney problem. This drug was approved in the United Sates in 2010 with issued warning of adverse effects that include thyroid cancer and pancreatitis.
Liraglutide in patients with NASH was assessed for safety and efficacy and was found to be effective considering histological resolution, safe, well tolerated, and warranting extensive long-term studies [91]. Liraglutide treatment helps in reducing weight loss, and decreased serum cholesterol and triglyceride levels, and hepatic fat content in T2DM patients with MAFLD; in a pilot 48-week treatment, it was able to stop the progression of liver fibrosis [91,92,93].
Semaglutide, another GLP-1 receptor agonist, boosts insulin secretion, controls blood sugar, is effective for weight loss, and reduces liver enzymes [94]. This GLP-1RAS shared common transient adverse effects with gastrointestinal symptoms, including nausea, vomiting, and abdominal pain. The probability of long-term injection may increase the risk of pancreatitis, pancreatic cancer, thyroid cancer, biliary disease, kidney injury, and retinopathy; however, no definite conclusion has been drawn [95]. Semaglutide improves insulin sensitivity and results in long-lasting weight loss in diabetic patients, whereas individuals with NASH in Phase 2 had considerable resolution but no improvement in the fibrosis stage [96].
Semaglutide is clinically effective in overweight or obese people for weight loss, to be used together with diet and exercise for weight loss at 3 and 6 months [97]. It is also effective in type 2 diabetes and lowers the risk of heart attack, stroke, or death in patients. Semaglutide treatment of overweight and obesity at a dose of 2.4 mg subcutaneously once a week consistently reduced the mean weight loss by 14.9%–17.4% in participants without diabetes and showed improvement in cardiometabolic risk factors, physical function, and quality of life [98]. Nova Nordisk Pharmaceutical Company developed semaglutide in both injectable formulations (Ozempic and Wegovy approved in 2017 in the US) and oral formulations (Rybelsus, approved in 2019 in the US).
Tirzapatide (Ly3298176, Eli Lilly and Company) is a dual GIP and GIP-1 agonist and a new therapeutic option for the treatment of type 2 diabetes. Side effects include tiredness, loss of appetite, nausea, vomiting, diarrhea, and abdominal pain. In comparison with dulaglutide, another GLP-1 agonist, a phase II RCT demonstrated improved glycemic control and weight loss with a favorable tolerability and safety profile [99]. In a phase IIb study, the efficacy and safety of tirzapatide in patients with NASH are still being evaluated (NCT04166773). Tirzapatide is prescribed subcutaneously once per week to treat type 2 diabetes and is sold under the brand name Mounjiaro. Swelling/redness/itching were also observed at the injection site. This drug was manufactured by Eli Lilly and was approved in 2022.
Cotadutide (MED 10382) is a dual glucagon and GLP-1 agonist that is effective against overweight T2DM in reducing weight and serum transaminase compared to placebo (NCT 03235050) in a phase IIb RCT [100]. By imitating the human hormones, glucagon-like peptide 1 and glucagon, which participate in blood sugar regulation, reduce blood glucose levels. This medication is a peptide injected under the skin. As of February 2021, this medication began phase II clinical trials. This investigational drug is under development for NASH and chronic kidney disease with type 2 diabetes.

5.5. Thyroid Hormone Receptor Beta Agonist: Resmetirom (MGL-3196)

The thyroid hormone receptor beta (THR-beta) agonist resmetirom (MGL-3196) is very selective. It is a liver-targeted drug with strong affinity for thyroid hormone receptor-beta, which is intended to reduce lipotoxicity and ameliorate NASH by accelerating hepatic fat metabolism. The drug was developed by Madrigal Pharmaceuticals (West Conshohocken, PA, USA). Resmetirom is a prospective lipid-lowering medication that also shows promise in the treatment of non-alcoholic fatty liver disease and hepatic insulin resistance. It is a liver-specific agonist of thyroid hormone receptor beta (TRBeta). TRBeta controls the metabolic pathway, which lowers triglyceride and cholesterol levels, improves insulin sensitivity, promotes liver fat metabolism, and decreases cell apoptosis. Resmetirom demonstrated efficacy in phase IIb NASH by reducing hepatic steatosis [101], and its efficacy and safety have been studied in phase III RCT (MAESTRO NASH) in stage 2–3 fibrosis NASH patients (NCT 03900429). Resmetirom was well tolerated, but had the most common gastrointestinal adverse events, such as diarrhea and nausea.

5.6. C-C Chemokine Receptor Type 2 (CCR2) and Type 5 (CCR5) Antagonist

Cenicriviroc is a dual CCR2/CCR5 antagonist. Although CCR5 promotes the proliferation of collagen-producing activated hepatic stellate cells/myofibroblasts and is linked to the advancement of fibrosis, CCR2 is crucial for the recruitment and activation of monocytes and macrophages at sites of hepatic damage. Cenicriviroc, however, was not very effective in treating NASH and required further validation in a phase3 trial [102], while in the phase III RCT of cenicriviroc in patients with NASH it lacked efficacy to treat liver fibrosis; further development was stopped due to lack of benefit [103]. Hence, at present, there are no approved drugs for patients with NASH from this family. Cenicriviroc was developed by Takeda and Tobira Therapeutics, with code numbers TAK-652 and TBR-652, respectively.

5.7. Antifibrotic Drugs

Selonsertib (GS-4997) is a selective inhibitor of apoptosis signal-regulating kinase1(ASK1). The treatment of NASH patients with this compound increases the risk of fibrosis, scarring, cirrhosis, liver failure, and cancer. In conclusion, selonsertib does not reduce fibrosis in NASH patients [104].
Simtuzumab (GS-6624) is a humanized monoclonal antibody designed to treat fibrosis. It acts as an immunomodulator by binding to enzyme lysyl oxidase-like2 (LOXL2). This product was developed by Gilead Sciences; however, in the phase 2 clinical study, it was terminated in January 2016 due to idiopathic pulmonary fibrosis and lack of efficacy. (Gilead Press release 5 January 2016). In the phase IIb trial of NASH patients, simtuzumab was ineffective in decreasing hepatic collagen content or hepatic venous pressure gradients (Clinical trial Govt NCT 01672866 and NCTO 1672879) [105].

5.8. Pancaspase Inhibitor

Caspase inhibitors are hepatoprotective drugs, and Idun Pharmaceuticals developed the prospective medication emricasan in 1998 for the treatment of human hepatic disease [106]. Emricasan (IDN-6556, PF-03491390) is an oral irreversible pan-caspase inhibitor and was found to be well tolerated in a phase II RCT but ineffective in reducing fibrosis in NASH patients with fibrosis stages 1–3 [107,108].

5.9. Natural Plant Drugs or Food Products

Natural plant medicine curcumin has anti-inflammatory, antioxidant, and lipid-modifying characteristics that may be helpful for MAFLD. Although a meta-analysis of RCTs demonstrated that curcumin improved the pathology of the liver and had a positive effect on lipid profiles, it failed to achieve statistical significance [109]. Functional foods or natural health products could be useful in the direction of decrease of obesity metabolic consequences and weight management [110]. However, scientific evidence is needed to support with clinical studies. Curcumin, silymarin, an extract from milk thistle seeds, resveratrol may benefit metabolic health [111,112] but natural plant drugs are inferior to metabolic targeted drugs for NAFLD [109].
Combination therapy of novel monotherapies targeting different mechanistic approaches is an attractive approach for investigation. Currently, FXR agonists are the most favorable agents and may be worth trying in combination, which will need additional clinical data in the evaluation of benefits in NASH as well as safety profiles.

5.10. Anti-Obesity Drugs Evaluated for MAFLD

Anti-obesity medications have also been evaluated for MAFLD. The following medications are currently licensed for the treatment of obesity in the United States and Europe: orlistat, naltrexone/bupropion, liraglutide, and semaglutide [113]. Orlistat did not alter liver histology in MAFLD patients, and further research is needed to determine its precise efficacy [114]. Similarly, the effectiveness of naltrexone may be associated with weight loss and may improve the liver health whereas other anti-obesity drugs for MAFLD are unknown and further research is required [115].

5.11. Anti-Hyperglycemic Agents for MAFLD

Two diseases, T2DM and MAFLD, frequently coexist and increase the risk of disease worsening [116,117]. Metformin and dipeptidyl peptide 4 (DPP4) inhibitors failed to demonstrate substantial efficacy in hepatic steatosis and fibrosis, whereas glucagon-like peptide-1 receptor agonists (GLP-1, RA), thiazolidinediones, and sodium-dependent glucose transporter 2 (SGLT2) inhibitors showed some improvement in the histological features of steatohepatitis.

5.12. Sodium–Glucose Cotransporter-2 (2SGLT-2) inhibitors

The use of sodium–glucose cotransporter-2 (2SGLT-2) inhibitors in T2DM patients at risk of cardiovascular disease is currently common [118]. For T2DM patients with MAFLD, other inhibitors in this group, including luseogliflozin, canagliflozin, ipragliflozin, and empagliflozin, all demonstrated improvements in liver fat content, ranging from 3.9% to 6.9% and reduced body fat and abdominal fat area and fibrosis regression [119,120,121,122].
Canagliflozin belongs to a class of SGLT2 inhibitors used in type 2 diabetes along with diet and exercise, or sometimes with other medications, to lower blood sugar levels. Canagliflozin reduces the risk of stroke and heart attack in patients with diabetes, as well as the risk of severe kidney disease. This medication is not used for the treatment of type 1 diabetes. Canagliflozin was developed for NASH in NASH activity for commercial preparation under the brand name Ivokana. Side effects include dizziness, lightheadedness, fainting when getting up, several urinary-related problems, and sometimes very serious ketoacidosis. The FDA is concerned about the cardiovascular safety of this drug.
Empagliflozin is also an SGLT2 inhibitor developed under the brand name Jardiance for type 2 diabetes that lowers the risk of cardiovascular death and hospitalization for heart failure. Jardiance reduces the hepatic fat content. This drug is not recommended for people with type1 diabetes and those with severe kidney problems. This medication can cause serious side effects such as ketoacidosis and urinary tract infection.

5.13. Metformin and Dipeptidyl Peptidase 4 Inhibitors

Metformin is well established drug for T2DM and helps in reducing obesity and improving liver dysfunctions [123]. Evidence for the use of metformin in reducing liver fat accumulation in MAFLD patients is needed to support clinical studies [124].

5.14. Fibroblast Growth Factors

Aldafermin is an engineered analog of fibroblast growth factor 19 (FGF 19 analog) (formerly NGM 282), and in a phase 2 trial, aldafermin therapy decreased the amount of liver fat in NASH patients as determined by MRI-PDFF and indicated a trend toward fibrosis improvement [125]. Aldafermin was well tolerated but had uncertainty over the efficacy of treatment and risk-benefit profile; hence, it could not be approved. The development of aldafermin was funded by NGM Biopharmaceuticals.
Efruxifermin (EFX) was developed to reverse fibrosis, reduce hepatic fat and inflammation, increase insulin sensitivity, and improve lipoprotein levels. It has been shown to dramatically decrease liver fat in patients with NASH and F1–F3 fibrosis, while being safe and well tolerated. After 16 weeks of treatment, the medication was successful in lowering the hepatic fat fraction liver injury marker level and improving the histology and metabolic criteria, indicating that efruxifermin may have the ability to alter the progression of NASH. Efruxifermin should be further examined in long-term studies with larger patient populations to be developed as a first-line therapy for NASH [126]. EFX was designed for convenient subcutaneous dosing once per week. This product is being developed by Akero Therapeutics and US FDA has recognized the potential of this medication and approved it for clinical research studies.
Both efruxifermin and pegbelfermin (formerly BMS* -986036*Bristol-Myers-squib) are both FGF21 analogues developed as injectable preparations for subcutaneous administration once per week for 16 weeks. Pegbelfermin is an investigational PEGylated fibroblast growth factor 21 analog that has been developed for the treatment of NASH. It has also been shown to improve liver damage and cardiometabolic parameters. Pegbelfermin treatment was well tolerated and significantly reduced the hepatic fat fraction in patients with non-alcoholic steatohepatitis. Further studies should be conducted in a larger number of patients with NASH using liver biopsies and their effects on liver histology to demonstrate reduced progression to cirrhosis. Most adverse events, observed such as nausea and diarrhea, are mild [127,128].
While efruxifermin showed benefits in serum lipid metabolism and glycemic management, as well as weight loss, pegbelfermin exhibited improvements in liver damage indicators and cardiometabolic parameters. To evaluate the effectiveness of histological endpoints, additional studies with efruxifermin in patients with NASH (NCT 04767529) and NASH-related compensated cirrhosis (NCT 05039450) are currently underway. Further studies with pegbelfermin should be carried out to demonstrate reduced progression to cirrhosis.
The pharmacological agents developed for MAFLD treatment, and their current status are summarized in Table 1.
Currently, there are no FDA-approved drugs for the treatment of fatty liver disease. The two best options affirmed by the American Association for the Study of Liver Diseases for biopsy-proven NASH are vitamin E (an antioxidant) and pioglitazone (used to treat diabetics). Amongst several pharmaceuticals researched, only a few molecules such as obeticholic acid, saroglitazar, liraglultide, semaglutide, tirzapatde, canagliflozin, and empagliflozin have been developed and are commercially available for the treatment at phases of NAFLD and or its associated disease MAFLD. The serious adverse effects of these drugs are taken into consideration during treatment.
In conclusion, regardless of the current or future progress in pharmacotherapy, a healthy lifestyle, good food habits, and weight loss remain the primary goal of MAFLD treatment. Second, (2) CPGs suggest utilizing pioglitazone and vitamin E in patients with considerable fibrosis (≥F2 fibrosis), whether they have T2DM or not, as an alternative strategy. Third, (3) many innovative therapeutics that target various pathogenetic pathways and the admixture of several types of targeted pharmacotherapies are now being researched to develop effective disease-treating drugs for individuals with MAFLD, NAFLD, and NASH.

6. Future Research Directions

1. Search for safe and effective novel therapeutic agents for MAFLD remain a priority for biomedical research in the near future.
2. New strategies combining multitarget drugs need to be studied.
3. Simple and reliable noninvasive diagnostic tools for disease identification are required.
4. The pathogenesis of the disease is not very well understood; therefore, research work is needed to explore the role of glucotoxicity, lipotoxicity, and antioxidant defense through diet.
5. Development of suitable animal models to explore the various aspects of the disease process and the evaluation of new therapeutic agents.

Author Contributions

K.K.B. wrote the manuscript. M.P.S. generated the graphic and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Authors acknowledge Bhupendra S. Kaphalia for his valuable comments and suggestions given for this manuscript. The figures presented in this manuscript were created using Biorender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yki-Jarvinen, H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome. Lancet Diabetes Endocrinol. 2014, 2, 901–910. [Google Scholar]
  2. Schaffner, F.; Thaler, H. Nonalcoholic fatty liver disease. Prog. Liver Dis. 1986, 8, 283–298. [Google Scholar] [PubMed]
  3. Huang, T.D.; Behary, J.; Zekry, A. Non-alcoholic fatty liver disease: A review of epidemiology, risk factors, diagnosis and management. Intern. Med. J. 2020, 50, 1038–1047. [Google Scholar] [PubMed]
  4. Eslam, M.; Sanyal, A.J.; George, J.; Consensus, P.I. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014. [Google Scholar]
  5. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wong, V.W.-S.; Dufour, J.F.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [PubMed]
  6. Lin, S.; Huang, J.; Wang, M.; Kumar, R.; Liu, Y.; Liu, S.; Wu, Y.; Wang, X.; Zhu, Y. Comparison of MAFLD and NAFLD diagnostic criteria in real world. Liver Int. 2020, 40, 2082–2089. [Google Scholar] [PubMed]
  7. Feng, G.; Byrne, C.D.; Targher, G.; Wang, F.; Zheng, M.H. Ferroptosis and metabolic dysfunction-associated fatty liver disease: Is there a link? Liver Int. 2022, 42, 1496–1502. [Google Scholar]
  8. Huang, Q.; Zou, X.; Wen, X.; Zhou, X.; Ji, L. NAFLD or MAFLD: Which Has Closer Association with All-Cause and Cause-Specific Mortality?—Results From NHANES III. Front. Med. 2021, 8, 693507. [Google Scholar]
  9. Eslam, M.; Sarin, S.K.; Wong, V.W.; Fan, J.G.; Kawaguchi, T.; Ahn, S.H.; Zheng, M.H.; Shiha, G.; Yilmaz, Y.; Gani, R.; et al. The Asian Pacific Association for the Study of the Liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. Hepatol. Int. 2020, 14, 889–919. [Google Scholar]
  10. Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [Google Scholar]
  11. Zheng, K.I.; Fan, J.G.; Shi, J.P.; Wong, V.W.; Eslam, M.; George, J.; Zheng, M.H. From NAFLD to MAFLD: A “redefining” moment for fatty liver disease. Chin. Med. J. 2020, 133, 2271–2273. [Google Scholar] [CrossRef]
  12. Younossi, Z.M.; Golabi, P.; de Avila, L.; Paik, J.M.; Srishord, M.; Fukui, N.; Qiu, Y.; Burns, L.; Afendy, A.; Nader, F. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J. Hepatol. 2019, 71, 793–801. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, J. Estimating global prevalence of metabolic dysfunction associated fatty liver disease in overweight or obese adults. Clin. Gastro Hepatol. 2022, 20, e573–e582. [Google Scholar] [CrossRef] [PubMed]
  14. European Association for the Study of The Liverl; European Association for the Study of Diabetes. EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 2016, 64, 1388–1402. [Google Scholar] [CrossRef]
  15. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef]
  16. Medina, J.; Fernandez-Salazar, L.I.; Garcia-Buey, L.; Moreno-Otero, R. Approach to the pathogenesis and treatment of nonalcoholic steatohepatitis. Diabetes Care 2004, 27, 2057–2066. [Google Scholar] [PubMed]
  17. Wong, V.W.; Chan, W.K.; Chitturi, S.; Chawla, Y.; Dan, Y.Y.; Duseja, A.; Fan, J.; Goh, K.L.; Hamaguchi, M.; Hashimoto, E.; et al. Asia-Pacific Working Party on Non-alcoholic Fatty Liver Disease guidelines 2017-Part 1: Definition, risk factors and assessment. J. Gastroenterol. Hepatol. 2018, 33, 70–85. [Google Scholar] [CrossRef]
  18. Diehl, A.M.; Day, C. Cause, Pathogenesis, and Treatment of Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2017, 377, 2063–2072. [Google Scholar] [CrossRef]
  19. Zhu, B.; Chan, S.L.; Li, J.; Li, K.; Wu, H.; Cui, K.; Chen, H. Non-alcoholic Steatohepatitis Pathogenesis, Diagnosis, and Treatment. Front. Cardiovasc. Med. 2021, 8, 742382. [Google Scholar]
  20. Lee, K.C.; Wu, P.S.; Lin, H.C. Pathogenesis and treatment of non-alcoholic steatohepatitis and its fibrosis. Clin. Mol. Hepatol. 2023, 29, 77–98. [Google Scholar] [CrossRef]
  21. Stefan, N.; Cusi, K. A global view of the interplay between non-alcoholic fatty liver disease and diabetes. Lancet Diabetes Endocrinol. 2022, 10, 284–296. [Google Scholar] [CrossRef] [PubMed]
  22. Powell, E.E.; Wong, V.W.; Rinella, M. Non-alcoholic fatty liver disease. Lancet 2021, 397, 2212–2224. [Google Scholar] [CrossRef]
  23. Wree, A.; Broderick, L.; Canbay, A.; Hoffman, H.M.; Feldstein, A.E. From NAFLD to NASH to cirrhosis-new insights into disease mechanisms. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 627–636. [Google Scholar] [CrossRef] [PubMed]
  24. Lim, J.S.; Mietus-Snyder, M.; Valente, A.; Schwarz, J.M.; Lustig, R.H. The role of fructose in the pathogenesis of NAFLD and the metabolic syndrome. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 251–264. [Google Scholar] [CrossRef]
  25. Leermakers, E.T.; Moreira, E.M.; Jong, J.C.K.-D.; Darweesh, S.K.; Visser, T.; Voortman, T.; Bautista, P.K.; Chowdhury, R.; Gorman, D.; Bramer, W.M.; et al. Effects of choline on health across the life course: A systematic review. Nutr. Rev. 2015, 73, 500–522. [Google Scholar] [CrossRef] [PubMed]
  26. Jensen, T.; Abdelmalek, M.F.; Sullivan, S.; Nadeau, K.J.; Green, M.; Roncal, C.; Nakagawa, T.; Kuwabara, M.; Sato, Y.; Kang, D.H.; et al. Fructose and sugar: A major mediator of non-alcoholic fatty liver disease. J. Hepatol. 2018, 68, 1063–1075. [Google Scholar] [CrossRef]
  27. Hashiba, M.; Ono, M.; Hyogo, H.; Ikeda, Y.; Masuda, K.; Yoshioka, R.; Ishikawa, Y.; Nagata, Y.; Munekage, K.; Ochi, T.; et al. Glycemic variability is an independent predictive factor for development of hepatic fibrosis in nonalcoholic fatty liver disease. PLoS ONE 2013, 8, e76161. [Google Scholar] [CrossRef]
  28. Wu, H.; Ballantyne, C.M. Metabolic Inflammation and Insulin Resistance in Obesity. Circ. Res. 2020, 126, 1549–1564. [Google Scholar] [CrossRef]
  29. Salvado, L.; Palomer, X.; Barroso, E.; Vazquez-Carrera, M. Targeting endoplasmic reticulum stress in insulin resistance. Trends Endocrinol. Metab. TEM 2015, 26, 438–448. [Google Scholar] [CrossRef]
  30. Parry, S.A.; Rosqvist, F.; Mozes, F.E.; Cornfield, T.; Hutchinson, M.; Piche, M.E.; Hulsmeier, A.J.; Hornemann, T.; Dyson, P.; Hodson, L. Intrahepatic Fat and Postprandial Glycemia Increase After Consumption of a Diet Enriched in Saturated Fat Compared with Free Sugars. Diabetes Care 2020, 43, 1134–1141. [Google Scholar] [CrossRef]
  31. Turner, N.; Kowalski, G.M.; Leslie, S.J.; Risis, S.; Yang, C.; Lee-Young, R.S.; Babb, J.R.; Meikle, P.J.; Lancaster, G.I.; Henstridge, D.C.; et al. Distinct patterns of tissue-specific lipid accumulation during the induction of insulin resistance in mice by high-fat feeding. Diabetologia 2013, 56, 1638–1648. [Google Scholar] [CrossRef] [PubMed]
  32. Lindeboom, L.; Nabuurs, C.I.; Hesselink, M.K.; Wildberger, J.E.; Schrauwen, P.; Schrauwen-Hinderling, V.B. Proton magnetic resonance spectroscopy reveals increased hepatic lipid content after a single high-fat meal with no additional modulation by added protein. Am. J. Clin. Nutr. 2015, 101, 65–71. [Google Scholar] [CrossRef] [PubMed]
  33. Donnelly, K.L.; Smith, C.I.; Schwarzenberg, S.J.; Jessurun, J.; Boldt, M.D.; Parks, E.J. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Investig. 2005, 115, 1343–1351. [Google Scholar] [CrossRef]
  34. Petersen, M.C.; Shulman, G.I. Roles of Diacylglycerols and Ceramides in Hepatic Insulin Resistance. Trends Pharmacol. Sci. 2017, 38, 649–665. [Google Scholar] [CrossRef] [PubMed]
  35. Vernon, G.; Baranova, A.; Younossi, Z.M. Systematic review: The epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment. Pharmacol. Ther. 2011, 34, 274–285. [Google Scholar] [CrossRef]
  36. Wondmkun, Y.T. Obesity, Insulin Resistance, and Type 2 Diabetes: Associations and Therapeutic Implications. Diabetes Metab. Syndr. Obes. 2020, 13, 3611–3616. [Google Scholar] [CrossRef]
  37. Miele, L.; Valenza, V.; La Torre, G.; Montalto, M.; Cammarota, G.; Ricci, R.; Masciana, R.; Forgione, A.; Gabrieli, M.L.; Perotti, G.; et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 2009, 49, 1877–1887. [Google Scholar] [CrossRef]
  38. Tokuhara, D. Role of the Gut Microbiota in Regulating Non-alcoholic Fatty Liver Disease in Children and Adolescents. Front. Nutr. 2021, 8, 700058. [Google Scholar] [CrossRef]
  39. Sharpton, S.R.; Ajmera, V.; Loomba, R. Emerging Role of the Gut Microbiome in Nonalcoholic Fatty Liver Disease: From Composition to Function. Clin. Gastroenterol. Hepatol. 2019, 17, 296–306. [Google Scholar] [CrossRef]
  40. Mehal, W.Z. The Gordian Knot of dysbiosis, obesity and NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 637–644. [Google Scholar] [CrossRef]
  41. Leung, C.; Rivera, L.; Furness, J.B.; Angus, P.W. The role of the gut microbiota in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 412–425. [Google Scholar] [CrossRef]
  42. Pickett-Blakely, O.; Young, K.; Carr, R.M. Micronutrients in Nonalcoholic Fatty Liver Disease Pathogenesis. Cell Mol. Gastroenterol. Hepatol. 2018, 6, 451–462. [Google Scholar] [CrossRef]
  43. Zhang, J.Z.; Cai, J.J.; Yu, Y.; She, Z.G.; Li, H. Nonalcoholic Fatty Liver Disease: An Update on the Diagnosis. Gene Expr. 2019, 19, 187–198. [Google Scholar] [CrossRef]
  44. Hernaez, R.; Lazo, M.; Bonekamp, S.; Kamel, I.; Brancati, F.L.; Guallar, E.; Clark, J.M. Diagnostic accuracy and reliability of ultrasonography for the detection of fatty liver: A meta-analysis. Hepatology 2011, 54, 1082–1090. [Google Scholar] [CrossRef]
  45. Esterson, Y.B.; Grimaldi, G.M. Radiologic Imaging in Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Clin. Liver Dis. 2018, 22, 93–108. [Google Scholar] [CrossRef] [PubMed]
  46. Maurice, J.; Manousou, P. Non-alcoholic fatty liver disease. Clin. Med. 2018, 18, 245–250. [Google Scholar] [CrossRef] [PubMed]
  47. Leoni, S.; Tovoli, F.; Napoli, L.; Serio, I.; Ferri, S.; Bolondi, L. Current guidelines for the management of non-alcoholic fatty liver disease: A systematic review with comparative analysis. World J. Gastroenterol. 2018, 24, 3361–3373. [Google Scholar] [CrossRef] [PubMed]
  48. Ofosu, A.; Ramai, D.; Reddy, M. Non-alcoholic fatty liver disease: Controlling an emerging epidemic, challenges, and future directions. Ann. Gastroenterol. 2018, 31, 288–295. [Google Scholar] [CrossRef]
  49. Pappachan, J.M.; Babu, S.; Krishnan, B.; Ravindran, N.C. Non-alcoholic Fatty Liver Disease: A Clinical Update. J. Clin. Transl. Hepatol. 2017, 5, 384–393. [Google Scholar] [CrossRef] [PubMed]
  50. Lee, D.H. Imaging evaluation of non-alcoholic fatty liver disease: Focused on quantification. Clin. Mol. Hepatol. 2017, 23, 290–301. [Google Scholar] [CrossRef] [PubMed]
  51. Newsome, P.N.; Sasso, M.; Deeks, J.J.; Paredes, A.; Boursier, J.; Chan, W.K.; Yilmaz, Y.; Czernichow, S.; Zheng, M.H.; Wong, V.W.; et al. FibroScan-AST (FAST) score for the non-invasive identification of patients with non-alcoholic steatohepatitis with significant activity and fibrosis: A prospective derivation and global validation study. Lancet Gastroenterol. Hepatol. 2020, 5, 362–373. [Google Scholar] [CrossRef]
  52. Tapper, E.B.; Castera, L.; Afdhal, N.H. FibroScan (vibration-controlled transient elastography): Where does it stand in the United States practice. Clin. Gastroenterol. Hepatol. 2015, 13, 27–36. [Google Scholar] [CrossRef] [PubMed]
  53. Imajo, K.; Kessoku, T.; Honda, Y.; Tomeno, W.; Ogawa, Y.; Mawatari, H.; Fujita, K.; Yoneda, M.; Taguri, M.; Hyogo, H.; et al. Magnetic Resonance Imaging More Accurately Classifies Steatosis and Fibrosis in Patients with Nonalcoholic Fatty Liver Disease Than Transient Elastography. Gastroenterology 2016, 150, 626–637.e7. [Google Scholar] [CrossRef] [PubMed]
  54. Noureddin, M.; Lam, J.; Peterson, M.R.; Middleton, M.; Hamilton, G.; Le, T.A.; Bettencourt, R.; Changchien, C.; Brenner, D.A.; Sirlin, C.; et al. Utility of magnetic resonance imaging versus histology for quantifying changes in liver fat in nonalcoholic fatty liver disease trials. Hepatology 2013, 58, 1930–1940. [Google Scholar] [CrossRef] [PubMed]
  55. Mikolasevic, I.; Milic, S.; Orlic, L.; Stimac, D.; Franjic, N.; Targher, G. Factors associated with significant liver steatosis and fibrosis as assessed by transient elastography in patients with one or more components of the metabolic syndrome. J. Diabetes Its Complicat. 2016, 30, 1347–1353. [Google Scholar] [CrossRef] [PubMed]
  56. Okamura, T.; Hashimoto, Y.; Hamaguchi, M.; Obora, A.; Kojima, T.; Fukui, M. The visceral adiposity index is a predictor of incident nonalcoholic fatty liver disease: A population-based longitudinal study. Clin. Res. Hepatol. Gastroenterol. 2020, 44, 375–383. [Google Scholar] [CrossRef]
  57. Chen, L.W.; Huang, P.R.; Chien, C.H.; Lin, C.L.; Chien, R.N. A community-based study on the application of fatty liver index in screening subjects with nonalcoholic fatty liver disease. J. Formos. Med. Assoc. 2020, 119, 173–181. [Google Scholar] [CrossRef]
  58. Bril, F.; McPhaul, M.J.; Caulfield, M.P.; Castille, J.M.; Poynard, T.; Soldevila-Pico, C.; Clark, V.C.; Firpi-Morell, R.J.; Lai, J.; Cusi, K. Performance of the SteatoTest, ActiTest, NashTest and FibroTest in a multiethnic cohort of patients with type 2 diabetes mellitus. J. Investig. Med. 2019, 67, 303–311. [Google Scholar] [CrossRef]
  59. Lin, B.; Ma, Y.; Wu, S.; Liu, Y.; Liu, L.; Wu, L. Novel Serum Biomarkers for Noninvasive Diagnosis and Screening of Nonalcoholic Fatty Liver Disease-Related Hepatic Fibrosis. OMICS 2019, 23, 181–189. [Google Scholar] [CrossRef]
  60. Xin, S.; Zhan, Q.; Chen, X.; Xu, J.; Yu, Y. Efficacy of serum miRNA test as a non-invasive method to diagnose nonalcoholic steatohepatitis: A systematic review and meta-analysis. BMC Gastroenterol. 2020, 20, 186. [Google Scholar] [CrossRef]
  61. Gunn, N.T.; Shiffman, M.L. The Use of Liver Biopsy in Nonalcoholic Fatty Liver Disease: When to Biopsy and in Whom. Clin. Liver Dis. 2018, 22, 109–119. [Google Scholar] [CrossRef]
  62. European Association for the Study of the Liver. EASL-ALEH Clinical Practice Guidelines: Non-invasive tests for evaluation of liver disease severity and prognosis. J. Hepatol. 2015, 63, 237–264. [Google Scholar] [CrossRef] [PubMed]
  63. Castera, L. Diagnosis of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: Non-invasive tests are enough. Liver Int. 2018, 38 (Suppl. S1), 67–70. [Google Scholar] [CrossRef] [PubMed]
  64. Boyd, A.; Cain, O.; Chauhan, A.; Webb, G.J. Medical liver biopsy: Background, indications, procedure and histopathology. Frontline Gastroenterol. 2020, 11, 40–47. [Google Scholar] [CrossRef] [PubMed]
  65. Ando, Y.; Jou, J.H. Nonalcoholic Fatty Liver Disease and Recent Guideline Updates. Clin. Liver Dis. 2021, 17, 23–28. [Google Scholar] [CrossRef]
  66. Said, A.; Akhter, A. Meta-Analysis of Randomized Controlled Trials of Pharmacologic Agents in Non-alcoholic Steatohepatitis. Ann. Hepatol. 2017, 16, 538–547. [Google Scholar] [CrossRef]
  67. Trauner, M.; Claudel, T.; Fickert, P.; Moustafa, T.; Wagner, M. Bile acids as regulators of hepatic lipid and glucose metabolism. Dig. Dis. 2010, 28, 220–224. [Google Scholar] [CrossRef]
  68. Wagner, M.; Zollner, G.; Trauner, M. Nuclear bile acid receptor farnesoid X receptor meets nuclear factor-kappaB: New insights into hepatic inflammation. Hepatology 2008, 48, 1383–1386. [Google Scholar] [CrossRef]
  69. Wang, Y.D.; Chen, W.D.; Wang, M.; Yu, D.; Forman, B.M.; Huang, W. Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology 2008, 48, 1632–1643. [Google Scholar] [CrossRef]
  70. Adorini, L.; Pruzanski, M.; Shapiro, D. Farnesoid X receptor targeting to treat nonalcoholic steatohepatitis. Drug Discov. Today 2012, 17, 988–997. [Google Scholar] [CrossRef]
  71. Mudaliar, S.; Henry, R.R.; Sanyal, A.J.; Morrow, L.; Marschall, H.U.; Kipnes, M.; Adorini, L.; Sciacca, C.I.; Clopton, P.; Castelloe, E.; et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology 2013, 145, 574–582.e1. [Google Scholar] [CrossRef] [PubMed]
  72. Schwabl, P.; Hambruch, E.; Budas, G.R.; Supper, P.; Burnet, M.; Liles, J.T.; Birkel, M.; Brusilovskaya, K.; Konigshofer, P.; Peck-Radosavljevic, M.; et al. The Non-Steroidal FXR Agonist Cilofexor Improves Portal Hypertension and Reduces Hepatic Fibrosis in a Rat NASH Model. Biomedicines 2021, 9, 60. [Google Scholar] [CrossRef]
  73. Patel, K.; Harrison, S.A.; Elkhashab, M.; Trotter, J.F.; Herring, R.; Rojter, S.E.; Kayali, Z.; Wong, V.W.; Greenbloom, S.; Jayakumar, S.; et al. Noureddin, Cilofexor, a Nonsteroidal FXR Agonist, in Patients With Noncirrhotic NASH: A Phase 2 Randomized Controlled Trial. Hepatology 2020, 72, 58–71. [Google Scholar] [CrossRef]
  74. Trauner, M.; Gulamhusein, A.; Hameed, B.; Caldwell, S.; Shiffman, M.L.; Landis, C.; Eksteen, B.; Agarwal, K.; Muir, A.; Rushbrook, S.; et al. The Nonsteroidal Farnesoid X Receptor Agonist Cilofexor (GS-9674) Improves Markers of Cholestasis and Liver Injury in Patients with Primary Sclerosing Cholangitis. Hepatology 2019, 70, 788–801. [Google Scholar] [CrossRef] [PubMed]
  75. Sanyal, A.J.; Lopez, P.; Lawitz, E.J.; Lucas, K.J.; Loeffler, J.; Kim, W.; Goh, G.B.B.; Huang, J.F.; Serra, C.; Andreone, P.; et al. Tropifexor for nonalcoholic steatohepatitis: An adaptive, randomized, placebo-controlled phase 2a/b trial. Nat. Med. 2023, 29, 392–400. [Google Scholar] [CrossRef]
  76. Pedrosa, M.; Seyedkazemi, S.; Francque, S.; Sanyal, A.; Rinella, M.; Charlton, M.; Loomba, R.; Ratziu, V.; Kochuparampil, J.; Fischer, L.; et al. A randomized, double-blind, multicenter, phase 2b study to evaluate the safety and efficacy of a combination of tropifexor and cenicriviroc in patients with nonalcoholic steatohepatitis and liver fibrosis: Study design of the TANDEM trial. Contemp. Clin. Trials 2020, 88, 105889. [Google Scholar] [CrossRef]
  77. Anstee, Q.M.; Lucas, K.J.; Francque, S.; Abdelmalek, M.F.; Sanyal, A.J.; Ratziu, V.; Gadano, A.C.; Rinella, M.; Charlton, M.; Loomba, R.; et al. Tropifexor plus cenicriviroc combination versus monotherapy in non-alcoholic steatohepatitis: Results from the Phase 2b TANDEM study. Hepatology 2023, 78, 1223–1239. [Google Scholar] [CrossRef]
  78. Grygiel-Gorniak, B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications—A review. Nutr. J. 2014, 13, 17. [Google Scholar] [CrossRef] [PubMed]
  79. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240. [Google Scholar] [CrossRef]
  80. Cariou, B.; Staels, B. GFT505 for the treatment of nonalcoholic steatohepatitis and type 2 diabetes. Expert. Opin. Investig. Drugs 2014, 23, 1441–1448. [Google Scholar] [CrossRef]
  81. Lefere, S.; Puengel, T.; Hundertmark, J.; Penners, C.; Frank, A.K.; Guillot, A.; de Muynck, K.; Heymann, F.; Adarbes, V.; Defrene, E.; et al. Differential effects of selective- and pan-PPAR agonists on experimental steatohepatitis and hepatic macrophages. J. Hepatol. 2020, 73, 757–770. [Google Scholar] [CrossRef]
  82. Munigoti, S.P.; Harinarayan, C.V. Role of Glitazars in atherogenic dyslipidemia and diabetes: Two birds with one stone? Indian J. Endocrinol. Metab. 2014, 18, 283–287. [Google Scholar] [CrossRef]
  83. Kaul, U.; Parmar, D.; Manjunath, K.; Shah, M.; Parmar, K.; Patil, K.P.; Jaiswal, A. New dual peroxisome proliferator activated receptor agonist-Saroglitazar in diabetic dyslipidemia and non-alcoholic fatty liver disease: Integrated analysis of the real-world evidence. Cardiovasc. Diabetol. 2019, 18, 80. [Google Scholar] [CrossRef]
  84. Gawrieh, S.; Noureddin, M.; Loo, N.; Mohseni, R.; Awasty, V.; Cusi, K.; Kowdley, K.V.; Lai, M.; Schiff, E.; Parmar, D.; et al. Saroglitazar, a PPAR-alpha/gamma Agonist, for Treatment of NAFLD: A Randomized Controlled Double-Blind Phase 2 Trial. Hepatology 2021, 74, 1809–1824. [Google Scholar] [CrossRef] [PubMed]
  85. Siddiqui, M.S.; Idowu, M.O.; Parmar, D.; Borg, B.B.; Denham, D.; Loo, N.M.; Lazas, D.; Younes, Z.; Sanyal, A.J.; Phase, A. Randomized Controlled Trial of Saroglitazar in Patients With Nonalcoholic Steatohepatitis. Clin. Gastroenterol. Hepatol. 2021, 19, 2670–2672. [Google Scholar] [CrossRef] [PubMed]
  86. Panebianco, C.; Oben, J.A.; Vinciguerra, M.; Pazienza, V. Senescence in hepatic stellate cells as a mechanism of liver fibrosis reversal: A putative synergy between retinoic acid and PPAR-gamma signalings. Clin. Exp. Med. 2017, 17, 269–280. [Google Scholar] [CrossRef] [PubMed]
  87. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  88. Gupta, N.A.; Mells, J.; Dunham, R.M.; Grakoui, A.; Handy, J.; Saxena, N.K.; Anania, F.A. Glucagon-like peptide-1 receptor is present on human hepatocytes and has a direct role in decreasing hepatic steatosis in vitro by modulating elements of the insulin signaling pathway. Hepatology 2010, 51, 1584–1592. [Google Scholar] [CrossRef] [PubMed]
  89. Holman, R.R.; Bethel, M.A.; Mentz, R.J.; Thompson, V.P.; Lokhnygina, Y.; Buse, J.B.; Chan, J.C.; Choi, J.; Gustavson, S.M.; Iqbal, N.; et al. Effects of Once-Weekly Exenatide on Cardiovascular Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 1228–1239. [Google Scholar] [CrossRef]
  90. Aroda, V.R. A review of GLP-1 receptor agonists: Evolution and advancement, through the lens of randomised controlled trials. Diabetes Obes. Metab. 2018, 20 (Suppl. 1), 22–33. [Google Scholar] [CrossRef]
  91. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; Abouda, G.; Aldersley, M.A.; 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, 679–690. [Google Scholar] [CrossRef]
  92. Petit, J.M.; Cercueil, J.P.; Loffroy, R.; Denimal, D.; Bouillet, B.; Fourmont, C.; Chevallier, O.; Duvillard, L.; Verges, B. Effect of Liraglutide Therapy on Liver Fat Content in Patients With Inadequately Controlled Type 2 Diabetes: The Lira-NAFLD Study. J. Clin. Endocrinol. Metab. 2017, 102, 407–415. [Google Scholar] [CrossRef] [PubMed]
  93. Yan, J.; Yao, B.; Kuang, H.; Yang, X.; Huang, Q.; Hong, T.; Li, Y.; Dou, J.; Yang, W.; Qin, G.; et al. Liraglutide, Sitagliptin, and Insulin Glargine Added to Metformin: The Effect on Body Weight and Intrahepatic Lipid in Patients with Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 2414–2426. [Google Scholar] [CrossRef] [PubMed]
  94. Capehorn, M.S.; Catarig, A.M.; Furberg, J.K.; Janez, A.; Price, H.C.; Tadayon, S.; Verges, B.; Marre, M. Efficacy and safety of once-weekly semaglutide 1.0mg vs once-daily liraglutide 1.2mg as add-on to 1–3 oral antidiabetic drugs in subjects with type 2 diabetes (SUSTAIN 10). Diabetes Metab. 2020, 46, 100–109. [Google Scholar] [CrossRef] [PubMed]
  95. Smits, M.M.; Van Raalte, D.H. Safety of Semaglutide. Front. Endocrinol. 2021, 12, 645563. [Google Scholar] [CrossRef]
  96. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.S.; Harrison, S.A.; Investigators, N.N. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124. [Google Scholar] [CrossRef]
  97. Ghusn, W.; De la Rosa, A.; Sacoto, D.; Cifuentes, L.; Campos, A.; Feris, F.; Hurtado, M.D.; Acosta, A. Weight Loss Outcomes Associated with Semaglutide Treatment for Patients With Overweight or Obesity. JAMA Netw. Open 2022, 5, e2231982. [Google Scholar] [CrossRef]
  98. Bergmann, N.C.; Davies, M.J.; Lingvay, I.; Knop, F.K. Semaglutide for the treatment of overweight and obesity: A review. Diabetes Obes. Metab. 2023, 25, 18–35. [Google Scholar] [CrossRef] [PubMed]
  99. Frias, J.P.; Nauck, M.A.; Van, J.; Kutner, M.E.; Cui, X.; Benson, C.; Urva, S.; Gimeno, R.E.; Milicevic, Z.; Robins, D.; 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, 2180–2193. [Google Scholar] [CrossRef]
  100. Ambery, P.; Parker, V.E.; Stumvoll, M.; Posch, M.G.; Heise, T.; Plum-Moerschel, L.; Tsai, L.F.; Robertson, D.; Jain, M.; Petrone, M.; et al. MEDI0382, a GLP-1 and glucagon receptor dual agonist, in obese or overweight patients with type 2 diabetes: A randomised, controlled, double-blind, ascending dose and phase 2a study. Lancet 2018, 391, 2607–2618. [Google Scholar] [CrossRef]
  101. Harrison, S.A.; Bashir, M.R.; Guy, C.D.; Zhou, R.; Moylan, C.A.; Frias, J.P.; Alkhouri, N.; Bansal, M.B.; Baum, S.; Neuschwander-Tetri, B.A.; et al. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: A multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2019, 394, 2012–2024. [Google Scholar] [CrossRef] [PubMed]
  102. Ratziu, V.; Sanyal, A.; Harrison, S.A.; Wong, V.W.; Francque, S.; Goodman, Z.; Aithal, G.P.; Kowdley, K.V.; Seyedkazemi, S.; Fischer, L.; et al. Cenicriviroc Treatment for Adults With Nonalcoholic Steatohepatitis and Fibrosis: Final Analysis of the Phase 2b CENTAUR Study. Hepatology 2020, 72, 892–905. [Google Scholar] [CrossRef] [PubMed]
  103. Anstee, Q.M.; Neuschwander-Tetri, B.A.; Wong, V.W.; Abdelmalek, M.F.; Landgren, H.; Bedossa, P.; Alkhouri, N.; Tacke, F.; Sanyal, A. Cenicriviroc for the treatment of liver fibrosis in adults with nonalcoholic steatohepatitis: AURORA Phase 3 study design. Contemp. Clin. Trials 2020, 89, 105922. [Google Scholar] [CrossRef] [PubMed]
  104. Harrison, S.A.; Wong, V.W.; Okanoue, T.; Bzowej, N.; Vuppalanchi, R.; Younes, Z.; Kohli, A.; Sarin, S.; Caldwell, S.H.; Alkhouri, N.; 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. [Google Scholar] [CrossRef]
  105. Harrison, S.A.; Abdelmalek, M.F.; Caldwell, S.; Shiffman, M.L.; Diehl, A.M.; Ghalib, R.; Lawitz, E.J.; Rockey, D.C.; Schall, R.A.; Jia, C.; et al. Simtuzumab Is Ineffective for Patients With Bridging Fibrosis or Compensated Cirrhosis Caused by Nonalcoholic Steatohepatitis. Gastroenterology 2018, 155, 1140–1153. [Google Scholar] [CrossRef]
  106. Linton, S.D.; Aja, T.; Armstrong, R.A.; Bai, X.; Chen, L.S.; Chen, N.; Ching, B.; Contreras, P.; Diaz, J.L.; Fisher, C.D.; et al. First-in-class pan caspase inhibitor developed for the treatment of liver disease. J. Med. Chem. 2005, 48, 6779–6782. [Google Scholar] [CrossRef]
  107. Harrison, S.A.; Goodman, Z.; Jabbar, A.; Vemulapalli, R.; Younes, Z.H.; Freilich, B.; Sheikh, M.Y.; Schattenberg, J.M.; Kayali, Z.; Zivony, A.; et al. A randomized, placebo-controlled trial of emricasan in patients with NASH and F1-F3 fibrosis. J. Hepatol. 2020, 72, 816–827. [Google Scholar] [CrossRef]
  108. Barreyro, F.J.; Holod, S.; Finocchietto, P.V.; Camino, A.M.; Aquino, J.B.; Avagnina, A.; Carreras, M.C.; Poderoso, J.J.; Gores, G.J. The pan-caspase inhibitor Emricasan (IDN-6556) decreases liver injury and fibrosis in a murine model of non-alcoholic steatohepatitis. Liver Int. 2015, 35, 953–966. [Google Scholar] [CrossRef]
  109. Zhou, J.; Chen, Y.; Yu, J.; Li, T.; Lu, Z.; Chen, Y.; Zhang, X.; Ye, F. The efficacy of novel metabolic targeted agents and natural plant drugs for nonalcoholic fatty liver disease treatment: A PRISMA-compliant network meta-analysis of randomized controlled trials. Medicine 2021, 100, e24884. [Google Scholar] [CrossRef]
  110. Konstantinidi, M.; Koutelidakis, A.E. Functional Foods and Bioactive Compounds: A Review of Its Possible Role on Weight Management and Obesity’s Metabolic Consequences. Medicines 2019, 6, 94. [Google Scholar] [CrossRef]
  111. Gillessen, A. Schmidt HH. Silymarin as Supportive Treatment in Liver Diseases: A Narrative Review. Adv. Ther. 2020, 37, 1279–1301. [Google Scholar] [CrossRef] [PubMed]
  112. Izzo, C.; Annunziata, M.; Melara, G.; Sciorio, R.; Dallio, M.; Masarone, M.; Federico, A.; Persico, M. The Role of Resveratrol in Liver Disease: A Comprehensive Review from In Vitro to Clinical Trials. Nutrients 2021, 13, 933. [Google Scholar] [CrossRef] [PubMed]
  113. Muller, T.D.; Bluher, M.; Tschop, M.H.; DiMarchi, R.D. Anti-obesity drug discovery: Advances; challenges. Nat. Rev. Drug Discov. 2022, 21, 201–223. [Google Scholar] [CrossRef]
  114. Harrison, S.A.; Fecht, W.; Brunt, E.M.; Neuschwander-Tetri, B.A. Orlistat for overweight subjects with nonalcoholic steatohepatitis: A randomized, prospective trial. Hepatology 2009, 49, 80–86. [Google Scholar] [CrossRef] [PubMed]
  115. Bajaj, H.S.; Burrows, M.; Blavignac, J.; Paron, E.; Camacho, F.; Gould, E.; Barakat, M. Extended-release naltrexone/bupropion and liver health: Pooled, post hoc analysis from four randomized controlled trials. Diabetes Obes. Metab. 2021, 23, 861–865. [Google Scholar] [CrossRef]
  116. Alexander, M.; Loomis, A.K.; van der Lei, J.; Duarte-Salles, T.; Prieto-Alhambra, D.; Ansell, D.; Pasqua, A.; Lapi, F.; Rijnbeek, P.; Mosseveld, M.; et al. Risks and clinical predictors of cirrhosis and hepatocellular carcinoma diagnoses in adults with diagnosed NAFLD: Real-world study of 18 million patients in four European cohorts. BMC Med. 2019, 17, 95. [Google Scholar] [CrossRef]
  117. Mantovani, A.; Petracca, G.; Beatrice, G.; Tilg, H.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and risk of incident diabetes mellitus: An updated meta-analysis of 501 022 adult individuals. Gut 2021, 70, 962–969. [Google Scholar] [CrossRef]
  118. Bolinder, J.; Ljunggren, O.; Kullberg, J.; Johansson, L.; Wilding, J.; Langkilde, A.M.; Sugg, J.; Parikh, S. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J. Clin. Endocrinol. Metab. 2012, 97, 1020–1031. [Google Scholar] [CrossRef]
  119. Loomba, R.; Noureddin, M.; Kowdley, K.V.; Kohli, A.; Sheikh, A.; Neff, G.; Bhandari, B.R.; Gunn, N.; Caldwell, S.H.; Goodman, Z.; et al. Combination Therapies Including Cilofexor and Firsocostat for Bridging Fibrosis and Cirrhosis Attributable to NASH. Hepatology 2021, 73, 625–643. [Google Scholar] [CrossRef]
  120. Ito, D.; Shimizu, S.; Inoue, K.; Saito, D.; Yanagisawa, M.; Inukai, K.; Akiyama, Y.; Morimoto, Y.; Noda, M.; Shimada, A. Comparison of Ipragliflozin and Pioglitazone Effects on Nonalcoholic Fatty Liver Disease in Patients With Type 2 Diabetes: A Randomized, 24-Week, Open-Label, Active-Controlled Trial. Diabetes Care 2017, 40, 1364–1372. [Google Scholar] [CrossRef]
  121. Sumida, Y.; Murotani, K.; Saito, M.; Tamasawa, A.; Osonoi, Y.; Yoneda, M.; Osonoi, T. Effect of luseogliflozin on hepatic fat content in type 2 diabetes patients with non-alcoholic fatty liver disease: A prospective, single-arm trial (LEAD trial). Hepatol. Res. 2019, 49, 64–71. [Google Scholar] [CrossRef]
  122. Cusi, K.; Bril, F.; Barb, D.; Polidori, D.; Sha, S.; Ghosh, A.; Farrell, K.; Sunny, N.E.; Kalavalapalli, S.; Pettus, J.; et al. Effect of canagliflozin treatment on hepatic triglyceride content and glucose metabolism in patients with type 2 diabetes. Diabetes Obes. Metab. 2019, 21, 812–821. [Google Scholar] [CrossRef]
  123. Lv, Z.; Guo, Y. Metformin and Its Benefits for Various Diseases. Front. Endocrinol. 2020, 11, 191. [Google Scholar] [CrossRef] [PubMed]
  124. Tajima, K.; Nakamura, A.; Shirakawa, J.; Togashi, Y.; Orime, K.; Sato, K.; Inoue, H.; Kaji, M.; Sakamoto, E.; Ito, Y.; et al. Metformin prevents liver tumorigenesis induced by high-fat diet in C57Bl/6 mice. American journal of physiology. Endocrinol. Metab. 2013, 305, E987–E998. [Google Scholar]
  125. Harrison, S.A.; Neff, G.; Guy, C.D.; Bashir, M.R.; Paredes, A.H.; Frias, J.P.; Younes, Z.; Trotter, J.F.; Gunn, N.T.; Moussa, S.E.; 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, 219–231.e1. [Google Scholar] [CrossRef]
  126. Harrison, S.A.; Ruane, P.J.; Freilich, B.L.; Neff, G.; Patil, R.; Behling, C.A.; Hu, C.; Fong, E.; de Temple, B.; Tillman, E.J.; et al. Efruxifermin in non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled, phase 2a trial. Nat. Med. 2021, 27, 1262–1271. [Google Scholar] [CrossRef]
  127. Sanyal, A.; Charles, E.D.; Neuschwander-Tetri, B.A.; Loomba, R.; Harrison, S.A.; Abdelmalek, M.F.; Lawitz, E.J.; Halegoua-DeMarzio, D.; Kundu, S.; Noviello, S.; et al. Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: A randomised, double-blind, placebo-controlled, phase 2a trial. Lancet 2019, 392, 2705–2717. [Google Scholar] [CrossRef]
  128. Verzijl, C.R.C.; Van De Peppel, I.P.; Struik, D.; Jonker, J.W. Pegbelfermin (BMS-986036): An investigational PEGylated fibroblast growth factor 21 analogue for the treatment of nonalcoholic steatohepatitis. Expert. Opin. Investig. Drugs 2020, 29, 125–133. [Google Scholar] [CrossRef] [PubMed]
  129. Ratziu, V.; Harrison, S.A.; Francque, S.; Bedossa, P.; Lehert, P.; Serfaty, L.; Romero-Gomez, M.; Boursier, J.; Abdelmalek, M.; Caldwell, S.; et al. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-alpha and -delta, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology 2016, 150, 1147–1159.e5. [Google Scholar] [CrossRef] [PubMed]
  130. Francque, S.M.; Bedossa, P.; Ratziu, V.; Anstee, Q.M.; Bugianesi, E.; Sanyal, A.J.; Loomba, R.; Harrison, S.A.; Balabanska, R.; Mateva, L.; et al. Controlled Trial of the Pan-PPAR Agonist Lanifibranor in NASH. N. Engl. J. Med. 2021, 385, 1547–1558. [Google Scholar] [CrossRef] [PubMed]
  131. Seo, Y.G. Side Effects Associated with Liraglutide Treatment for Obesity as Well as Diabetes. J. Obes. Metab. Syndr. 2021, 30, 12–19. [Google Scholar] [CrossRef]
  132. Mishra, R.; Raj, R.; Elshimy, G.; Zapata, I.; Kannan, L.; Majety, P.; Edem, D.; Correa, R. Adverse Events Related to Tirzepatide. J. Endocr. Soc. 2023, 7, bvad016. [Google Scholar] [CrossRef] [PubMed]
  133. Chavda, V.P.; Ajabiya, J.; Teli, D.; Bojarska, J.; Apostolopoulos, V. Tirzepatide, a New Era of Dual-Targeted Treatment for Diabetes and Obesity: A Mini-Review. Molecules 2022, 27, 4315. [Google Scholar] [CrossRef]
  134. Parker, V.E.R.; Hoang, T.; Schlichthaar, H.; Gibb, F.W.; Wenzel, B.; Posch, M.G.; Rose, L.; Chang, Y.T.; Petrone, M.; Hansen, L.; et al. Efficacy and safety of cotadutide, a dual glucagon-like peptide-1 and glucagon receptor agonist, in a randomized phase 2a study of patients with type 2 diabetes and chronic kidney disease. Diabetes Obes. Metab. 2022, 24, 1360–1369. [Google Scholar] [CrossRef] [PubMed]
  135. Nahra, R.; Wang, T.; Gadde, K.M.; Oscarsson, J.; Stumvoll, M.; Jermutus, L.; Hirshberg, B.; Ambery, P. Effects of Cotadutide on Metabolic and Hepatic Parameters in Adults With Overweight or Obesity and Type 2 Diabetes: A 54-Week Randomized Phase 2b Study. Diabetes Care 2021, 44, 1433–1442. [Google Scholar] [CrossRef] [PubMed]
  136. Neal, B.; Perkovic, V.; Mahaffey, K.W.; de Zeeuw, D.; Fulcher, G.; Erondu, N.; Shaw, W.; Law, G.; Desai, M.; Matthews, D.R.; et al. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 644–657. [Google Scholar] [CrossRef]
  137. Jakher, H.; Chang, T.I.; Tan, M.; Mahaffey, K.W. Canagliflozin review—Safety and efficacy profile in patients with T2DM. Diabetes Metab. Syndr. Obes. 2019, 12, 209–215. [Google Scholar] [CrossRef]
  138. Frampton, J.E. Empagliflozin: A Review in Type 2 Diabetes. Drugs 2018, 78, 1037–1048. [Google Scholar] [CrossRef]
Figure 1. Differential features of NAFLD and MAFLD. Steatosis is a common feature in both NAFLD and MAFLD. A strong association of overweight, obesity, type 2 diabetes, and metabolic risk factors such as hypertension, insulin resistance, high triglycerides level, low LDL cholesterol, and high blood glucose level differentiate MAFLD from NAFLD.
Figure 1. Differential features of NAFLD and MAFLD. Steatosis is a common feature in both NAFLD and MAFLD. A strong association of overweight, obesity, type 2 diabetes, and metabolic risk factors such as hypertension, insulin resistance, high triglycerides level, low LDL cholesterol, and high blood glucose level differentiate MAFLD from NAFLD.
Livers 03 00040 g001
Figure 2. Various etiological agents responsible for the initiation and progression of NAFLD/MAFLD. Various etiological agents such as unhealthy diet, high fat diet, malnutrition, high carbohydrate diet are responsible for the development of metabolic disease such as obesity and or diabetes. Bacterial toxins and gut microbiome disbalance are also associated with the development of disease. As a result, exposure of these agents to subjects develops fatty liver and leads to disease progression as inflammation may lead to fibrosis, cirrhosis, and hepatocellular carcinoma depending on persistence of etiological agents. Age, race, gender, and genetics of persons show variations in disease pattern.
Figure 2. Various etiological agents responsible for the initiation and progression of NAFLD/MAFLD. Various etiological agents such as unhealthy diet, high fat diet, malnutrition, high carbohydrate diet are responsible for the development of metabolic disease such as obesity and or diabetes. Bacterial toxins and gut microbiome disbalance are also associated with the development of disease. As a result, exposure of these agents to subjects develops fatty liver and leads to disease progression as inflammation may lead to fibrosis, cirrhosis, and hepatocellular carcinoma depending on persistence of etiological agents. Age, race, gender, and genetics of persons show variations in disease pattern.
Livers 03 00040 g002
Table 1. Therapeutic profile of pharmacological agents developed for MAFLD/NAFLD.
Table 1. Therapeutic profile of pharmacological agents developed for MAFLD/NAFLD.
Pharmacological
Agents
Therapeutic
Class
Activity
Profile
Adverse/
Side Effect
Current
Status
Commercialized
Trade Name
Remarks
Obeticholic acid
(OCA) [70,71]
FXRFor primary biliary cholangitis, possibly may have beneficial role in NASH, reduced inflammation,
fibrosis, insulin sensitivity, supports further clinical studies.
Lipid increased abnormality, cardiovascular abnormality, mild to moderate pruritus. May cause liver damage. Medication for PBC
primary
biliary cholangitis.
OcalivaOcaliva may cause serious side effects including liver failure
Cilofexor [72,73]FXR agonist
2nd generation
NAFLD, NASH (Non-cirrhotic), PSC (primary sclerosis cholangitis), reduction in steatosisGenerally well tolerated, most frequently headache,
with combination of Firsocostat caused pruritis or skin itching, headache, diarrhea and nausea
Phase II Not approved by US FDA
no NASH resolution, no
decreased fibrosis
Tropifexor [75,76,77]FXR, 2nd generationCholestatic liver disease, NASH, reduction in steatosis, improvement in liver fibrosis in combination with cenicrivirocSignificant pruritus,
increased LDL cholesterol
Phase II in progress Discontinued because of side effects
Elafibranor [80,129]Dual PPAR
alpha/delta
agonist
Diabetes2, IR, dyslepidemia,
NAFLD, NASH resolution,
no fibrosis decreased
Generally, well tolerated, renal impairment, elevated serum creatinine level Phase IIb
Phase III in clinical pathway, FDA grants approval for PBC
Investigational.
not approved for any particular hepatic condition. elevated serum creatinine concern about its safety.
Lanifibranor [81,130]Pan-PPAR
Agonist of PPAR alpha, PPAR beta and delta, PPAR gamma.
Reduction in NASH, lipid profile, glycemic control Highest risk of diarrhea, constipation, nausea, abdominal painApproved for Phase III
and other studies are in progress
Research is in progress
Saroglitazar [82,83,84,85]Dual PPAR, alpha/
delta agonist
Type 2 diabetes, dislipidemia. potential therapeutic option for NAFLD, approved for NASH diabetic nephropathy,
hypertriglyceridemia
No major but increased serum creatin may cause concern. May cause
nausea, vomiting, diarrhea, headache
Phase IILipaglynLongterm cardiovascular
safety not established,
qualified for NASH treatment
In India
Liraglutide [91,92,93,131]GLP-1R receptor agonistType 2 diabetes,
chronic obesity
atherosclerotic cardiovascular disease NASH resolution, no decreased fibrosis
GI side effect
low blood glucose
thyroid cancer,
pancreatitis
Phase IIVictoza,
Saxenda
Semaglutide [94,95,96,97,98]GLP-1RDiabetic Type 2,
obesity, plus diet & exercise
NASH resolution but no decrease in fibrosis.
GI side effect. GERD OzempicContraindication in people with thyroid carcinoma
Tirzapatde [99,132,133]Dual GIP and GLP1 agonistType 2 diabetes GI side effects, abdominal pain, nausea, vomiting. Diarrhea, loss of appetite, tiredness.Phase IIbMounjiaroEfficacy in NASH is under evaluation
Cotadutide [100,134,135]Glucagon and GLP1agonistType 2 diabetes
Reducing weight, supports for further evaluation after NASH and CKD with type 2 diabetes.
Increased incidence of gastrointestinal disordersPhaseII,
Phase III
Investigational drug
Resmetirom [101]THR-BNAFLD
NASH resolution
GI side effect, nausea, diarrhea, otherwise well tolerated.Phase III Investigational drug
Cenicriviroc [102,103]CCR2/CCR5
antagonist
No NASH resolution
decreased Fibrosis
Phase IIb
No benefit in NASH, hence stopped.
Not approved.
Selonsertib [104]Antifibrotic inhibitor of ASK1treatment of NASHIncreases risk of cirrhosis fibrosis, scarring cirrhosis, liver failure and cancer Does not reduce fibrosis. Not developed
Simtuzumab [105]Antifibrotic drug
humanised monoclonal antibody
FibrosisCaused IPF (idiopathic pulmonary fibrosisPhase II terminated Lack of efficacy
Emericasan [106]Pancaspase inhibitorLiver disease, NASH and cirrhosis, developed for decreased fibrosisWell toleratedPhase II Investigational drug
Canagliflozin [136,137]SGL2 inhibitorType2 diabetes, reduces risk of heart attack
ESKD (End Stage kidney disease), improvement in liver fat content
Low blood sugar, G.I. problem, kidney problem, UTI, increased risk of diabetic ketoacidosis Phase II trial
For treatment of obesity in US and Europe
Invokana, Sulisent,
Prominand
FDA Concern about cardiovascular safety.
Increased risk of leg and foot amputation
Empagliflozin [138]SGL2 inhibitorType2 diabetes, proven lower risk of cardiovascular death and hospitalization for heart failure.
Improvement in liver fat content.
Hyperventilation,
diabetes ketoacidosis,
urinary tract infection, female genital mycotic infection
US FDA approvedJardiance
Aldafermin [125]Analogue of fibroblast growth factor 19Decreases amount of liver fat in NASH and F1–F3, liver fatGenerally mild to moderate diarrhea, headache, nausea and joint painPhase II Not developed due to uncertainty of efficacy
Efruxifermin [126]FGF21 Fibroblast growth factorsDecreases hepatic fat in NASH and F1–F3 fibrosis, improves insulin sensitivity, lipoproteinWell toleratedPhase IIa
Progress in longterm studies for NASH underway
Pegbelfermin [127,128]PEGylated fibroblast growth factor 21 analogueFor the treatment of NASH, improvement in liver damage and cardiometabolic diseases. Under phase II
trial
Investigational
drug
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bhopale, K.K.; Srinivasan, M.P. Therapeutics for Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD). Livers 2023, 3, 597-617. https://doi.org/10.3390/livers3040040

AMA Style

Bhopale KK, Srinivasan MP. Therapeutics for Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD). Livers. 2023; 3(4):597-617. https://doi.org/10.3390/livers3040040

Chicago/Turabian Style

Bhopale, Kamlesh K., and Mukund P. Srinivasan. 2023. "Therapeutics for Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD)" Livers 3, no. 4: 597-617. https://doi.org/10.3390/livers3040040

Article Metrics

Back to TopTop