Next Article in Journal
Cytotoxic Metabolites from Calophyllum tacamahaca Willd.: Isolation and Detection through Feature-Based Molecular Networking
Previous Article in Journal
Thymoquinone Antifungal Activity against Candida glabrata Oral Isolates from Patients in Intensive Care Units—An In Vitro Study
Previous Article in Special Issue
Exercise Augments the Effect of SGLT2 Inhibitor Dapagliflozin on Experimentally Induced Diabetic Cardiomyopathy, Possible Underlying Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glucagon-like Peptide-1 Receptor Agonists in Patients with Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease—Current Background, Hopes, and Perspectives

by
Georgiana-Diana Cazac
1,
Cristina-Mihaela Lăcătușu
1,2,*,
Gabriela Ștefănescu
3,4,
Cătălina Mihai
3,4,
Elena-Daniela Grigorescu
1,*,
Alina Onofriescu
1,2 and
Bogdan-Mircea Mihai
1,2
1
Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
2
Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
3
Unit of Medical Semiology and Gastroenterology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
4
Institute of Gastroenterology and Hepatology, “Sf. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
*
Authors to whom correspondence should be addressed.
Metabolites 2023, 13(5), 581; https://doi.org/10.3390/metabo13050581
Submission received: 22 March 2023 / Revised: 20 April 2023 / Accepted: 21 April 2023 / Published: 23 April 2023
(This article belongs to the Special Issue Key Role of Drug Intervention on Diabetic Complications)

Abstract

:
Nonalcoholic fatty liver disease (NAFLD) represents the most common chronic liver disease worldwide, reaching one of the highest prevalences in patients with type 2 diabetes mellitus (T2DM). For now, no specific pharmacologic therapies are approved to prevent or treat NAFLD. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are currently evaluated as potential candidates for NAFLD treatment in patients with T2DM. Some representatives of this class of antihyperglycemic agents emerged as potentially beneficial in patients with NAFLD after several research studies suggested they reduce hepatic steatosis, ameliorate lesions of nonalcoholic steatohepatitis (NASH), or delay the progression of fibrosis in this population. The aim of this review is to summarize the body of evidence supporting the effectiveness of GLP-1RA therapy in the management of T2DM complicated with NAFLD, describing the studies that evaluated the effects of these glucose-lowering agents in fatty liver disease and fibrosis, their possible mechanistic justification, current evidence-based recommendations, and the next steps to be developed in the field of pharmacological innovation.

1. Introduction

Type 2 diabetes mellitus (T2DM) is a chronic health condition that shares a wide range of risk factors and pathogenic elements with obesity and nonalcoholic fatty liver disease (NAFLD) [1]. The prevalence of diabetes in adults aged 20–79 years worldwide is estimated at 10.5%, of whom more than 90% have T2DM [2].
Along with the diabetes pandemic, today, NAFLD represents the most prevalent liver disease worldwide and is perceived as the hepatic manifestation of the metabolic syndrome [3]. The global prevalence of NAFLD among adults is currently 25–30% [4] and is predicted to reach 55.7% in 2040 [5]. More than 90% of the patients with severe obesity, and 50% of T2DM patients have NAFLD [4,6]. T2DM is often associated with multiple organ dysfunction, including eye, kidney, liver, and cardiovascular disease. NAFLD represents the least screened diabetes mellitus complication today; this is a discouraging finding, given the rapid progression towards nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, or even hepatocellular carcinoma, all leading to a rising liver-related mortality [7].
The relationship between NAFLD and T2DM is bidirectional and requires increased awareness [8]. The presence of diabetes promotes the progression of NAFLD toward advanced liver disease, while the presence of NAFLD increases the risk of developing other diabetes-related complications or even worsening the already present ones [9,10].
Accumulating evidence supports the use of some antidiabetic agents, such as pioglitazone and glucagon-like peptide-1 receptor agonists (GLP-1RAs), to improve histological findings of NASH and fibrosis in T2DM patients [11]. Well-designed trials with these agents have been published, but the insufficient duration of the studies for both these classes limits the significance of their results [12]. In the case of GLP-1RAs, the hepatic benefits found in several studies that focused on NAFLD-associating patients have added to the already acknowledged effects of improving cardiovascular outcomes, preventing the nephropathy progression and reducing all-cause mortality in T2DM patients [13].
GLP-1RAs can be an effective treatment in patients with T2DM and NAFLD, so the aim of this comprehensive review is to summarize the existing evidence on the effectiveness of GLP-1RA therapy in the management of NAFLD that complicates T2DM, describing the studies that evaluated the effects of these glucose-lowering agents in fatty liver disease and fibrosis, their potential mechanistic justification, current evidence-based recommendations, and the next steps to be developed in the field of pharmacological innovation.

2. Basic Concepts in the Management of Patients with T2DM and NAFLD

Screening of people at risk for liver disease is advised for all patients with T2DM, as well as in individuals who are overweight or living with obesity, in people with metabolic syndrome, or in those living with cardiovascular disease [14,15].
Once the diagnosis of NAFLD is established, an intensive management strategy needs to be implemented. The cornerstone of NAFLD treatment remains lifestyle optimization; paradoxically enough, given the increased incidence of the disease and the vital risks involved by its progression, there are still no drugs specifically approved for NAFLD therapy [14,16]. The treatment objectives involve avoidance or delayed progression of diabetes-related complications (cardiovascular disease and liver cancer included) by improving the metabolic profile, and the reduction of the liver fat load, inflammation, or even fibrosis, given the liver’s ability to regenerate [14].
Lifestyle modification in the management of NAFLD includes initiation and maintenance of weight loss through nutritional intervention and increased physical activity patterns [17,18]. Nevertheless, nonpharmacological therapy displays limited efficacy for weight-related outcomes, which impairs its ability to change the course of liver disease [19].
Antihyperglycemic agents supporting weight loss in patients with T2DM have shown beneficial effects in NAFLD [20]. However, it appears that improvement of glycemic control in patients with co-existent NAFLD and diabetes may be associated with an improvement in NAFLD independently of the lost weight [21]. Glycemic outcomes cannot be considered outside the need to tackle metabolic traits such as dyslipidemia or obesity, which improves the liver condition in patients with or without diabetes [22].
Along with sodium-glucose cotransporter 2 (SGLT2) inhibitors, GLP-1RAs have become the first-line therapy for T2DM due to their potent glucose-lowering effects and their ability to reduce the cardiovascular risk irrespective of the glycemic improvement or the weight loss [23].
GLP-1RAs belong to the incretin-based therapies with subcutaneous or oral administration that are currently approved for the treatment of T2DM and obesity (Table 1) [24,25]. They are intensively scrutinized, given their beneficial impact on the glycemic control, their ability to reduce the visceral adipose tissue and the total body weight, and their cardio-reno-metabolic benefits in patients with T2DM and obesity (Figure 1) [26,27,28]. Therefore, the inference of their benefits in NAFLD-complicated diabetes, or even in NAFLD without associated diabetes, was only a step further [29]. Some data even suggest that GLP-1RAs have metabolic effects related to NAFLD pathophysiology [22].
Some of the high-impact guidelines for NAFLD have already embedded the use of GLP-1RAs (Table 2). United States guidelines were the first to support the use of GLP-1RAs in patients with T2DM and NASH [12,15]. As of now, further studies are still needed to clarify the role of GLP-1RAs as an NAFLD-targeted therapy in persons with or without diabetes, including their ability to interfere with each of the histological changes seen in this metabolic liver disease, as a valuable addition to their well-recognized capacity to improve cardiometabolic health [39].

3. Search Methods

We performed an extensive literature search of PubMed and ClinicalTrials.Gov databases using the terms “GLP-1 receptor agonist” or “GLP-1RA” combined with “fatty liver” or “NAFLD” or “NASH” or “fibrosis” and “diabetes mellitus” or “DM” or “diabetic patients” or “T2DM”. Further research was made using the generic names of the GLP-1RA representatives, specifically “exenatide”, “lixisenatide”, “dulaglutide”, “liraglutide”, “semaglutide”, “albiglutide”, and “efpeglenatide” for trials that studied their effect on NAFLD in patients with T2DM at any time up to February 2023.
Clinical trials, randomized controlled trials (RCT), and multicenter studies examining the effectiveness of GLP-1RA therapy on NAFLD in patients with T2DM were included. Studies that did not include subjects with T2DM were excluded. Only articles available in English and in full text were considered. To avoid omissions, other types of articles such as reviews, guidelines, systematic reviews, and meta-analyses were consulted. We also carefully examined the reference lists of the previously selected articles for other eligible and relevant articles. All the relevant information derived from the selected articles has been compiled into text or table form.
Relevant data were extracted by two independent authors who selected information related to the study characteristics (author, year of publication, country of origin, sample size, follow-up duration), the therapeutic regimen, diagnostic methods, and treatment outcomes (changes in liver biomarkers, steatosis and fibrosis indices, liver fat content, and histological modifications). An independent third author performed blinded checking of the data and disparities were resolved through consensus.

4. Search Results

Table 3 summarizes all data (clinical trials, RCT, multicenter studies, post hoc analyses) with at least a 12-week follow-up collected from patients with T2DM and NAFLD based on the use of exenatide, liraglutide, dulaglutide, and semaglutide in various doses compared with either placebo or other glucose-lowering agents, regardless of the population sample, primary endpoints, or investigation methods. No studies with efpeglenatide, lixisenatide, or albiglutide were found. All cited references summarized results of human studies, with both male and female adult patients included. Patients in various studies had different degrees of NAFLD, from steatosis to NASH or fibrosis. A total of 32 studies were found, including eight studies with exenatide, sixteen with liraglutide, six studies with semaglutide, and two studies with dulaglutide, all with positive results of improving NAFLD. Among all, liraglutide was by far the most investigated GLP-1RA for the management of NAFLD in patients with T2DM. In addition to the effects of ameliorating liver biomarkers, steatosis and fibrosis indices, liver fat content, and histological data, some studies also correlated the amounts of visceral adipose tissue and subcutaneous adipose tissue to NAFLD, and found improvements after the treatment with GLP-1RAs was implemented.

4.1. Effects of GLP-1RAs on Diabetes-Related Liver Disease

4.1.1. Changes in Serum Liver Enzymes

Exenatide was the first GLP-1RA to show, besides its favorable effects on HbA1c, insulin resistance, body weight, and blood pressure, an improvement of the hepatic enzymes in patients with T2DM [45,49]. Nowadays, exenatide is less used because of the lack of cardiorenal benefits [74] that were later demonstrated by the new generation of long-acting GLP-1RAs, such as liraglutide, dulaglutide, and semaglutide [75].
Liraglutide appears to be, for now, the most studied GLP-1RA in NAFLD, raising high hopes for its ability to prevent and treat the metabolic liver disease [76,77]. A meta-analysis of the LEAD program that included 4442 patients with T2DM assessed the effects of liraglutide on liver parameters after a 26-week therapy [78]. Liraglutide 1.8 mg per day improved liver enzymes, while smaller doses did not show significant effects after adjustments to body weight were applied [78].
The findings of a post hoc analysis of 1499 subjects from the AWARD trials with dulaglutide also showed a decrease in serum aminotransferases vs. placebo (−8.8 IU/L vs. −6.7 IU/L) in patients with T2DM and NASH after the administration of dulaglutide 1.5 mg per week [79].
Semaglutide is another GLP-1RA that has also gained interest in the treatment of steatohepatitis, as several studies in patients with T2DM and obesity reported beneficial changes in liver enzymes and inflammatory biomarkers such as high-sensitivity C-reactive protein [80,81].
An earlier meta-analysis of patients with T2DM and a high risk of NAFLD concluded that lixisenatide might have beneficial effects on liver enzymes compared to the control group [82].

4.1.2. Effects on Composite Indices of Hepatic Steatosis and Fibrosis

Improvement of noninvasive scores for NAFLD was observed in a trial by Gastaldelli et al., which assessed the efficacy of exenatide once weekly in association with dapagliflozin in T2DM patients. Fatty liver index (FLI), NAFLD liver fat score (NLFS), Fibrosis-4 (FIB-4) index, and NAFLD fibrosis score (NFS) decreased under the synergic effects of exenatide and dapagliflozin [42]. On the other hand, multiple glucose-lowering agents were able to improve NAFLD scores in patients with T2DM, independently of their ability to induce weight loss. In a retrospective study, Colosimo et al. demonstrated a correlation between the glycated hemoglobin level, which was used to identify good glycemic responders, and the changes in FLI and FIB-4, independently of the body mass index (BMI) or the nature of the antidiabetic medication, represented by either GLP-1RAs, dipeptidyl peptidase-4 (DPP-4) inhibitors, or SGLT2 inhibitors [21].
Gameil et al. performed a study that compared the effect of liraglutide and dulaglutide vs. conventional treatment on FLI and FIB-4 index in patients with T2DM complicated with NAFLD. Liraglutide and dulaglutide led to significant improvements in the FLI and FIB-4 values, with a greater change in the liraglutide group [52].

4.1.3. Changes in Liver Fat Content or Fibrosis Evaluated by Imaging Techniques

Several studies investigated, using imaging techniques, the GLP-1RAs effect on the reduction of liver fat content as a primary outcome in patients with T2DM and NAFLD. The reduction of liver fat content was assessed by either ultrasound [45,56,59,69] or magnetic-resonance-based imaging methods [43,64,72]. Liraglutide improved the intrahepatic fat content, leading to a decrease of approximately 60% from baseline when compared with metformin and gliclazide, along with reducing mean HbA1c levels from 8.9% to 5.9% and transaminase levels [59]. Another study with liraglutide pointed to a close correlation between the degree of hepatic steatosis evaluated by ultrasound and the extent of weight loss [60]. Moreover, other authors suggest that the diminution of liver steatosis due to the significant decreases in body weight and HbA1c values can also lead to a reduction of the hepatic inflammation [83].
However, mixed results are reported in this category. Smits et al. did not observe any valuable effect after a 12-week liraglutide or sitagliptin administration in T2DM, where proton magnetic resonance spectroscopy (1H-MRS) was used to measure hepatic fat and NFS, FIB-4 index, and AST to platelet ratio index (APRI) scores to assess fibrosis [62]. Another early study on T2DM patients randomized to insulin glargine or liraglutide for 12 weeks showed no significant change in liver proton density fat fraction measured by MRS [64], while dulaglutide was also not able to demonstrate a reduction of the intrahepatic fat estimated by transient elastography in NAFLD patients, probably due to the short duration of follow-up [84].
Contrariwise, Flint et al. found that semaglutide can determine more than 30% reduction in hepatic steatosis vs. placebo; however, no significant improvement of the liver stiffness assessed by magnetic resonance elastography (MRE) and of the magnetic resonance imaging-estimated proton density fat fraction (MRI-PDFF) was seen [70].
In short, the majority of studies acknowledge that GLP-1RAs decrease or even normalize the serum liver enzyme levels and reduce the liver fat content on imaging in people with NAFLD and T2DM [29,76].

4.1.4. Effects on Biopsy-Proven Histopathological Modifications

Liver biopsy and subsequent histologic examination represent the “gold standard” in diagnosing NAFLD [11]. There are only two important RCTs that have assessed the evolution of liver biopsy-proven histopathological abnormalities in diabetic and nondiabetic subjects with NASH [29,85]. The 48-week LEAN trial led by Armstrong and colleagues showed that liraglutide improved some features of NASH by delaying fibrosis progression when compared to placebo (39% vs. 9%, p = 0.019). However, the NAFLD Activity Score (NAS) did not display any significant changes during the trial [61]. The other RCT compared various doses of an atypical administration of once-daily subcutaneous semaglutide versus placebo for 72 weeks in patients with or without T2DM with histologic evidence of NASH. The study concluded that NASH resolution was dose-dependent, seen in 59% of those in the 0.4 mg/day semaglutide group (equivalent to 2.4 mg/week) versus 17% in the placebo group (p < 0.001) and due to the significant weight loss [71]. The treatment group exhibited a slower progression of fibrosis (4.9% vs. 18.8% in the placebo group), but no significant improvement in the severity of fibrosis [71]. Therefore, liraglutide and semaglutide achieved the histological resolution of NASH in 40 to 60% of participants, but with no influence on the overall fibrosis outcome [81,86]. The Effect of Semaglutide in Subjects with Noncirrhotic Nonalcoholic Steatohepatitis (ESSENCE) study is an ongoing phase 3 larger trial assessing the effects of semaglutide in NASH-related fibrosis [15,87].

4.1.5. Effects Assessed by Combined Investigations

Simeone et al. randomized patients with newly diagnosed T2DM and prediabetes to liraglutide 1.8 mg/dose or lifestyle counseling until achieving a target of approximately 7% less than the initial body weight. Both groups displayed significant comparable weight loss, improvement of the glycemic values, and reductions of the BMI, IL-1β level, and NAFLD degree assessed by magnetic resonance [54].
A 24-week, oral administration of semaglutide study assessed its efficacy and safety in T2DM complicated with NAFLD. A greater improvement was observed in the impaired liver function tests, hypertriglyceridemia, insulin resistance, and hepatic steatosis, led by the favorable effects on glycemic control and body weight. Controlled attenuation parameter (CAP) values significantly decreased from 344 to 279 dB/m at 24 weeks, the FIB-4 index decreased from 1.42 to 1.1, ferritin decreased from 4.1 ng/mL to 3.5 ng/mL, and hepatic enzymes were normalized at 24 weeks. However, liver stiffness measurement (LSM) values showed no significant changes in fibrosis after the 24 weeks of semaglutide treatment [67].
A systematic review and meta-analysis of 26 studies by Kumar et al. found that GLP-1RAs are associated with a significant reduction in ALT (mean difference (MD) −27.98, p = 0.04) and GGT (MD −40.65, p = 0.03), with no statistically significant influence upon AST values. They also demonstrated a significant improvement in liver steatosis (standard MD −2.53, p = 0.03), with insufficient data to support an effect on inflammation and fibrosis [88].
According to a meta-analysis of seven multinational RCTs assessing NAFLD by biopsy or imaging techniques, exenatide, and liraglutide proved to have consistent beneficial effects on serum liver enzymes, liver fat content, intraabdominal adipose tissue, and histological resolution of NASH [89].
At present, more large RCTs with biopsy-assessed outcomes are needed to accurately evaluate the effects of GLP-1RAs on various histopathological findings of NAFLD in patients with or without diabetes [77,83].

5. Potential Beneficial Mechanisms Underlying the Effects of GLP-1RAs in Diabetes-Related Liver Disease

5.1. Indirect Effects That Promote NAFLD Improvement

The mechanisms involved in NAFLD pathophysiology are not completely understood. Current data link the development of fatty liver disease to insulin resistance, de novo lipogenesis, oxidative stress, microbiome dysregulation, immune or cytokine-related anomalies, mitochondrial injury, and apoptosis, overlapping a pre-existing genetic predisposition [7,90]. This multifactorial determinism suggests that NAFLD therapy should be as personalized as possible to optimally interfere with its predominant underlying mechanisms in each individual.
GLP-1RAs appear to have pleiotropic effects that can improve the metabolic liver disease in patients with T2DM [91]. Their effects on NAFLD may be due to systemic benefits upon various metabolic pathways, and not to the activation of the local GLP-1 receptors, which are not expressed in the hepatic tissue in significant amounts [92]. Among such metabolic effects, the GLP-1RA class mainly includes the stimulation of insulin release and the inhibition of glucagon secretion, a decreased hepatic glucose production, an increased insulin sensitivity in hepatocytes and adipocytes, a delayed gastric emptying, and the suppression of appetite [61,93].
This perspective is supported by the results of earlier studies, which indicated that an intensive glycemic control may ameliorate liver fibrosis seen on serial hepatic biopsies in patients with T2DM [94].
The loss of body weight by appetite suppression and inducing satiety is one of the main intermediate steps leading to the beneficial effects of GLP-1RAs. One example is the Lira-NAFLD study, where liraglutide 1.2 mg per day induced a mean weight loss of 3.6 kg and a proportional decrease of the liver fat content from 17.3% to 11.9% [60].
GLP-1RAs also have antioxidant and anti-inflammatory properties leading to a significant diminution of the oxidative stress and inflammatory biomarkers, which could determine NASH amelioration and some protection against the natural evolution of the liver fibrosis [95].
The gut microbiome plays an important role in the occurrence and development of NAFLD and T2DM, and can therefore emerge as a forthcoming target in the therapy of these metabolic diseases [96,97]. Animal studies provide some evidence that liraglutide has a beneficial impact on components of the intestinal flora that are related to inflammation and glucolipid metabolism, thus improving the fatty liver disease [98]. Liraglutide also seems to have a beneficial effect on the human intestinal microbiome in studies on patients with NAFLD, where it decreased the inflammatory factors and improved the liver function and the adipose content [99].

5.2. Direct Mechanisms That Promote NAFLD Improvement

Another pathway that warrants future investigation is the involvement of glycerophospholipid perturbance in T2DM pathogenesis [100]. Du et al. tried to use the changes in metabolites after treatment with two GLP-1RAs (liraglutide and dulaglutide) to evaluate the metabolic remodeling effects [101]. After assessing various metabolites in patients with T2DM, many studies pointed out that some of them contribute to specific metabolic dysfunctions, such as insulin resistance, obesity, glucose impairment, and NAFLD. At present, the most preeminent class of T2DM-associated metabolites is represented by glycerophospholipids. The authors of the abovementioned study point out that, for now, there is no clear understanding yet of how GLP-1RAs reduce major adverse cardiovascular events (MACE) or how they ameliorate NASH, as these effects are not fully explained by the indirect beneficial effects on weight, lipid profile, or blood pressure. They hypothesize that such effects may be determined by the effects liraglutide and dulaglutide have on different metabolic pathways, among which the alteration of glycerophospholipid metabolism may play an important role in the pathophysiology of NAFLD [101].
Another study investigated the effect of liraglutide on thyroid resistance in the liver of T2DM patients [102]. Relevant findings suggest that the intrahepatic impairment of the thyroid hormone (TH) pathway due to thyroid hormone receptor beta (THRB) gene mutations may lead to fatty liver; TH resistance can thus take part in NAFLD development [29]. In the abovementioned study, liraglutide decreased thyroid hormone levels in the NAFLD group of patients with T2DM. The results suggest that a mechanism by which liraglutide can improve hepatic TH resistance by restoring the impaired THRB expression is likely possible [102].

6. Perspectives

The twincretin tirzepatide is a novel dual glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonist approved by the FDA in May 2022 for the treatment of T2DM, based on the results of the Study of Tirzepatide in Participants with T2DM Not Controlled with Diet and Exercise Alone (SURPASS) clinical trials [103,104]; benefits in patients with obesity were also proved later [105]. A randomized trial of patients with T2DM who received tirzepatide revealed significant decreases in biomarkers related to NASH such as AST/ALT, procollagen III, keratin 18, and an increase in adiponectin levels, which is believed to activate antifibrotic and antisteatogenic effects in the liver [106]. A once-weekly subcutaneous administration of tirzepatide for 72 weeks in obese patients without diabetes demonstrated weight losses of 15% with the 5 mg dose, 19.5% with the 10 mg dose, and 20.9% with the 15 mg dose vs. a 3.1% weight loss in the placebo group. This RCT also showed changes in the fasting insulin levels, lipid profile, and blood pressure, along with a specific reduction of the fat mass [105]. This may be the primary indirect pathway leading to NAFLD improvement, but additional investigations are needed. Moreover, the prediabetes group that received tirzepatide had regressed to normoglycemia [105]. Given these beneficial effects in the metabolic profile and intrahepatic fat, an RCT with tirzepatide in biopsy-proven NASH individuals with or without T2DM is currently ongoing and will complete its results in 2024 [107].
Cotadutide is a dual GLP-1/glucagon (Gcg) receptor dual-agonist investigated at present for T2DM, kidney disease, heart failure, and NASH [108]. Cotadutide exerts a unique dual agonism mode of action upon the glucose and lipid metabolism that appears to be superior compared with GLP-1 agonism alone, given the indirect effects of reduced inflammation and reversed fibrosis found in preclinical models, in addition to significant properties to induce HbA1c and weight reduction and to improve insulin resistance [109]. In a 54-week phase 2b study, cotadutide led to positive changes in body weight, serum ALT levels, and scores of hepatic steatosis and fibrosis (NFS and FIB-4) in patients with NASH and T2DM [110].
Another relevant trial is the one-year administration of liraglutide to 82 women with NAFLD and history of gestational diabetes, followed by a major reduction of steatosis vs. placebo [111].
Last, but not least, it must be mentioned that multiple ongoing or forthcoming large trials are aiming to investigate, using liver biopsy or MRI techniques, the effects of GLP-1RAs and their newly derived molecules on patients with T2DM and NAFLD (Table 4). Their results will no doubt shape the clinical guidelines for pharmacological therapy in T2DM and NAFLD patients.
The increased appearance of NAFLD in patients with T2DM in a large proportion of patients, added to the still-incomplete knowledge of the mechanical changes that these therapies induce in the liver, supports the need for more studies that seek effective treatments for both pathologies. A new contingent of drugs under development based on the principle of GLP-1 receptor agonism may represent a turning point in this field in the coming years.

7. Conclusions

Given the increasing worldwide incidence of both T2DM and NAFLD, this comprehensive review shows that in type 2 diabetic patients, GLP-1RAs may contribute to the reduction of liver fat content, induce NASH resolution, and reduce the progression of fibrosis.

Author Contributions

Conceptualization, G.-D.C., C.-M.L., G.Ș. and B.-M.M.; methodology, G.-D.C., C.-M.L. and E.-D.G.; validation, G.-D.C., C.-M.L. and B.-M.M.; formal analysis, G.Ș., C.M., E.-D.G. and A.O.; investigation, G.Ș., C.M., E.-D.G. and A.O.; resources, G.-D.C., C.-M.L. and E.-D.G.; writing—original draft preparation, G.-D.C., C.-M.L. and E.-D.G.; writing—review and editing, G.Ș., C.M., A.O. and B.-M.M.; visualization, G.-D.C. and C.-M.L.; supervision, B.-M.M.; project administration, C.-M.L. and E.-D.G. All authors had equal contributions to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cotter, T.G.; Rinella, M. Nonalcoholic Fatty Liver Disease 2020: The State of the Disease. Gastroenterology 2020, 158, 1851–1864. [Google Scholar] [CrossRef]
  2. IDF Diabetes Atlas. Available online: https://idf.org/e-library/epidemiology-research/diabetes-atlas.html?id=171 (accessed on 20 March 2023).
  3. Kotronen, A.; Yki-Järvinen, H. Fatty Liver: A Novel Component of the Metabolic Syndrome. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 27–38. [Google Scholar] [CrossRef] [PubMed]
  4. Younossi, Z.M.; Henry, L. Epidemiology of Non-Alcoholic Fatty Liver Disease and Hepatocellular Carcinoma. JHEP Rep. 2021, 3, 100305. [Google Scholar] [CrossRef]
  5. Le, M.H.; Yeo, Y.H.; Zou, B.; Barnet, S.; Henry, L.; Cheung, R.; Nguyen, M.H. Forecasted 2040 Global Prevalence of Nonalcoholic Fatty Liver Disease Using Hierarchical Bayesian Approach. Clin. Mol. Hepatol. 2022, 28, 841–850. [Google Scholar] [CrossRef] [PubMed]
  6. 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]
  7. Gariani, K.; Jornayvaz, F.R. Pathophysiology of NASH in Endocrine Diseases. Endocr. Connect. 2021, 10, R52–R65. [Google Scholar] [CrossRef] [PubMed]
  8. Xia, M.-F.; Bian, H.; Gao, X. NAFLD and Diabetes: Two Sides of the Same Coin? Rationale for Gene-Based Personalized NAFLD Treatment. Front. Pharmacol. 2019, 10, 877. [Google Scholar] [CrossRef]
  9. Tilg, H.; Moschen, A.R.; Roden, M. NAFLD and Diabetes Mellitus. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 32–42. [Google Scholar] [CrossRef]
  10. Hazlehurst, J.M.; Woods, C.; Marjot, T.; Cobbold, J.F.; Tomlinson, J.W. Non-Alcoholic Fatty Liver Disease and Diabetes. Metabolism 2016, 65, 1096–1108. [Google Scholar] [CrossRef] [PubMed]
  11. 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]
  12. Cusi, K.; Isaacs, S.; Barb, D.; Basu, R.; Caprio, S.; Garvey, W.T.; Kashyap, S.; Mechanick, J.I.; Mouzaki, M.; Nadolsky, K.; et al. American Association of Clinical Endocrinology Clinical Practice Guideline for the Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Primary Care and Endocrinology Clinical Settings: Co-Sponsored by the American Association for the Study of Liver Diseases (AASLD). Endocr. Pract. 2022, 28, 528–562. [Google Scholar] [CrossRef]
  13. Nauck, M.A.; Quast, D.R.; Wefers, J.; Meier, J.J. GLP-1 Receptor Agonists in the Treatment of Type 2 Diabetes—State-of-the-Art. Mol. Metab. 2021, 46, 101102. [Google Scholar] [CrossRef]
  14. Francque, S.M.; Marchesini, G.; Kautz, A.; Walmsley, M.; Dorner, R.; Lazarus, J.V.; Zelber-Sagi, S.; Hallsworth, K.; Busetto, L.; Frühbeck, G.; et al. Non-Alcoholic Fatty Liver Disease: A Patient Guideline. JHEP Rep. 2021, 3, 100322. [Google Scholar] [CrossRef] [PubMed]
  15. Rinella, M.E.; Neuschwander-Tetri, B.A.; Siddiqui, M.S.; Abdelmalek, M.F.; Caldwell, S.; Barb, D.; Kleiner, D.E.; Loomba, R. AASLD Practice Guidance on the Clinical Assessment and Management of Nonalcoholic Fatty Liver Disease. Hepatology 2023, 77, 1797–1835. [Google Scholar] [CrossRef] [PubMed]
  16. Seghieri, M.; Christensen, A.S.; Andersen, A.; Solini, A.; Knop, F.K.; Vilsbøll, T. Future Perspectives on GLP-1 Receptor Agonists and GLP-1/Glucagon Receptor Co-Agonists in the Treatment of NAFLD. Front. Endocrinol. 2018, 9, 649. [Google Scholar] [CrossRef] [PubMed]
  17. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the Management of Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2016, 64, 1388–1402. [Google Scholar] [CrossRef]
  18. Hydes, T.; Alam, U.; Cuthbertson, D.J. The Impact of Macronutrient Intake on Non-Alcoholic Fatty Liver Disease (NAFLD): Too Much Fat, Too Much Carbohydrate, or Just Too Many Calories? Front. Nutr. 2021, 8, 640557. [Google Scholar] [CrossRef]
  19. Neuschwander-Tetri, B.A. Therapeutic Landscape for NAFLD in 2020. Gastroenterology 2020, 158, 1984–1998.e3. [Google Scholar] [CrossRef]
  20. Diaconu, C.-T.; Guja, C. Nonalcoholic Fatty Liver Disease and Its Complex Relation with Type 2 Diabetes Mellitus-From Prevalence to Diagnostic Approach and Treatment Strategies. J. Clin. Med. 2022, 11, 5144. [Google Scholar] [CrossRef]
  21. Colosimo, S.; Tan, G.D.; Petroni, M.L.; Marchesini, G.; Tomlinson, J.W. Improved Glycaemic Control in Patients with Type 2 Diabetes Has a Beneficial Impact on NAFLD, Independent of Change in BMI or Glucose Lowering Agent. Nutr. Metab. Cardiovasc. Dis. 2023, 33, 640–648. [Google Scholar] [CrossRef]
  22. Binet, Q.; Loumaye, A.; Preumont, V.; Thissen, J.P.; Hermans, M.P.; Lanthier, N. Non-Invasive Screening, Staging and Management of Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) in Type 2 Diabetes Mellitus Patients: What Do We Know so Far? Acta Gastroenterol. Belg. 2022, 85, 346–357. [Google Scholar] [CrossRef]
  23. Davies, M.J.; Aroda, V.R.; Collins, B.S.; Gabbay, R.A.; Green, J.; Maruthur, N.M.; Rosas, S.E.; Del Prato, S.; Mathieu, C.; Mingrone, G.; et al. Management of Hyperglycemia in Type 2 Diabetes, 2022. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2022, 45, 2753–2786. [Google Scholar] [CrossRef]
  24. Hinnen, D. Glucagon-Like Peptide 1 Receptor Agonists for Type 2 Diabetes. Diabetes Spectr. 2017, 30, 202–210. [Google Scholar] [CrossRef]
  25. Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.D.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989–1002. [Google Scholar] [CrossRef]
  26. Neeland, I.J.; Marso, S.P.; Ayers, C.R.; Lewis, B.; Oslica, R.; Francis, W.; Rodder, S.; Pandey, A.; Joshi, P.H. Effects of Liraglutide on Visceral and Ectopic Fat in Adults with Overweight and Obesity at High Cardiovascular Risk: A Randomised, Double-Blind, Placebo-Controlled, Clinical Trial. Lancet Diabetes Endocrinol. 2021, 9, 595–605. [Google Scholar] [CrossRef] [PubMed]
  27. Sattar, N.; Lee, M.M.Y.; Kristensen, S.L.; Branch, K.R.H.; Del Prato, S.; Khurmi, N.S.; Lam, C.S.P.; Lopes, R.D.; McMurray, J.J.V.; Pratley, R.E.; et al. Cardiovascular, Mortality, and Kidney Outcomes with GLP-1 Receptor Agonists in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis of Randomised Trials. Lancet Diabetes Endocrinol. 2021, 9, 653–662. [Google Scholar] [CrossRef] [PubMed]
  28. Marsico, F.; Paolillo, S.; Gargiulo, P.; Bruzzese, D.; Dell’Aversana, S.; Esposito, I.; Renga, F.; Esposito, L.; Marciano, C.; Dellegrottaglie, S.; et al. Effects of Glucagon-like Peptide-1 Receptor Agonists on Major Cardiovascular Events in Patients with Type 2 Diabetes Mellitus with or without Established Cardiovascular Disease: A Meta-Analysis of Randomized Controlled Trials. Eur. Heart J. 2020, 41, 3346–3358. [Google Scholar] [CrossRef]
  29. Patel Chavez, C.; Cusi, K.; Kadiyala, S. The Emerging Role of Glucagon-like Peptide-1 Receptor Agonists for the Management of NAFLD. J. Clin. Endocrinol. Metab. 2022, 107, 29–38. [Google Scholar] [CrossRef] [PubMed]
  30. Gillenwater, B.A.; Wakefield, A.N.; Triboletti, J.; Gonzalvo, J.D.; Meredith, A.H. GLP-101: A Diabetes Educator’s Guide to Glucagon-Like-Peptide-1 Receptor Agonists. AADE Pract. 2019, 7, 32–41. [Google Scholar] [CrossRef]
  31. Brown, E.; Heerspink, H.J.L.; Cuthbertson, D.J.; Wilding, J.P.H. SGLT2 Inhibitors and GLP-1 Receptor Agonists: Established and Emerging Indications. Lancet 2021, 398, 262–276. [Google Scholar] [CrossRef]
  32. Kolterman, O.G.; Kim, D.D.; Shen, L.; Ruggles, J.A.; Nielsen, L.L.; Fineman, M.S.; Baron, A.D. Pharmacokinetics, Pharmacodynamics, and Safety of Exenatide in Patients with Type 2 Diabetes Mellitus. Am. J. Health Syst. Pharm. 2005, 62, 173–181. [Google Scholar] [CrossRef] [PubMed]
  33. Becker, R.H.A.; Stechl, J.; Steinstraesser, A.; Golor, G.; Pellissier, F. Lixisenatide Reduces Postprandial Hyperglycaemia via Gastrostatic and Insulinotropic Effects. Diabetes Metab. Res. Rev. 2015, 31, 610–618. [Google Scholar] [CrossRef] [PubMed]
  34. Matthews, J.E.; Stewart, M.W.; De Boever, E.H.; Dobbins, R.L.; Hodge, R.J.; Walker, S.E.; Holland, M.C.; Bush, M.A. Albiglutide Study Group Pharmacodynamics, Pharmacokinetics, Safety, and Tolerability of Albiglutide, a Long-Acting Glucagon-like Peptide-1 Mimetic, in Patients with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2008, 93, 4810–4817. [Google Scholar] [CrossRef] [PubMed]
  35. Damholt, B.; Golor, G.; Wierich, W.; Pedersen, P.; Ekblom, M.; Zdravkovic, M. An Open-Label, Parallel Group Study Investigating the Effects of Age and Gender on the Pharmacokinetics of the Once-Daily Glucagon-like Peptide-1 Analogue Liraglutide. J. Clin. Pharmacol. 2006, 46, 635–641. [Google Scholar] [CrossRef] [PubMed]
  36. Geiser, J.S.; Heathman, M.A.; Cui, X.; Martin, J.; Loghin, C.; Chien, J.Y.; de la Peña, A. Clinical Pharmacokinetics of Dulaglutide in Patients with Type 2 Diabetes: Analyses of Data from Clinical Trials. Clin. Pharmacokinet. 2016, 55, 625–634. [Google Scholar] [CrossRef]
  37. Marbury, T.C.; Flint, A.; Jacobsen, J.B.; Derving Karsbøl, J.; Lasseter, K. Pharmacokinetics and Tolerability of a Single Dose of Semaglutide, a Human Glucagon-Like Peptide-1 Analog, in Subjects With and Without Renal Impairment. Clin. Pharmacokinet. 2017, 56, 1381–1390. [Google Scholar] [CrossRef]
  38. Granhall, C.; Donsmark, M.; Blicher, T.M.; Golor, G.; Søndergaard, F.L.; Thomsen, M.; Bækdal, T.A. Safety and Pharmacokinetics of Single and Multiple Ascending Doses of the Novel Oral Human GLP-1 Analogue, Oral Semaglutide, in Healthy Subjects and Subjects with Type 2 Diabetes. Clin. Pharmacokinet. 2019, 58, 781–791. [Google Scholar] [CrossRef]
  39. Napoli, R.; Avogaro, A.; Formoso, G.; Piro, S.; Purrello, F.; Targher, G.; Consoli, A. Beneficial Effects of Glucagon-like Peptide 1 Receptor Agonists on Glucose Control, Cardiovascular Risk Profile, and Non-Alcoholic Fatty Liver Disease. An Expert Opinion of the Italian Diabetes Society. Nutr. Metab. Cardiovasc. Dis. 2021, 31, 3257–3270. [Google Scholar] [CrossRef]
  40. Eslam, M.; Sarin, S.K.; Wong, V.W.-S.; 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] [CrossRef]
  41. Recommendations|Non-Alcoholic Fatty Liver Disease (NAFLD): Assessment and Management|Guidance|NICE. Available online: https://www.nice.org.uk/guidance/ng49/chapter/Recommendations (accessed on 20 March 2023).
  42. Gastaldelli, A.; Repetto, E.; Guja, C.; Hardy, E.; Han, J.; Jabbour, S.A.; Ferrannini, E. Exenatide and Dapagliflozin Combination Improves Markers of Liver Steatosis and Fibrosis in Patients with Type 2 Diabetes. Diabetes Obes. Metab. 2020, 22, 393–403. [Google Scholar] [CrossRef]
  43. Liu, L.; Yan, H.; Xia, M.; Zhao, L.; Lv, M.; Zhao, N.; Rao, S.; Yao, X.; Wu, W.; Pan, B.; et al. Efficacy of Exenatide and Insulin Glargine on Nonalcoholic Fatty Liver Disease in Patients with Type 2 Diabetes. Diabetes Metab. Res. Rev. 2020, 36, e3292. [Google Scholar] [CrossRef] [PubMed]
  44. Dutour, A.; Abdesselam, I.; Ancel, P.; Kober, F.; Mrad, G.; Darmon, P.; Ronsin, O.; Pradel, V.; Lesavre, N.; Martin, J.C.; et al. Exenatide Decreases Liver Fat Content and Epicardial Adipose Tissue in Patients with Obesity and Type 2 Diabetes: A Prospective Randomized Clinical Trial Using Magnetic Resonance Imaging and Spectroscopy. Diabetes Obes. Metab. 2016, 18, 882–891. [Google Scholar] [CrossRef]
  45. Shao, N.; Kuang, H.Y.; Hao, M.; Gao, X.Y.; Lin, W.J.; Zou, W. Benefits of Exenatide on Obesity and Non-Alcoholic Fatty Liver Disease with Elevated Liver Enzymes in Patients with Type 2 Diabetes. Diabetes Metab. Res. Rev. 2014, 30, 521–529. [Google Scholar] [CrossRef] [PubMed]
  46. Fan, H.; Pan, Q.; Xu, Y.; Yang, X. Exenatide Improves Type 2 Diabetes Concomitant with Non-Alcoholic Fatty Liver Disease. Arq. Bras. Endocrinol. Metabol. 2013, 57, 702–708. [Google Scholar] [CrossRef] [PubMed]
  47. Cuthbertson, D.J.; Irwin, A.; Gardner, C.J.; Daousi, C.; Purewal, T.; Furlong, N.; Goenka, N.; Thomas, E.L.; Adams, V.L.; Pushpakom, S.P.; et al. Improved Glycaemia Correlates with Liver Fat Reduction in Obese, Type 2 Diabetes, Patients given Glucagon-like Peptide-1 (GLP-1) Receptor Agonists. PLoS ONE 2012, 7, e50117. [Google Scholar] [CrossRef]
  48. Sathyanarayana, P.; Jogi, M.; Muthupillai, R.; Krishnamurthy, R.; Samson, S.L.; Bajaj, M. Effects of Combined Exenatide and Pioglitazone Therapy on Hepatic Fat Content in Type 2 Diabetes. Obesity 2011, 19, 2310–2315. [Google Scholar] [CrossRef]
  49. Buse, J.B.; Klonoff, D.C.; Nielsen, L.L.; Guan, X.; Bowlus, C.L.; Holcombe, J.H.; Maggs, D.G.; Wintle, M.E. Metabolic Effects of Two Years of Exenatide Treatment on Diabetes, Obesity, and Hepatic Biomarkers in Patients with Type 2 Diabetes: An Interim Analysis of Data from the Open-Label, Uncontrolled Extension of Three Double-Blind, Placebo-Controlled Trials. Clin. Ther. 2007, 29, 139–153. [Google Scholar] [CrossRef]
  50. Tan, Y.; Zhen, Q.; Ding, X.; Shen, T.; Liu, F.; Wang, Y.; Zhang, Q.; Lin, R.; Chen, L.; Peng, Y.; et al. Association between Use of Liraglutide and Liver Fibrosis in Patients with Type 2 Diabetes. Front. Endocrinol. 2022, 13, 935180. [Google Scholar] [CrossRef]
  51. Bizino, M.B.; Jazet, I.M.; de Heer, P.; van Eyk, H.J.; Dekkers, I.A.; Rensen, P.C.N.; Paiman, E.H.M.; Lamb, H.J.; Smit, J.W. Placebo-Controlled Randomised Trial with Liraglutide on Magnetic Resonance Endpoints in Individuals with Type 2 Diabetes: A Pre-Specified Secondary Study on Ectopic Fat Accumulation. Diabetologia 2020, 63, 65–74. [Google Scholar] [CrossRef]
  52. Gameil, M.A.; Rozaik, S.E.; Elsebaie, A.; Marzouk, R. Influence of Liraglutide, Dulaglutide versus Conventional Treatment on Fatty Liver Index and Fibrosis-4 Score in Egyptian Patients with Type 2 Diabetes Mellitus and Non-Alcoholic Fatty Liver Disease. Med. J. Viral Hepat. 2020, 5, 25–32. [Google Scholar] [CrossRef]
  53. Guo, W.; Tian, W.; Lin, L.; Xu, X. Liraglutide or Insulin Glargine Treatments Improves Hepatic Fat in Obese Patients with Type 2 Diabetes and Nonalcoholic Fatty Liver Disease in Twenty-Six Weeks: A Randomized Placebo-Controlled Trial. Diabetes Res. Clin. Pract. 2020, 170, 108487. [Google Scholar] [CrossRef]
  54. Simeone, P.G.; Costantino, S.; Tripaldi, R.; Liani, R.; Ciotti, S.; Tartaro, A.; Guagnano, M.T.; Cosentino, F.; Consoli, A.; Paneni, F.; et al. Baseline Interleukin1beta Expression in Peripheral Blood Monocytes Predicts the Extent of Weight Loss and Nonalcoholic Fatty Liver Improvement in Obese Subjects with Prediabetes or Type 2 Diabetes. Eur. Heart J. 2020, 41, ehaa946.3033. [Google Scholar] [CrossRef]
  55. 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]
  56. Tian, F.; Zheng, Z.; Zhang, D.; He, S.; Shen, J. Efficacy of Liraglutide in Treating Type 2 Diabetes Mellitus Complicated with Non-Alcoholic Fatty Liver Disease. Biosci. Rep. 2018, 38, BSR20181304. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, Z.; Qi, Y.; Kong, W.; Jin, Q.; Wang, X.; Dong, Y.; Wang, Y.; Li, H. Efficacy and Clinical Value of Liraglutide for Treatment of Diabetes Mellitus Complicated by Non-Alcoholic Fatty Liver Disease. Med. Sci. Monit. 2018, 24, 7399–7404. [Google Scholar] [CrossRef]
  58. Bouchi, R.; Nakano, Y.; Fukuda, T.; Takeuchi, T.; Murakami, M.; Minami, I.; Izumiyama, H.; Hashimoto, K.; Yoshimoto, T.; Ogawa, Y. Reduction of Visceral Fat by Liraglutide Is Associated with Ameliorations of Hepatic Steatosis, Albuminuria, and Micro-Inflammation in Type 2 Diabetic Patients with Insulin Treatment: A Randomized Control Trial. Endocr. J. 2017, 64, 269–281. [Google Scholar] [CrossRef]
  59. Feng, W.; Gao, C.; Bi, Y.; Wu, M.; Li, P.; Shen, S.; Chen, W.; Yin, T.; Zhu, D. Randomized Trial Comparing the Effects of Gliclazide, Liraglutide, and Metformin on Diabetes with Non-Alcoholic Fatty Liver Disease. J. Diabetes 2017, 9, 800–809. [Google Scholar] [CrossRef] [PubMed]
  60. Petit, J.-M.; Cercueil, J.-P.; Loffroy, R.; Denimal, D.; Bouillet, B.; Fourmont, C.; Chevallier, O.; Duvillard, L.; Vergès, 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]
  61. 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]
  62. Smits, M.M.; Tonneijck, L.; Muskiet, M.H.A.; Kramer, M.H.H.; Pouwels, P.J.W.; Pieters-van den Bos, I.C.; Hoekstra, T.; Diamant, M.; van Raalte, D.H.; Cahen, D.L. Twelve Week Liraglutide or Sitagliptin Does Not Affect Hepatic Fat in Type 2 Diabetes: A Randomised Placebo-Controlled Trial. Diabetologia 2016, 59, 2588–2593. [Google Scholar] [CrossRef]
  63. Vanderheiden, A.; Harrison, L.B.; Warshauer, J.T.; Adams-Huet, B.; Li, X.; Yuan, Q.; Hulsey, K.; Dimitrov, I.; Yokoo, T.; Jaster, A.W.; et al. Mechanisms of Action of Liraglutide in Patients with Type 2 Diabetes Treated With High-Dose Insulin. J. Clin. Endocrinol. Metab. 2016, 101, 1798–1806. [Google Scholar] [CrossRef] [PubMed]
  64. Tang, A.; Rabasa-Lhoret, R.; Castel, H.; Wartelle-Bladou, C.; Gilbert, G.; Massicotte-Tisluck, K.; Chartrand, G.; Olivié, D.; Julien, A.-S.; de Guise, J.; et al. Effects of Insulin Glargine and Liraglutide Therapy on Liver Fat as Measured by Magnetic Resonance in Patients With Type 2 Diabetes: A Randomized Trial. Diabetes Care 2015, 38, 1339–1346. [Google Scholar] [CrossRef] [PubMed]
  65. Ohki, T.; Isogawa, A.; Iwamoto, M.; Ohsugi, M.; Yoshida, H.; Toda, N.; Tagawa, K.; Omata, M.; Koike, K. The Effectiveness of Liraglutide in Nonalcoholic Fatty Liver Disease Patients with Type 2 Diabetes Mellitus Compared to Sitagliptin and Pioglitazone. Sci. World J. 2012, 2012, e496453. [Google Scholar] [CrossRef]
  66. Carretero-Gómez, J.; Carrasco-Sánchez, F.J.; Fernández-Rodríguez, J.M.; Casado-Escribano, P.; Miramontes-González, J.P.; Seguí-Ripoll, J.M.; Ena, J.; Arévalo-Lorido, J.C. Effect of Semaglutide on Fatty Liver Disease Biomarkers in Patients with Diabetes and Obesity. Rev. Clín. Española (Engl. Ed.) 2023, 223, 134–143. [Google Scholar] [CrossRef]
  67. Arai, T.; Atsukawa, M.; Tsubota, A.; Ono, H.; Kawano, T.; Yoshida, Y.; Okubo, T.; Hayama, K.; Nakagawa-Iwashita, A.; Itokawa, N.; et al. Efficacy and Safety of Oral Semaglutide in Patients with Non-Alcoholic Fatty Liver Disease Complicated by Type 2 Diabetes Mellitus: A Pilot Study. JGH Open 2022, 6, 503–511. [Google Scholar] [CrossRef] [PubMed]
  68. Ding, L.P.; Liu, S.K. The Effect of Semaglutide on Glycolipid Metabolism in Type 2 Diabetes Mellitus and Non-Alcoholic Fatty Liver Disease. Indian J. Pharm. Sci. 2022, 84, 171–175. [Google Scholar] [CrossRef]
  69. Volpe, S.; Lisco, G.; Fanelli, M.; Racaniello, D.; Colaianni, V.; Triggiani, D.; Donghia, R.; Crudele, L.; Rinaldi, R.; Sabbà, C.; et al. Once-Weekly Subcutaneous Semaglutide Improves Fatty Liver Disease in Patients with Type 2 Diabetes: A 52-Week Prospective Real-Life Study. Nutrients 2022, 14, 4673. [Google Scholar] [CrossRef]
  70. Flint, A.; Andersen, G.; Hockings, P.; Johansson, L.; Morsing, A.; Sundby Palle, M.; Vogl, T.; Loomba, R.; Plum-Mörschel, L. Randomised Clinical Trial: Semaglutide versus Placebo Reduced Liver Steatosis but Not Liver Stiffness in Subjects with Non-Alcoholic Fatty Liver Disease Assessed by Magnetic Resonance Imaging. Aliment. Pharmacol. Ther. 2021, 54, 1150–1161. [Google Scholar] [CrossRef]
  71. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.-S.; Harrison, S.A. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124. [Google Scholar] [CrossRef]
  72. Kuchay, M.S.; Krishan, S.; Mishra, S.K.; Choudhary, N.S.; Singh, M.K.; Wasir, J.S.; Kaur, P.; Gill, H.K.; Bano, T.; Farooqui, K.J.; et al. Effect of Dulaglutide on Liver Fat in Patients with Type 2 Diabetes and NAFLD: Randomised Controlled Trial (D-LIFT Trial). Diabetologia 2020, 63, 2434–2445. [Google Scholar] [CrossRef]
  73. Bogomolov, P.O.; Bueverov, A.O.; Dreval, A.V.; Nechaeva, O.A.; Mayorov, A.Y.; Mishina, E.E.; Fedosina, E.A.; Koblov, S.V.; Sumtsova, O.V.; Jallow, A.; et al. Preliminary results of dulaglutide treatment in patients with non-alcoholic fatty liver disease (DEMOS study). Exp. Clin. Gastroenterol. 2019, 9, 4–10. [Google Scholar] [CrossRef]
  74. Giugliano, D.; Scappaticcio, L.; Longo, M.; Caruso, P.; Maiorino, M.I.; Bellastella, G.; Ceriello, A.; Chiodini, P.; Esposito, K. GLP-1 Receptor Agonists and Cardiorenal Outcomes in Type 2 Diabetes: An Updated Meta-Analysis of Eight CVOTs. Cardiovasc. Diabetol. 2021, 20, 189. [Google Scholar] [CrossRef]
  75. Boyle, J.G.; Livingstone, R.; Petrie, J.R. Cardiovascular Benefits of GLP-1 Agonists in Type 2 Diabetes: A Comparative Review. Clin. Sci. 2018, 132, 1699–1709. [Google Scholar] [CrossRef]
  76. Cusi, K. Incretin-Based Therapies for the Management of Nonalcoholic Fatty Liver Disease in Patients with Type 2 Diabetes. Hepatology 2019, 69, 2318–2322. [Google Scholar] [CrossRef]
  77. Lian, J.; Fu, J. Efficacy of Various Hypoglycemic Agents in the Treatment of Patients With Nonalcoholic Liver Disease With or Without Diabetes: A Network Meta-Analysis. Front. Endocrinol. 2021, 12, 649018. [Google Scholar] [CrossRef] [PubMed]
  78. Armstrong, M.J.; Houlihan, D.D.; Rowe, I.A.; Clausen, W.H.O.; Elbrønd, B.; Gough, S.C.L.; Tomlinson, J.W.; Newsome, P.N. Safety and Efficacy of Liraglutide in Patients with Type 2 Diabetes and Elevated Liver Enzymes: Individual Patient Data Meta-Analysis of the LEAD Program. Aliment. Pharmacol. Ther. 2013, 37, 234–242. [Google Scholar] [CrossRef]
  79. Cusi, K.; Sattar, N.; García-Pérez, L.-E.; Pavo, I.; Yu, M.; Robertson, K.E.; Karanikas, C.A.; Haupt, A. Dulaglutide Decreases Plasma Aminotransferases in People with Type 2 Diabetes in a Pattern Consistent with Liver Fat Reduction: A Post Hoc Analysis of the Award Programme. Diabet. Med. 2018, 35, 1434–1439. [Google Scholar] [CrossRef]
  80. Newsome, P.; Francque, S.; Harrison, S.; Ratziu, V.; Van Gaal, L.; Calanna, S.; Hansen, M.; Linder, M.; Sanyal, A. Effect of Semaglutide on Liver Enzymes and Markers of Inflammation in Subjects with Type 2 Diabetes and/or Obesity. Aliment. Pharmacol. Ther. 2019, 50, 193–203. [Google Scholar] [CrossRef] [PubMed]
  81. Ahmed, N.R.; Kulkarni, V.V.; Pokhrel, S.; Akram, H.; Abdelgadir, A.; Chatterjee, A.; Khan, S.; Ahmed, N.R.; Kulkarni, V.V.; Pokhrel, S.; et al. Comparing the Efficacy and Safety of Obeticholic Acid and Semaglutide in Patients with Non-Alcoholic Fatty Liver Disease: A Systematic Review. Cureus 2022, 14, e24829. [Google Scholar] [CrossRef] [PubMed]
  82. Gluud, L.L.; Knop, F.K.; Vilsbøll, T. Effects of Lixisenatide on Elevated Liver Transaminases: Systematic Review with Individual Patient Data Meta-Analysis of Randomised Controlled Trials on Patients with Type 2 Diabetes. BMJ Open 2014, 4, e005325. [Google Scholar] [CrossRef]
  83. Ghosal, S.; Datta, D.; Sinha, B. A Meta-Analysis of the Effects of Glucagon-like-Peptide 1 Receptor Agonist (GLP1-RA) in Nonalcoholic Fatty Liver Disease (NAFLD) with Type 2 Diabetes (T2D). Sci. Rep. 2021, 11, 22063. [Google Scholar] [CrossRef]
  84. Seko, Y.; Sumida, Y.; Tanaka, S.; Mori, K.; Taketani, H.; Ishiba, H.; Hara, T.; Okajima, A.; Umemura, A.; Nishikawa, T.; et al. Effect of 12-Week Dulaglutide Therapy in Japanese Patients with Biopsy-Proven Non-Alcoholic Fatty Liver Disease and Type 2 Diabetes Mellitus. Hepatol. Res. 2017, 47, 1206–1211. [Google Scholar] [CrossRef] [PubMed]
  85. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Cusi, K.; Hilliard, M.E.; Isaacs, D.; et al. 4. Comprehensive Medical Evaluation and Assessment of Comorbidities: Standards of Care in Diabetes—2023. Diabetes Care 2022, 46, S49–S67. [Google Scholar] [CrossRef]
  86. Barritt, A.S.; Marshman, E.; Noureddin, M. Review Article: Role of Glucagon-like Peptide-1 Receptor Agonists in Non-Alcoholic Steatohepatitis, Obesity and Diabetes-What Hepatologists Need to Know. Aliment. Pharmacol. Ther. 2022, 55, 944–959. [Google Scholar] [CrossRef] [PubMed]
  87. Novo Nordisk A/S. The Effect of Semaglutide in Subjects with Non-Cirrhotic Non-Alcoholic Steatohepatitis; ClinicalTrials.gov: Bethesda, MD, USA, 2022. [Google Scholar]
  88. Kumar, J.; Memon, R.S.; Shahid, I.; Rizwan, T.; Zaman, M.; Menezes, R.G.; Kumar, S.; Siddiqi, T.J.; Usman, M.S. Antidiabetic Drugs and Non-Alcoholic Fatty Liver Disease: A Systematic Review, Meta-Analysis and Evidence Map. Dig. Liver Dis. 2021, 53, 44–51. [Google Scholar] [CrossRef] [PubMed]
  89. Mantovani, A.; Beatrice, G.; Petracca, G.; Pampagnin, F.; Sandri, D.; Targher, G. GLP-1 Receptor Agonists for NAFLD Treatment in Patients with and without Type 2 Diabetes: An Updated Meta-Analysis. Explor. Med. 2020, 1, 108–123. [Google Scholar] [CrossRef]
  90. Caturano, A.; Acierno, C.; Nevola, R.; Pafundi, P.C.; Galiero, R.; Rinaldi, L.; Salvatore, T.; Adinolfi, L.E.; Sasso, F.C. Non-Alcoholic Fatty Liver Disease: From Pathogenesis to Clinical Impact. Processes 2021, 9, 135. [Google Scholar] [CrossRef]
  91. Marušić, M.; Paić, M.; Knobloch, M.; Liberati Pršo, A.-M. NAFLD, Insulin Resistance, and Diabetes Mellitus Type 2. Can. J. Gastroenterol. Hepatol. 2021, 2021, 6613827. [Google Scholar] [CrossRef]
  92. Müller, T.D.; Finan, B.; Bloom, S.R.; D’Alessio, D.; Drucker, D.J.; Flatt, P.R.; Fritsche, A.; Gribble, F.; Grill, H.J.; Habener, J.F.; et al. Glucagon-like Peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef]
  93. Ferguson, D.; Finck, B.N. Emerging Therapeutic Approaches for the Treatment of NAFLD and Type 2 Diabetes Mellitus. Nat. Rev. Endocrinol. 2021, 17, 484–495. [Google Scholar] [CrossRef]
  94. Hamaguchi, E.; Takamura, T.; Sakurai, M.; Mizukoshi, E.; Zen, Y.; Takeshita, Y.; Kurita, S.; Arai, K.; Yamashita, T.; Sasaki, M.; et al. Histological Course of Nonalcoholic Fatty Liver Disease in Japanese Patients: Tight Glycemic Control, Rather than Weight Reduction, Ameliorates Liver Fibrosis. Diabetes Care 2010, 33, 284–286. [Google Scholar] [CrossRef]
  95. Bray, J.J.H.; Foster-Davies, H.; Salem, A.; Hoole, A.L.; Obaid, D.R.; Halcox, J.P.J.; Stephens, J.W. Glucagon-like Peptide-1 Receptor Agonists Improve Biomarkers of Inflammation and Oxidative Stress: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Diabetes Obes. Metab. 2021, 23, 1806–1822. [Google Scholar] [CrossRef] [PubMed]
  96. Leung, H.; Long, X.; Ni, Y.; Qian, L.; Nychas, E.; Siliceo, S.L.; Pohl, D.; Hanhineva, K.; Liu, Y.; Xu, A.; et al. Risk Assessment with Gut Microbiome and Metabolite Markers in NAFLD Development. Sci. Transl. Med. 2022, 14, eabk0855. [Google Scholar] [CrossRef] [PubMed]
  97. Pezzino, S.; Sofia, M.; Faletra, G.; Mazzone, C.; Litrico, G.; La Greca, G.; Latteri, S. Gut-Liver Axis and Non-Alcoholic Fatty Liver Disease: A Vicious Circle of Dysfunctions Orchestrated by the Gut Microbiome. Biology 2022, 11, 1622. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, Q.; Cai, B.-Y.; Zhu, L.-X.; Xin, X.; Wang, X.; An, Z.-M.; Li, S.; Hu, Y.-Y.; Feng, Q. Liraglutide Modulates Gut Microbiome and Attenuates Nonalcoholic Fatty Liver in Db/Db Mice. Life Sci. 2020, 261, 118457. [Google Scholar] [CrossRef] [PubMed]
  99. Ying, X.; Rongjiong, Z.; Kahaer, M.; Chunhui, J.; Wulasihan, M. Therapeutic Efficacy of Liraglutide versus Metformin in Modulating the Gut Microbiota for Treating Type 2 Diabetes Mellitus Complicated with Nonalcoholic Fatty Liver Disease. Front. Microbiol. 2023, 14, 1088187. [Google Scholar] [CrossRef]
  100. Guasch-Ferré, M.; Hruby, A.; Toledo, E.; Clish, C.B.; Martínez-González, M.A.; Salas-Salvadó, J.; Hu, F.B. Metabolomics in Prediabetes and Diabetes: A Systematic Review and Meta-Analysis. Diabetes Care 2016, 39, 833–846. [Google Scholar] [CrossRef]
  101. Du, J.; Xi, L.; Zhang, Z.; Ge, X.; Li, W.; Peng, W.; Jiang, X.; Liu, W.; Zhao, N.; Wang, X.; et al. Metabolic Remodeling of Glycerophospholipids Acts as a Signature of Dulaglutide and Liraglutide Treatment in Recent-Onset Type 2 Diabetes Mellitus. Front. Endocrinol. 2022, 13, 1097612. [Google Scholar] [CrossRef]
  102. Ye, J.; Xu, J.; Wen, W.; Huang, B. Effect of Liraglutide on Serum TSH Levels in Patients with NAFLD and Its Underlying Mechanisms. Int. J. Clin. Pract. 2022, 2022, 1786559. [Google Scholar] [CrossRef]
  103. 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]
  104. Frías, J.P.; Davies, M.J.; Rosenstock, J.; Pérez Manghi, F.C.; Fernández Landó, L.; Bergman, B.K.; Liu, B.; Cui, X.; Brown, K. Tirzepatide versus Semaglutide Once Weekly in Patients with Type 2 Diabetes. N. Engl. J. Med. 2021, 385, 503–515. [Google Scholar] [CrossRef] [PubMed]
  105. Jastreboff, A.M.; Aronne, L.J.; Ahmad, N.N.; Wharton, S.; Connery, L.; Alves, B.; Kiyosue, A.; Zhang, S.; Liu, B.; Bunck, M.C.; et al. Tirzepatide Once Weekly for the Treatment of Obesity. N. Engl. J. Med. 2022, 387, 205–216. [Google Scholar] [CrossRef]
  106. Hartman, M.L.; Sanyal, A.J.; Loomba, R.; Wilson, J.M.; Nikooienejad, A.; Bray, R.; Karanikas, C.A.; Duffin, K.L.; Robins, D.A.; Haupt, A. Effects of Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide on Biomarkers of Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes. Diabetes Care 2020, 43, 1352–1355. [Google Scholar] [CrossRef]
  107. Clayton-Chubb, D.; Kemp, W.; Majeed, A.; Lubel, J.S.; Hodge, A.; Roberts, S.K. Understanding NAFLD: From Case Identification to Interventions, Outcomes, and Future Perspectives. Nutrients 2023, 15, 687. [Google Scholar] [CrossRef] [PubMed]
  108. Parker, V.E.R.; Robertson, D.; Wang, T.; Hornigold, D.C.; Petrone, M.; Cooper, A.T.; Posch, M.G.; Heise, T.; Plum-Moerschel, L.; Schlichthaar, H.; et al. Efficacy, Safety, and Mechanistic Insights of Cotadutide, a Dual Receptor Glucagon-Like Peptide-1 and Glucagon Agonist. J. Clin. Endocrinol. Metab. 2020, 105, 803–820. [Google Scholar] [CrossRef]
  109. Boland, M.L.; Laker, R.C.; Mather, K.; Nawrocki, A.; Oldham, S.; Boland, B.B.; Lewis, H.; Conway, J.; Naylor, J.; Guionaud, S.; et al. Resolution of NASH and Hepatic Fibrosis by the GLP-1R and GCGR Dual-Agonist Cotadutide via Modulating Mitochondrial Function and Lipogenesis. Nat. Metab. 2020, 2, 413–431. [Google Scholar] [CrossRef]
  110. 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]
  111. Vedtofte, L.; Bahne, E.; Foghsgaard, S.; Bagger, J.I.; Andreasen, C.; Strandberg, C.; Gørtz, P.M.; Holst, J.J.; Grønbæk, H.; Svare, J.A.; et al. One Year’s Treatment with the Glucagon-Like Peptide 1 Receptor Agonist Liraglutide Decreases Hepatic Fat Content in Women with Nonalcoholic Fatty Liver Disease and Prior Gestational Diabetes Mellitus in a Randomized, Placebo-Controlled Trial. J. Clin. Med. 2020, 9, 3213. [Google Scholar] [CrossRef]
  112. Eli Lilly and Company. A Randomized, Double-Blind, Placebo-Controlled Phase 2 Study Comparing the Efficacy and Safety of Tirzepatide versus Placebo in Patients with Nonalcoholic Steatohepatitis (NASH); ClinicalTrials.gov: Bethesda, MD, USA, 2023. [Google Scholar]
  113. Central Hospital, Nancy, France. A Multicentre Controlled and Randomized Study Assessing the Effect of Dulaglutide Add-On to Dietary Reinforcement versus Dietary Reinforcement Alone in Patients with Type 2 Diabetes and Carriers of a Non-Alcoholic Steatohepatitis; ClinicalTrials.gov: Bethesda, MD, USA, 2019. [Google Scholar]
  114. The Deutsche Diabetes Forschungsgesellschaft e.V. Combined Active Treatment in Type 2 Diabetes with NASH; ClinicalTrials.gov: Bethesda, MD, USA, 2022. [Google Scholar]
  115. AstraZeneca. A Phase IIb/III Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety and Efficacy of Cotadutide in Participants with Non-Cirrhotic Non-Alcoholic Steatohepatitis with Fibrosis; ClinicalTrials.gov: Bethesda, MD, USA, 2023. [Google Scholar]
  116. Agha, D.A. Effect on Non-Alcoholic Fatty Liver Disease with Advanced Fibrosis in Patients with Type 2 Diabetes Mellitus on Treatment with Gastric Inhibitory Polypeptide/Glucagon Like Peptide-1 Analogue (Tirzpatide); ClinicalTrials.gov: Bethesda, MD, USA, 2023. [Google Scholar]
  117. Caprio, S. Semaglutide, 2.4 mg, Once Weekly: Effects on Beta-Cell Preservation and Reduction of Intrahepatic Triglyceride Content in Obese Youth with Prediabetes (IGT)/Early Type 2 Diabetes (T2D) and Non-Alcoholic Fatty Liver Disease (NAFLD); ClinicalTrials.gov: Bethesda, MD, USA, 2022. [Google Scholar]
Figure 1. The beneficial effects of GLP-1RAs on various cardiometabolic components associated with T2DM.
Figure 1. The beneficial effects of GLP-1RAs on various cardiometabolic components associated with T2DM.
Metabolites 13 00581 g001
Table 1. Characteristics of GLP-1RAs approved for the treatment of T2DM [24,30].
Table 1. Characteristics of GLP-1RAs approved for the treatment of T2DM [24,30].
GLP-1RA
Generic Name, Ref.
Trade Name
Manufacturer
First ApprovedDose and Route of AdministrationAdministration ScheduleSide Effects
Short-acting
  • Nausea, vomiting, diarrhea.
  • Pruritus and erythema in the injection area of administration.
  • Risk of pancreatitis and pancreatic cancer.
  • Risk of gallbladder and biliary disease [31].
Exenatide [32]Byetta®
AstraZeneca, Cambridge, England
2005 (US)
2006 (EU)
5–10 mcg scTwice daily prior to meals
Lixisenatide [33]Adlyxin®, Lyxumia®
Sanofi, Paris, France
2013 (EU)
2016 (US)
10–20 mg scOnce daily prior to the first meal
Long-acting
Exenatide ER [32]Bydureon®
AstraZeneca, Cambridge, England
2012 (US)
2011 (EU)
2 mg scOnce weekly, unrelated to meals
Albiglutide [34]Eperzan®
Tanzeum®
GlaxoSmithKline, London, England
2014 (EU)
2014 (US)
30–50 mg scOnce weekly, unrelated to meals
Liraglutide [35]Victoza®
Novo Nordisk, Bagsvaerd, Denmark
2010 (US)
2009 (EU)
0.6–1.8 mg scOnce daily, unrelated to meals
Dulaglutide [36]Trulicity®
Eli Lilly and Company, Indianapolis, Indiana, USA
2014 (US, EU)0.75–1.5 mg scOnce weekly, unrelated to meals
Semaglutide [37,38]Ozempic®
Novo Nordisk, Bagsvaerd, Denmark
2017 (US)
2018 (EU)
0.25–1 mg scOnce weekly, unrelated to meals
Rybelsus®
Novo Nordisk, Bagsvaerd, Denmark
2019 (US)
2020 (EU)
3–14 mg poOnce daily, one hour prior to the first meal
Abbreviations: GLP-1RAs, glucagon-like peptide-1 receptor agonists; Ref., reference; US, United States; EU, Europe; sc, subcutaneously; po, oral.
Table 2. The place of GLP-1RAs among various guidelines as a clinical approach to NAFLD in T2DM patients.
Table 2. The place of GLP-1RAs among various guidelines as a clinical approach to NAFLD in T2DM patients.
Guidelines/GuidanceRecommendations
AASLD Practice Guidance on the Clinical Assessment and Management of Nonalcoholic Fatty
Liver Disease, 2023 [15]
Semaglutide can be considered for T2DM or obesity in patients with NASH, adding cardiovascular benefit and improving NASH.
AACE Clinical Practice Guideline for the Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Primary Care and Endocrinology Clinical Settings, 2022 [12]Recommend pioglitazone and GLP-1RAs for people with T2DM and biopsy-proved NASH.
Consider treating T2DM with GLP-1RAs or pioglitazone in the situation of possible NASH with modified noninvasive tests and elevated levels of hepatic enzymes, also offering cardiometabolic benefits, even in pediatric obesity and T2DM.
APASL clinical practice guidelines for the diagnosis and management of metabolic-associated fatty liver disease, 2020 [40]Insufficient evidence in the Asian population.
EASL–EASD–EASO Clinical Practice Guidelines for the management of nonalcoholic fatty liver disease, 2016 [17]Pharmacotherapy reserved for patients with NASH and significant fibrosis (≥stage F2) or NASH with a high risk for disease progression (elevated ALT, T2DM, MS).
No firm recommendation for the use of pioglitazone or vitamin E in NASH.
Insufficient evidence for GLP-1RAs.
NICE Guideline on Liver Disease (NAFLD), 2016 [41]GLP-1RAs not mentioned.
Insufficient evidence for pioglitazone and vitamin E.
Abbreviations: AASLD, American Association for the Study of Liver Diseases; AACE, American Association of Clinical Endocrinology; APASL, The Asian Pacific Association for the Study of the Liver; EASL, European Association for the Study of the Liver; EASD, European Association for the Study of Diabetes; EASO, European Association for the Study of Obesity; NICE, National Institute for Health and Care Excellence; NASH, nonalcoholic steatohepatitis, GLP-1RA, glucagon-like peptide-1 receptor agonist; T2DM, type 2 diabetes mellitus; MS, metabolic syndrome; ALT, alanine aminotransferase.
Table 3. Summary of studies that evaluated the effects of GLP-1RAs in patients with T2DM and NAFLD.
Table 3. Summary of studies that evaluated the effects of GLP-1RAs in patients with T2DM and NAFLD.
GLP-1RAAuthor, Year, Ref.CountryTotal Patients (GLP-1RA/Control)Control ArmGLP-1RA ArmFollow-Up
Duration
Diagnosis MethodResults of GLP-1RAs
ExenatideGastaldelli et al., 2020 [42]Italy228/227/230Dapagliflozin + placebo
Exenatide + placebo
Exenatide 2 mg OW and Dapagliflozin 10 mg daily104 weeksSteatosis and fibrosis scoresImproved biomarkers of steatosis and fibrosis in the exenatide and dapagliflozin group vs. exenatide or dapagliflozin alone.
Liu et al., 2020 [43]China35/36Glargine insulinExenatide 5 μg BID/4 weeks, then 10 μg BID/20 weeks24 weeks1H-MRSGreater reductions in body weight, visceral fat area, liver enzymes, FIB-4, postprandial plasma glucose, and LDL-C.
Dutour et al., 2016 [44]France22/22Standard of care without GLP-1RAExenatide 5 μg BID/4 weeks, then 10 μg BID/22 weeks26 weeks1H-MRSReduced liver fat content and epicardial fat dependent on weight loss.
Shao et al., 2014 [45]China30/30Insulin-based therapyExenatide 5 μg BID/4 weeks, then 10 μg BID/8 weeks12 weeksUSLower AST, ALT, and GGT (p < 0.001) correlated with body weight change.
Fan et al., 2013 [46]China49/68MetforminExenatide 5 μg BID/4 weeks, then 10 μg BID/8 weeks12 weeksUSControlled blood glucose, reduced body weight, and improved hepatic enzymes.
Cuthbertson et al., 2012 [47]United Kingdom25
19/6
-Exenatide 5 μg BID/4 weeks, then 10 μg BID/20 weeks
Or Liraglutide 0.6–1.2 mg daily
24 weeks1H-MRS
MRI
Correlation between intrahepatic lipid and AST, ALT, and GGT levels (p < 0.05).
Significant body weight relative reduction of 4.3%.
Reduction in VAT and SAT volume.
Sathyanarayana et al., 2011 [48]USA11/10PioglitazoneExenatide 5 μg BID/2 weeks, then 10 μg BID/50 weeks52 weeksMRS,
plasma adiponectin
Significantly greater reduction in hepatic fat (Δ = 61% vs. 41%, p < 0.05), and greater increase in circulating adiponectin in combination therapy vs. pioglitazone alone.
Buse, 2007 [49]USA283PlaceboExenatide 5 μg or 10 μg BID2 yearsLiver enzymesHbA1c and ALT reduction, insulin resistance improvement, weight loss, and blood pressure.
LiraglutideTan et al., 2022 [50]China262/1503Conventional drug therapyLiraglutide daily12 monthsFibrosis scores
LSM
Decrease of NFS (p = 0.043), FIB-4 (p = 0.044), and LSM (p = 0.007).
Reduced prevalence of advanced fibrosis (3.1% vs. 6.1%, p = 0.218) in the liraglutide group vs. control group.
Bizino et al., 2020 [51]Netherlands23/26PlaceboLiraglutide 1.8 mg daily26 weeksMRISignificantly reduced SAT
No significant change in VAT, hepatic, epicardial, and myocardial fat content.
Gameil et al., 2020 [52]Egypt79/80/65Conventional treatmentLiraglutide 1.8 mg daily
Or Dulaglutide 1.5 mg OW
24 weeksFLI
FIB-4 score
Significant reduction of median FLI and FIB-4 score in the liraglutide and dulaglutide groups vs. the conventional treatment group (p < 0.001).
Greater change in the liraglutide group vs. dulaglutide group (p = 0.027).
Guo et al., 2020 [53]China32/32/32Insulin glargine
Placebo
Liraglutide 1.8 mg daily26 weeks1H-MRSSignificantly decreased IHCL
(26.4% ± 3.2% to 20.6% ± 3.9%, p < 0.05).
Significantly decreased of SAT and VATgroup (SAT, 331.7 ± 79.0 cm2 to 295.3 ± 80.3 cm2, p < 0.05; and VAT, 235.6 ± 30.8 cm2 to 188.2 ± 26.6 cm2, p < 0.05).
Simeone et al., 2020 [54]Italy16 people with T2DM, 16 with prediabetesLifestyle counselingLiraglutide 1.8 mg dailyNot mentionedMRIImprovement in glycemic control, CRP, IL-1β level, BMI, and NAFLD degree.
Yan et al., 2019 [55]China24/27Sitagliptin
Glargine
Liraglutide 1.8 mg daily26 weeksMRI-PDFFReduced body weight, intrahepatic lipid, and VAT in addition to improving glycemic control.
Tian et al., 2018 [56]China52/75Metformin 1–1.5 g/dayLiraglutide 0.6–1.8 mg daily12 weeksB-mode Ultrasonic
Scanning
Significant decrease of 2 h plasma glucose, AST, ALT, and adiponectin levels.
Zhang et al., 2018 [57]China424/411Conventional drug therapyLiraglutide 1.2 mg daily24 weeksBiochemical analyzerSignificant improvement in blood glucose level, HbA1c, lipid profile, and liver function.
Bouchi et al., 2017 [58]Japan8/9InsulinLiraglutide 0.9 mg daily36 weeksCTSignificantly reduced VFA, LAI, ACR, and CRP levels.
Feng et al., 2017 [59]China29/29Metformin
Gliclazide
Liraglutide daily24 weeksUSLower HbA1c, improved liver enzymes, weight loss on liraglutide and metformin.
Decreased liver fat content in all groups, greater in liraglutide compared with others.
Petit et al., 2017 [60]France68Conventional drug therapyLiraglutide 1.2 mg daily24 weeksMRSSignificantly reduced LFC (−31%, p = 0.0001) and body weight.
Armstrong et al., 2016 [61]United Kingdom26/26PlaceboLiraglutide 1.8 mg daily48 weeks
(extension to 72 weeks)
Liver biopsySignificantly higher NASH resolution
(39% liraglutide vs. 9% placebo).
No worsening of fibrosis.
Smits et al., 2016 [62]Netherlands17/17Sitagliptin 100 mg
Placebo
Liraglutide 1.8 mg daily12 weeks1H-MRS
Fibrosis scores
Reduced glycemia, HbA1c, insulin levels.
Reduced steatosis by 10% (p = 0.98).
No influence on hepatic fibrosis.
Reduced plasma albumin levels (p = 0.03).
Vanderheiden et al., 2016 [63]USA35/36PlaceboLiraglutide 1.8 mg daily24 weeksMRI/MRSSignificant reduction in abdominal SAT, no change in VAT, and the ratio of visceral to total fat.
Tang et al., 2015 [64]Canada18/17GlargineLiraglutide 1.8 mg daily12 weeksMRI-PDFFSimilar improvement in the liver fat fraction in both groups (p = 0.34).
No weight gain from insulin therapy.
Ohki et al., 2012 [65]Japan26/36
26/20
Sitagliptin
Pioglitazone
Liraglutide daily
0.3 mg/first week, 0.6 mg/next week, and finally up to the limit dose of 0.9 mg if necessary
48 weeks for liraglutideUSBody weight decreased from 81.1 to 78 kg.
BMI from 30.1 to 28.6 kg/m2.
Fasting glycemia from 207 to 168 mg/dL.
HbA1c from 8.4% to 7.6%.
AST from 50 IU/L to 35 IU/L.
ALT from 65 IU/L to 48 IU/L.
APRI index from 0.73 to 0.49.
SemaglutideCarretero-Gomez et al., 2023 [66]Spain213-Semaglutide sc OW24 weeksHSI
FIB-4
Significant reduction in HSI (−2.36, p < 0.00001) and FIB-4 (−0.075, p < 0.016), related to the decrease of body weight, triglyceride levels, insulin resistance, and liver enzymes.
Arai et al., 2022 [67]Japan16-Oral semaglutide daily
3 mg/4 weeks, then 7 mg/4 weeks, then 14 mg/16 weeks
24 weeksCAP, LSMReduced HbA1c, HOMA-IR, ferritin normalized hepatic enzymes.
Decreased FIB-4 index from 1.42 to 1.1.
Significantly decreased CAP values from 344 to 279 dB/m.
Ding et al., 2022 [68]China75/75MetforminSemaglutide sc OW
0.6 mg/1.2 mg/1.8 mg
12 weeksUSDecreased AST, ALT, and GGT levels (p < 0.05).
Significantly reduced moderate to severe NAFLD patients.
Volpe et al., 2022 [69]Italy48-Semaglutide sc 0.25 mg OW for 4 weeks,
then 0.5 mg OW for
20 weeks
52 weeksUS
US-Liver Steatosis Score
Reduced body weight, HbA1c, HOMA-IR, serum lipid, AST, ALT, GGT, FLI.
Significant improvement in liver steatosis severity, body composition.
Flint et al., 2021 [70]Germany34/33PlaceboSemaglutide sc 0.4 mg daily72 weeksMRE
MRI-PDFF
≥30% reduction in liver fat content, but no significant change in liver stiffness.
Decrease of HbA1c, liver enzymes, body weight.
Newsome et al., 2021 [71]United Kingdom82/80PlaceboSemaglutide sc 0.4 mg daily72 weeksLiver biopsyResolution of NASH (59% vs. 17% placebo, p < 0.001).
Slowed fibrosis progression (4.9% vs. 18.8% placebo), but no significant reduction in fibrosis stages.
DulaglutideKuchay MS, 2020 [72]India32/32Standard of care without GLP-1RADulaglutide 0.75 mg OW for 4 weeks,
then 1.5 mg OW for
20 weeks
24 weeksMRI-PDFFSignificant reduction of LFC (−3.5%, p = 0.025) and GGT levels (−13.1 IU/L, p = 0.025) in patients with NAFLD.
No significant reductions in PFC, liver stiffness, serum AST and ALT levels.
Bogomolov et al., 2020 [73]Russia65-Dulaglutide 0.75 mg OW for 2 weeks, then 1.5 mg OW for 24 weeks26 weeksFLISignificant reduction of body weight, BMI, waist circumference, glucose, HbA1c, insulin resistance indexes, transaminases, and GGT.
Decreased FLI and liver stiffness.
Abbreviations: GLP-1RA, glucagon-like peptide-1 receptor agonist; OW, once weekly; BID, twice a day; 1H-MRS, proton magnetic resonance spectroscopy; FIB-4, fibrosis-4, LDL-C, low-density lipoprotein cholesterol; ALT, alanine aminotransferase; AST, aspartate aminotransferase; GGT, gamma-glutamyl transferase; MRI, magnetic resonance imaging; VAT, visceral adipose tissue; SAT, subcutaneous adipose tissue; MRS, magnetic resonance spectroscopy; US, ultrasonography; LFC, liver fat content; T2DM, type 2 diabetes mellitus; VFA, visceral fat area; LAI, liver attenuation index; ACR, albumin-to-creatinine ratio; CRP, C-reactive protein; IL-1β, Interleukin-1β; BMI, body mass index; APRI, AST to platelet ratio index; NAFLD, nonalcoholic fatty liver disease; NFS, NAFLD fibrosis score; HSI, hepatic steatosis index; LSM, liver stiffness measurement, CAP, controlled attenuation parameter; NASH, nonalcoholic steatohepatitis; MRI-PDFF, magnetic resonance imaging-proton density fat fraction; IHCL, intra-hepatocellular lipid content; CT, computed tomography; MRE, magnetic resonance elastography; HOMA-IR, homeostatic model assessment for insulin resistance; FLI, fatty liver index; PFC, pancreatic fat content.
Table 4. Ongoing trials investigating the effects of GLP-1RAs and next-generation-related molecules in patients with T2DM and NAFLD (www.clinicaltrial.gov, accessed 1 February 2023).
Table 4. Ongoing trials investigating the effects of GLP-1RAs and next-generation-related molecules in patients with T2DM and NAFLD (www.clinicaltrial.gov, accessed 1 February 2023).
Trial NameEstimated EnrollmentStart DateCompletion DateInterventionCriteria for DiabetesPrimary OutcomeSecondary Outcome
A Randomized, Double-Blind, Placebo-Controlled Phase 2 Study Comparing the Efficacy and Safety of Tirzepatide Versus Placebo in Patients with Nonalcoholic Steatohepatitis
(SYNERGY-NASH) [112]
196 participants19 November 201913 February 2024Tirzepatide 5/10/15 mg sc OW vs. placeboPatients with T2DM (HbA1c ≤ 9.5%) or without T2DM
  • Percentage of participants with absence of NASH with no worsening of fibrosis on liver histology.
  • ≥1 point decrease in fibrosis stage with no worsening of NASH on liver histology.
  • ≥1 point increase in fibrosis stage on liver histology.
  • ≥2 point decrease in NAS on liver histology, with ≥1 point reduction in at least 2 NAS components.
  • Change in liver fat content by MRI-PDFF.
  • Change in body weight.
Researching an Effect of GLP-1 Agonist on Liver Steatosis (REALIST) [113]93 participants1 September 201930 March 2024Dulaglutide 1.5 mg sc OWModerately controlled T2DM under OADs stable dose for at least 3 months
  • Histological improvement (regression of NASH) without worsening fibrosis over 52 weeks.
  • Changes in Fibrosis Kleiner score, Fibrotest score measurement, Fibrosis marker parameter, serum levels of ALT and AST, lipid profile, glycemic control, fat mass, quality of life, weight over 52 weeks, and changes in weight and AST and ALT levels over 24 weeks.
Combined Active Treatment in Type 2 Diabetes with NASH (COMBAT_T2_NASH) [114]192 participants26 March 2021December 2023Empagliflozin
10 mg/d + semaglutide
1 mg/week
vs. empagliflozin
10 mg/d vs. placebo
Patient with T2D (HbA1c ≤ 9.5%) and NASH (F1–F3 fibrosis stage)
  • Histological resolution of NASH without worsening of fibrosis over 48 weeks.
  • Overall NAS, fibrosis stage, activity component of NASH, grade of steatosis over 48 weeks.
The Effect of Semaglutide in Subjects with Noncirrhotic Nonalcoholic Steatohepatitis
(ESSENCE) [87]
1200 participants1 April 202126 May 2028Semaglutide sc OWPatients with T2DM, without any glucose-lowering agents
  • Resolution of NASH and no worsening of liver fibrosis.
  • Improvement in liver fibrosis with no worsening of NASH.
  • Time to first liver-related clinical event.
  • Progression of liver fibrosis, NASH.
  • Change in body weight, AST, ALT, lipid profile, histology-assessed liver collagen proportionate area, liver stiffness assessment.
A Phase IIb/III Randomized, Double-blind, Placebo-controlled Study to Evaluate the Safety and Efficacy of Cotadutide in Participants with Noncirrhotic Nonalcoholic Steatohepatitis with Fibrosis
(PROXYMO-ADV) [115]
1860 participants14 July 202226 March 2026Cotadutide 300 μg/600 μg sc once daily vs. placeboPatients with or without T2DM
  • Resolution of NASH without worsening of liver fibrosis based on biopsy.
  • Liver fibrosis improvement by at least one stage without worsening of NASH.
  • Change in body weight, HbA1c, triglycerides.
  • Progression to cirrhosis based on biopsy.
Effect on Nonalcoholic Fatty Liver Disease with Advanced Fibrosis in Patients with Type 2 Diabetes Mellitus on Treatment with Gastric Inhibitory Polypeptide/Glucagon-Like Peptide-1 Analogue
NCT05751720 [116]
30 participantsApril 2023February 2025Tirzepatide sc 0.25 mg OW/4 weeks then 0.5 mg OW, or
Oral semaglutide 3 mg daily/first month, then 7 mg daily for 6 months
Patients with T2DM for > 1 year in
presence of NAFLD advanced fibrosis
  • Change in liver stiffness in terms of kPa.
  • Change in liver fat quantification.
  • Change in BMI.
  • Glycemic control.
Semaglutide, 2.4 mg, Once Weekly: Effects on Beta-cell Preservation and Reduction of Intrahepatic Triglyceride Content in Obese Youth with Prediabetes (IGT)/Early Type 2 Diabetes (T2D) and Nonalcoholic Fatty Liver Disease (NAFLD)
NCT05067621 [117]
60 participantsJanuary 2023January 2027Semaglutide sc 0.24 mg/0.5 mg/1.0 mg/1.7 mg/2.4 mg OWImpaired glucose tolerance or new onset T2DM (<6 months duration)
  • Change in oDI.
  • Change in PDFF.
  • Change in oral glucose tolerance test (OGTT) derived biomarkers: oDI, fasting insulin, C-peptide.
  • Time to glucose peak.
  • Fractional rates of de novo lipogenesis.
  • Changes in lipidic profile and liver fibrosis.
Abbreviations: T2DM, type 2 diabetes mellitus; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; GLP-1, glucagon-like peptide-1; NAS, NAFLD Activity Score; MRI-PDFF, magnetic resonance imaging—proton density fat fraction; oDI, Oral Disposition Index; OAD, oral antidiabetics; sc, subcutaneously; ALT, alanine aminotransferase; AST, aspartate aminotransferase; kPa, kilopascals; BMI, body mass index; IGT, impaired glucose tolerance.
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

Cazac, G.-D.; Lăcătușu, C.-M.; Ștefănescu, G.; Mihai, C.; Grigorescu, E.-D.; Onofriescu, A.; Mihai, B.-M. Glucagon-like Peptide-1 Receptor Agonists in Patients with Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease—Current Background, Hopes, and Perspectives. Metabolites 2023, 13, 581. https://doi.org/10.3390/metabo13050581

AMA Style

Cazac G-D, Lăcătușu C-M, Ștefănescu G, Mihai C, Grigorescu E-D, Onofriescu A, Mihai B-M. Glucagon-like Peptide-1 Receptor Agonists in Patients with Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease—Current Background, Hopes, and Perspectives. Metabolites. 2023; 13(5):581. https://doi.org/10.3390/metabo13050581

Chicago/Turabian Style

Cazac, Georgiana-Diana, Cristina-Mihaela Lăcătușu, Gabriela Ștefănescu, Cătălina Mihai, Elena-Daniela Grigorescu, Alina Onofriescu, and Bogdan-Mircea Mihai. 2023. "Glucagon-like Peptide-1 Receptor Agonists in Patients with Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease—Current Background, Hopes, and Perspectives" Metabolites 13, no. 5: 581. https://doi.org/10.3390/metabo13050581

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

Article Metrics

Back to TopTop