Minireviews Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Methodol. Jun 20, 2024; 14(2): 91319
Published online Jun 20, 2024. doi: 10.5662/wjm.v14.i2.91319
Non-alcoholic fatty liver disease in type 2 diabetes: Emerging evidence of benefit of peroxisome proliferator-activated receptors agonists and incretin-based therapies
Subhodip Pramanik, Department of Endocrinology, Neotia Getwel Multispecialty Hospital, Siliguri 734010, West Bengal, India
Partha Pal, Department of Medical Gastroenterology, Asian Institute of Gastroenterology, Hyderabad 500082, India
Sayantan Ray, Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar 751019, Odisha, India
ORCID number: Subhodip Pramanik (0000-0002-3196-1192); Partha Pal (0000-0002-7090-9004); Sayantan Ray (0000-0002-6274-465X).
Author contributions: Pramanik S performed the literature search, wrote the first draft, and provided intellectual input; Pal P conceptualized the work, performed a literature search, and supervised the writing; Ray S supervised the literature search, and the writing, provided intellectual input and critically revised the manuscript.
Conflict-of-interest statement: All authors have no conflicts of interest to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Sayantan Ray, MBBS, MD, Assistant Professor, Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar, Sijua, Patrapada, Bhubaneswar 751019, Odisha, India. sayantan.ray30@gmail.com
Received: December 27, 2023
Peer-review started: December 27, 2023
First decision: January 5, 2024
Revised: January 20, 2024
Accepted: February 27, 2024
Article in press: February 27, 2024
Published online: June 20, 2024

Abstract

Nonalcoholic fatty liver disease (NAFLD) is a global epidemic, affecting more than half of the people living with type 2 diabetes (T2D). The relationship between NAFLD and T2D is bidirectional and the presence of one perpetuates the other, which significantly increases the hepatic as well as extrahepatic complications. Until recently, there was no approved pharmacological treatment for NAFLD/ nonalcoholic steatohepatitits (NASH). However, there is evidence that drugs used for diabetes may have beneficial effects on NAFLD. Insulin sensitizers acting through peroxisome proliferator-activated receptor (PPAR) modulation act on multiple levels of NAFLD pathogenesis. Pioglitazone (PPARγ agonist) and saroglitazar (PPARα/γ agonist) are particularly beneficial and recommended by several authoritative bodies for treating NAFLD in T2D, although data on biopsy-proven NASH are lacking with the latter. Initial data on elafibanor (PPAR α/δ agonist) and Lanifibranor (pan PPAR agonist) are promising. On the other hand, incretin therapies based on glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RA) and dual- and triple-hormone receptor co-agonists reported impressive weight loss and may have anti-inflammatory and antifibrotic properties. GLP-1 RAs have shown beneficial effects on NAFLD/NASH and more studies on potential direct effects on liver function by dual- and triple-agonists are required. Furthermore, the long-term safety of these therapies in NAFLD needs to be established. Collaborative efforts among healthcare providers such as primary care doctors, hepatologists, and endocrinologists are warranted for selecting patients for the best possible management of NAFLD in T2D.

Key Words: Non-alcoholic fatty liver disease, Type 2 diabetes, Evidence, PPAR agonists, Incretin-based therapies

Core Tip: Co-existence of non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) synergistically act to increase the risk of adverse hepatic and extra hepatic outcomes. T2DM is an established risk factor for NAFLD progression to NASH, advanced fibrosis or cirrhosis. Timely intervention in these populations could have a significant effect on liver- related outcomes. Newer dual and pan-PPAR agonists show promising results in patients with NAFLD/NASH and T2DM. Incretin-based therapy for the treatment of NAFLD is currently being explored. With better understanding of the complex interaction between T2DM and NAFLD, PPAR agonists and incretin-based therapies are likely to provide more effective approach of NAFLD management in T2DM.



INTRODUCTION

The coexistence of non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D) synergistically act to increase the risk of adverse hepatic and extrahepatic outcomes. The global prevalence of NAFLD in people with T2D has been estimated at nearly 56% in a large meta-analysis of observational studies. More than one-third of people with T2D have nonalcoholic steatohepatitis (NASH), and around 1 in 6 carries advanced fibrosis[1]. T2D is an established risk factor for NAFLD progression to NASH, advanced fibrosis, or cirrhosis. Considering the prospective clinical implications of T2D in NAFLD, timely intervention in these patients could have a significant effect on liver-related outcomes. NAFLD and T2D share underlying altered pathophysiological mechanisms, including insulin resistance (IR). Expectedly, some anti-diabetic drug classes might exert favorable effects on NAFLD, with prevention of adverse cardiovascular (CV) events as well[2]. However, an unmet need for more focused research is there on how glycemic control through insulin sensitizers or incretin-based therapies might halt or revert the progression of NAFLD in individuals with T2D. The present mini-review summarizes the evidence on strong and significant association between T2D and the risk of adverse clinical outcomes in NAFLD, the therapeutic potential of peroxisome proliferator-activated receptors (PPAR) agonists for NASH and NASH-related hepatic morbidity, and lastly the key scientific advances regarding the use of incretins for treating NAFLD.

NAFLD AND T2D- THE COMPLEX LINK
From diabetes to NAFLD/NASH

The association between T2D and NAFLD is more complex than earlier thought and the relationship appears to be bidirectional[3]. The underlying pathophysiological mechanisms for the development of NAFLD are primarily the glucose and lipid metabolism alterations, with IR, elucidating the close link between T2D and NAFLD. Increased adiposity, often found in NAFLD and T2D, is coupled with adipocyte IR and dysfunction. Dysfunctional adipose tissue (AT) exhibits resistance to the antilipolytic effect of insulin. The consequent enhanced lipolysis and release of free fatty acids (FFAs) and glycerol are responsible for the accumulation of triglyceride and lipotoxicity in the liver, and other organs. Overflow of FAs to the liver also results in increased cellular levels of toxic lipids such as diacylglycerols, ceramides, and long-chain fatty acyl-coenzyme A (CoA), which are involved in inflammatory pathways (Figure 1)[4]. Excess FFAs also induce mitochondrial dysfunction, an increase in oxidative stress, and uncoupled oxidative phosphorylation. A fibrogenic response in hepatic stellate cells may be activated that can stimulate the progression to NASH and cirrhosis, and reactive oxygen species production (Figure 1)[5]. Dysfunction of AT and IR leads to the enhanced release of pro-inflammatory cytokines and decreased release of anti-inflammatory adipokines by AT. These cytokines can damage the liver directly, or indirectly, by raising oxidative stress, liver fibrosis, and the development of tumors[6,7]. Being closely interrelated, both glucotoxicity and lipotoxicity contribute to worsening IR and impaired insulin secretion (Figure 1)[8]. Hyperglycemia induces hepatic de-novo lipogenesis (DNL). Saturated fatty acid palmitate, the primary product of lipogenesis can induce hepatic inflammation and endoplasmic reticulum stress. Persistently elevated blood glucose levels promote DNL through direct and indirect mechanisms: the direct mechanism is by enhancing the activity of TCA cycle and Acyl CoA synthesis that acts as a substrate of both DNL and gluconeogenesis, and the indirect mechanism is through the transcription factor carbohydrate response element binding protein and liver X receptor αwhich sequentially promote unfavorable gene transcriptions.

Figure 1
Figure 1 Relationship between lipo- and glucotoxicity, insulin resistance, and beta-cell function. DAG: Diacylglycerol; DNL: De novo lipogenesis; NAFLD: Non-alcoholic fatty liver disease; ROS: Reactive oxygen species; T2D: Type 2 diabetes.
Progression of NAFLD in T2D

T2D is one of the strongest risk factors for the progression of NAFLD to NASH, cirrhosis, or hepatocellular carcinoma (HCC). An unhealthy lifestyle, via the promotion of gut dysbiosis and T2D-associated hyperglycemia and hyperinsulinemia, influences the transition from isolated steatosis to NASH and the progression of NASH. T2D-associated hyperglycemia also promotes the progression from NASH to hepatic fibrosis and HCC (Figure 2)[8]. Swedish National Diabetes Registry showed that severe liver disease risk is higher in T2D patients than in the general population. In people with T2D, risk factors associated with severe liver disease were older age, male gender, higher body mass index, smoking, hypertension, and microalbuminuria[9]. Among persons with NAFLD, those with T2D are at greater risk of developing advanced stages of fibrosis and subsequently hepatic decompensation, HCC, and liver-related mortality than those without T2D[10,11]. Huang et al[12] report the findings of a recent large meta-analysis comprising 2016 ethnically diverse participants. Compared with individuals without diabetes, participants with T2D had an elevated risk of hepatic decompensation and HCC despite adjustment for multiple factors having a known association with the progression of NAFLD. In general, liver fibrosis progresses by one stage over 7 years for people with NASH, but data are limited regarding the time to fibrosis progression in people with T2D compared with people without T2D[13]. In a recent comprehensive, multicentric study of patients with paired liver biopsies from the Nonalcoholic Steatohepatitis Clinical Research Network, the authors found that the cumulative incidence of fibrosis progression was considerably higher in participants with T2D compared with individuals without T2D. T2D remained an important predictor of the progression of fibrosis, even after controlling for several potential confounders[14]. Altogether, these findings provide convincing clinical data to point out that T2D is associated with advanced-stage liver disease, faster disease progression, and worse hepatic outcomes in people with NAFLD. Consequently, it is compulsory to screen for the presence and severity of NAFLD in all adults with T2D with liver enzymes in a wider range than usually considered[15]. Poor glycemic control has been linked to severe long-term complications, including advanced liver disease, and adverse outcomes in NAFLD; however, an ideal HbA1c cutoff value to detect people at risk of those outcomes is yet to be identified.

Figure 2
Figure 2 Modifiable and non-modifiable risk factors of non-alcoholic fatty liver disease and its progression to severe liver disease. Dashed arrows indicate factors that promote or predispose to disease. Among non-modifiable risk factors, most important genetic variants of nonalcoholic fatty liver disease are being extensively studied. Type 2 diabetes-associated hyperglycemia induces progression from nonalcoholic steatohepatitis to hepatic fibrosis and HCC. CV: Cardiovascular; HCC: Hepatocellular carcinoma; NAFLD: Nonalcoholic fatty liver disease; NASH: Nonalcoholic steatohepatitis.
THERAPEUTIC ROLE OF PPAR AGONISTS
Rationale

PPARs are nuclear receptors for specific endogenous ligands and act as transcription factors involved in the regulation of lipid metabolism, glucose homeostasis, and inflammation[16]. Endogenous ligands include free fatty acids (FAs), eicosanoids, and various complex lipids. PPAR forms a heterodimer with retinoid X receptor once ligands bind to it, and the complex binds to the response element that regulates the gene expression which is important for fatty acid metabolism[17]. There are three PPAR isoforms, PPAR α is largely present in the liver involved in fatty acid oxidation, and exerts anti-inflammatory action through complex regulation of nuclear factor kappa-light-chain-enhancer of activated B cells[18]. PPAR β/δ is expressed mainly in skeletal muscle but is also present in adipose tissue and skin and has a significant role in controlling lipid metabolism[19]. PPAR γ is highly expressed in adipose tissue and plays an important role in the regulation of adipogenesis, adipocyte differentiation, and fatty acid metabolism[20]. Together, PPAR agonists have a role in insulin sensitivity, inflammation, and dyslipidemia and thus act on multiple levels in NAFLD pathogenesis. This makes it an attractive option for drug development.

PPAR single agonists

Given the differential actions of different PPAR agonists, there has been a significant effort in the development of agonists for single receptors. They have been tried in different conditions including NAFLD, the results of which are encouraging. The agents, and trials with biochemical and histological outcomes of single agonists are summarized in Table 1[21-27].

Table 1 Summary of studies of peroxisome proliferator-activated receptors single agonists in nonalcoholic fatty liver disease.
Drug class and mechanism
Agent
Trial design
Biochemical response
Histological response
Comments
PPARα (Fatty acid oxidation and Anti-inflammatory)[21,22]Fenofibrate16 patients, 48 wk vs placeboSignificant reduction in triglyceride and liver enzymesDecreased ballooning, grade steatosis, inflammation/fibrosis no changeLimited efficacy
Clofibrate40 patients 1 yr vs UDCAReduced ALTNo change
PPAR β/δ (glucose homeostasis and insulin sensitivity)[23,24]GW5015166 patients, 2 wk vs placeboReduced TG and LDLNo dataAbandoned due to cancer risk in preclinical studies
MBX-8025181 overweight patients, 2 wk vs placeboFavorable lipid profile and decreased liver enzymesNo dataNeed more data
PPARγ (adipogenesis, insulin sensitization, fatty acid oxidation)[25-27]Rosiglitazone63 patients, 52 wk vs placeboNormalized transaminase levels (38% vs 7%, P = 0.005)Improved steatosis (47% vs 16%; P = 0.014), although only half of the patients responded, no change of other histologic parametersWeight gain and painful swollen legs in rosiglitazone arm
PioglitazoneRCT, 61 patients, 12 months placebo or pioglitazone had paired biopsies Improvement of ALT and GGTHepatocellular injury (P = 0.005), Mallory–Denk bodies (P = 0.004), and fibrosis (P = 0.05) were reduced in patients treated with pioglitazoneWeight gain with pioglitazone
RCT, 259 patients pioglitazone vs vitamin E vs placebo 96 wkImprovement of transaminases in the vitamin E and pioglitazone armImprovement in NASH as compared with placebo (pioglitazone P = 0.04), significant reductions in steatosis, lobular inflammation and fibrosis in pioglitazone armWeight gain in pioglitazone group

PPARα agonists: Fibrates are considered as PPARα agonists, albeit weak, and have limited clinical efficacy in NAFLD. In a study by Fernández-Miranda et al[21] 16 patients with NAFLD were treated with fenofibrate 200 mg/d for 48 wk and found a significant reduction in triglyceride and other liver enzymes. However, there was no significant improvement in histologic parameters. Another fibrate, clofibrate was compared with ursodeoxycholic acid in 40 patients for 1 year but no advantage was found in the clofibrate group. Newer selective PPARα-specific agonists, known as selective PPAR modulators, are in different phases of development[28,29].

PPAR β/δ agonists: Data is sparse with these PPAR isotypes. Two agents GW501516 and MBX-8025 have been tried in short trials[24,25]. They have shown some benefit in metabolic parameters (Table 1) but no histologic data is available. Further studies are required to establish any role of these agents in NAFLD.

PPARγ agonists: Thiazolidinediones are strong PPARγ agonists widely investigated in NAFLD. They are insulin-sensitizing agents and have a significant role in lipid metabolism. Rosiglitazone was compared with placebo for 1-year therapy in FLIRT trial[25] and found to have improved steatosis (47% vs 16%) and transaminitis (38% vs 7%), but lacked improvement in fibrosis and NAFLD activity score. Weight gain and painful swollen legs were significant side effects in the rosiglitazone arm. In the PIVENS trial[27], Pioglitazone was compared with vitamin E and placebo for 96 wk in patients with NASH where pioglitazone demonstrated a reduction in hepatic steatosis, lobular inflammation, and hepatic enzymes. A meta-analysis including 3 high-quality randomized controlled trials (RCTs) comparing pioglitazone vs placebo in NAFLD showed significant improvement in liver fibrosis[30]. Currently, pioglitazone is recommended in persons with T2D and biopsy-proven NASH by the American Association of Clinical Endocrinologists (AACE)[31] and recommended in both diabetic and non-diabetic adult patients with or without fibrosis by the Indian National Association for the study of the liver (INASL)[32] (Strong recommendation, high strength of evidence). However, the use of PPAR agonists can be associated with poor bone quality (in post-menopausal females) and dose-dependent pedal edema and weight gain.

Dual and pan PPAR agonists

Combination therapy using dual or pan agonists is an attractive choice as the most beneficial effects on NAFLD can be seen with the least side effects. For example, they can produce an antihyperlipidemic (PPARα) effect with insulin sensitization (PPARγ) and increase β-oxidation in the liver and skeletal muscle (α and β/δ). The efficacy can also be improved by synergistic action of different isoforms on fatty acid metabolism and insulin sensitivity. On the other hand, the side effects seen with the individual class of drugs like heart failure or weight gain with pioglitazone can be minimized. The agents and trials with biochemical and histological outcomes of dual/pan agonists are summarized in Table 2[33-40].

Table 2 Summary of studies of peroxisome proliferator-activated receptors dual and pan agonists in nonalcoholic fatty liver disease.
Drug class and mechanism
Agent
Trial design
Biochemical response
Histological response
Comments
PPAR α/δ agonist[33]Elafibranor276 patients, Elafibranor 80 mg vs 120 mg vs placebo, 52 wk, GOLDEN trialLiver enzymes, lipids, glucose profiles, and markers of systemic inflammation were significantly reduced in the elafibranor 120-mg groupElafibranor 120 mg was superior to the placebo, with NASH resolution without worsening of fibrosis in 19% versus 12% in the placebo group (P = 0.045), based on a post hoc analysis for the modified definitionNo change in primary outcome in intention to treat analysis
1070 patients, Elafibranor 120 mg vs placebo, 72 wk, RESOLVE IT trialImprovement in TG, ALT, and GGT 138 (19.2%) patients in the Ela group and 52 (14.7%) patients in the placebo group achieved resolution of NASH without worsening of fibrosis (P = 0.066)Despite the absence of safety signals, the RESOLVE-IT trial was discontinued due to the limited effect of Ela on surrogate efficacy endpoints
PPAR α/γ agonist[34-38]Tesaglitazar, Muraglitazar, AleglitazarFavorable lipid profile with muraglitazarNo changeTrials terminated early due to nephrotoxicity, cardiotoxicity and gastrointestinal hemorrhage respectively
Saroglitazar106 patients, 16 wk vs placeboImprovement in ALT and lipid profileNo dataDCGI approved for NAFLD in India
85 patients, 12 wk vs placeboImprovement in ALT and TGSignificant reduction in liver fibrosis (fibroscan)
Pan PPAR agonists[39,40]BezafibrateImprove HbA1c and atherogenic dyslipidemia in miceNo dataMostly animal data. Human studies are ongoing
LanifibranorImprove insulin sensitivity in miceImprove steatosis and fibrosis in liver tissue in mice

PPAR α/δ agonist: Elafibranor, an agonist of PPARα and PPARδ, works on insulin sensitivity, glucose homeostasis, and lipid metabolism. It has been studied in an RCT by Ratziu et al[33] (Golden trial) where two doses (80 mg and 120 mg) of Elafibranor were compared with a placebo. In the intention-to-treat analysis, there was no improvement in the resolution of NASH without worsening fibrosis (primary outcome). However, Elafibranor 120 mg became superior to placebo in 19% vs 12% in the placebo group (P = 0.045), based on a post hoc analysis for the modified definition of NASH resolution after excluding patients with mild steatohepatitis from the analysis. Another study RESOLVE IT (NCT02704403) was terminated early due to the limited effect of Elafibranor on surrogate efficacy endpoints. No safety signal was found in the studies.

PPAR α/γ agonists: Glitazars are PPAR α/γ agonists and improve dyslipidemia through action and insulin sensitivity through g action thus addressing two important issues of NAFLD. Initial glitazars, Tesaglitazar, Muraglitazar, and Aleglitazar, although showed significant improvement in dyslipidemia, trials for NFALD were terminated early due to nephrotoxicity, cardiotoxicity, and gastrointestinal hemorrhage respectively[34-36]. Saroglitazar is the new molecule in this class containing a unique pyrol moiety and lacks the glitazone ring and unlike its congeners has predominant PPARα action and moderate PPARγ action. Thus, it lacks the typical glitazone side effects. In mice models, saroglitazar has been shown to reduce histological NASH as compared with pioglitazone and fenofibrate[41]. Serum transaminase levels improved in patients with T2D treated with saroglitazar as compared with placebo[38,42]. A similar trial is ongoing at the United Nations with saroglitazar (NCT03061721) which will evaluate the improvement of serum transaminases and hepatic steatosis on MR imaging-estimated proton density fat fraction (MR-PDFF). Additionally, a recent phase III study (CTRI/2015/10/006236) in India is currently evaluating the histological efficacy of saroglitazar in comparison with a placebo in patients with biopsy-proven NASH.

Pan PPAR agonist: Pan PPAR agonists are still in various phases of development and many animal data are published. Bezafibrate and Lanifibranor have shown anti-lipid, anti-inflammatory, and antifibrotic properties in rats. An ongoing study is evaluating the efficacy of lanifibranor in patients with diabetes and NAFLD (NCT03459079). These agents may have a promising role in the management of NAFLD.

To conclude, given the multiple actions of the PPAR on lipid metabolism, oxidation of FAs, glucose homeostasis, and inflammation, PPAR agonists are attractive targets for the treatment of patients with NAFLD. Of all the PPAR agonists, the PPARα agonist pioglitazone is the most extensively evaluated and barring a few side effects, is most useful in patients with NAFLD and recommended by AACE and INASL guidelines. Emerging data of dual PPAR agonists and pan-PPAR agonists appear encouraging and may hold promise for patients with NAFLD.

THERAPEUTIC ROLE OF INCRETIN-BASED THERAPIES

In response to the intake of food, incretins play an important role in the regulation of blood glucose levels by enhancing insulin secretion from β-cells of the pancreas after meals. There are two main incretin hormones which are glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). GLP-1 is produced by L cells in the ileum and colon whereas GIP is produced from K cells in the duodenum and jejunum. The release of both is triggered as a response to the presence of nutrients (carbohydrates for both GLP1 and GIP, fat for GIP) in the digestive system. GLP-1 and GIP enhance glucose-dependent insulin release and inhibit glucagon secretion (glucagon stimulates the liver to release glucose) into the bloodstream. In addition, GLP-1 delays gastric emptying, regulates nutrient absorption, and promotes satiety whereas GIP promotes fat storage in adipose tissue[43].

Incretin-based therapies were originally developed for T2D which include GLP-1 receptor agonists (GLP-1 RAs) and dipeptidyl peptidase-4 (DPP-4) inhibitors. GLP-1 RAs (e.g., exenatide, liraglutide, semaglutide) bind to the GLP-1R and increase insulin secretion, inhibit glucagon release, slow gastric emptying, and promote satiety. DPP-4 inhibitors (sitagliptin, saxagliptin, linagliptin) increase the endogenous concentration of GLP-1 by inhibiting the DDP-4 enzyme which degrades GLP-1[44].

NASH is a multi-system disease with underlying metabolic stress driving its manifestations in several organs. The classical progression of NAFLD to NASH to cirrhosis and hepatocellular carcinoma is accelerated in T2D (Figure 3)[45]. Hence agents directed towards underlying metabolic dysfunction like incretin-based therapies could be helpful. These agents have pleiotropic effects apart from regulation of glucose metabolism such as anti-inflammatory and anti-fibrotic effects[46]. Several studies have assessed the clinical efficacy of incretin-based therapies in NASH.

Figure 3
Figure 3 Natural history of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. HCC: Hepatocellular carcinoma; NASH: Nonalcoholic steatohepatitis; NAFLD: Nonalcoholic fatty liver disease.
Mechanisms of action of incretin-based therapies in NASH

Insulin sensitivity improvement: GLP-1RAs act through augmenting incretins actions, namely GLP-1. GLP-1 is produced from the gut and has an impact on the pancreas to produce insulin and thus improves IR[47,48].

Effect on central nervous system: GLP-1 also has effects on central nervous system which reduce appetite and can lead to weight loss. GLP-1-based therapies are the mainstay of drug-induced weight loss. Within 6-12 months, semaglutide (a GLP-1 RA) induces 10%-15% weight loss comparable to bariatric procedures[49].

Anti-inflammatory effects and reduction of liver fat content: GLP-1 receptors are present in pancreas, gastrointestinal tract, CV system, kidney and many others organs. It promotes glucose uptake, and storage of glycogen in the liver and prevents gluconeogenesis[43]. Stimulation of FXR and LXR in the liver using GLP-1 RA may reduce liver fat as shown in animal models/human studies and inflammation.

Anti-Fibrotic Properties: GLP-1RAs like semaglutide have been shown to have dose dose-dependent trend for fibrosis reduction in NASH. With higher doses of GLP-1 RAs, significantly lower fibrosis worsening has been noted highlighting its potential for slowing NASH progression[50].

Improvement in the extra-hepatic milieu in NASH: GLP-1 has many biological effects other than those mentioned above. It has a cardio-protective effect and promotes diuresis/natriuresis in kidneys improving blood pressure (Figure 4). NASH is a multi-system disease with metabolic stress and systemic inflammation[46]. The prerequisites of NASH therapy are that it should improve liver disease and at least be neutral or beneficial with other comorbidities. Incretin-based therapies fulfill these criteria.

Figure 4
Figure 4 Pleotropic effects of glucagon-like peptide 1 and its effect of several organ systems in systematic inflammation characteristic of nonalcoholic steatohepatitis. GLP-1: Glucagon-like peptide 1.
Clinical evidence

Several clinical trials have explored the efficacy of incretin-based therapies in NASH (Table 3)[50-64]. The earlier studies were with liraglutide. The landmark LEAN study in 2016 showed that subcutaneous liraglutide 1.8 mg/d led to the resolution of biopsy-proven NASH in 39% of cases compared to 9% in placebo at 48 wk. Moreover, progression of fibrosis was seen in only 9% compared to 36% in placebo. GLP-1 RA also improved body weight, liver enzymes, and liver fat content[55].

Table 3 Summary of trials exploring the efficacy of incretin-based therapies in nonalcoholic steatohepatitis.
Ref.
Agent
Comparator
Participants
Duration
Outcomes
Newsome et al[50], 2021Semaglutide 0.1, 0.2 or 0.4 mg s/c once daily PlaceboNASH with F1, F2, or F3 fibrosis stage72 wkNASH resolution without worsening of fibrosis: 40% (0.1 mg), 36% (0.2 mg), 59% (0.4 mg), 33% (placebo); Improvement in fibrosis (P < 0.001 for 0.4 mg vs placebo); 43% (0.4 mg), 33% (placebo) (P = 0.48); Weight loss: 13% (0.4 mg), 1% placebo
Loomba et al[51], 2023Semaglutide 2.4 mg s/c Placebo (1: 2)NASH with compensated cirrhosis 48 wkImprovement in liver fibrosis by ≥ 1 stage without worsening NASH: 11% Semaglutide, 29% placebo (P = 0.087); Resolution of NASH at 48 wk: 34% semaglutide, 21% placebo (P = 0.29); No new safety concerns
Romero-Gomez et al[52], 2023 Efinopegdutide 10 mg s/c once weekly Semaglutide 1 mg s/c once weekly NAFLD with liver fat content (LFC) ≥ 10% 24 wkRelative reduction in LFC from baseline higher with efinopegdutide (72.7%) than semaglutide (42.3%); Similar reduction in body weight
Alkhouri et al[53], 2022Semaglutide 2.4 mg s/c + cilofexor once daily (30 or 100 mg)+ fircosostat (20 mg)Semaglutide alone
Semaglutide+cilofexor/fircosostat
NASH (MRI-PDFF > 10%, elastography ≥ 7 kPa24 wkCombination well tolerated; Greater reduction in liver fat with combination groups vs semaglutide (10%-11% vs 8.6%); Similar weight loss in all the groups
Flint et al[54], 2021 Semaglutide 0.4 mg s/c once daily PlaceboNAFLD, steatosis ≥ 10% (MRI-PDFF), MRE: 2.5-4.63 kPa48 wk≥ 30% reduction in liver steatosis at 24, 48 and 72 wk (64.7%, 76.5%, 73.5%) significantly higher than placebo; Change in liver stiffness in NAFLD not significantly different compared to placebo; Improvement in liver enzymes, body weight and HbA1c
Armstrong et al[55], 2016Liraglutide 1.8 mg/d s/cPlaceboBiopsy proven NASH48 wk39% had resolution of NASH (vs 9% placebo); Progression of fibrosis: 9% Liraglutide, 36% placebo
Khoo et al[56], 2017 Liraglutide 3.0 mg/dDiet and moderate exercise Obesity and NAFLD (Liver fat content > 5% on MRI) 26 wkSimilar reduction in weight, liver fat, liver enzymes in both the groups without any significant difference between the two
Yan et al[57], 2019 Liraglutide 1.8 mg/dInsulin glargine or sitagliptinT2D and NAFLD26 wkLiraglutide and sitagliptin along with metformin reduced body weight, liver fat content, visceral adipose tissue in addition to glycemic control in contrast to Insulin, subcutaneous fat also decreased in liraglutide arm
Bizino et al[58], 2020 Liraglutide 1.8 mg/dPlaceboT2D and NAFLD26 wkLiraglutide reduced significantly more body weight and subcutaneous fat but not visceral fat
Guo et al[59], 2020 Liraglutide 1.8 mg/dInsulin glargine or placeboT2D and NAFLD26 wkLiraglutide plus 2 gm metformin for 26 wk significantly reduced liver, subcutaneous and visceral fat
Zhang et al[60], 2020 Liraglutide 1.8 mg/dPioglitazone T2D and NAFLD24 wkLiraglutide reduced liver fat significantly compare to pioglitazone which could be attributed to weight loss
Liu et al[61], 2020 Exenatide 1.8 mg/d Insulin glargineT2D and NAFLD24 wkBoth reduced liver fat but exenatide led to higher reduction in body weight, visceral fat, liver enzymes
Miyake et al[62], 2022 (Trial Registration: jRCTs061210009)Semaglutide 0.5 mg/wk + Luseogliflozin 2.5 mg/dSemaglutide 0.5 mg/wk T2D and NASH52 wkOngoing study
Gastaldelli et al[63], 2022Dual GIP/GLP-1R agonist: tirzepatide10 mg or 15 mg/wk Insulin degludecT2D and NAFLD52 wkSignificant decrease in liver fat content, visceral and subcutaneous adipose tissue compared to insulin degludec
Kuchay et al[64], 2020 Glucagon-like peptide-1 receptor (GLP-1r) agonist (GLP-1 RA)Usual care T2D and NAFLD24 wkDulaglutide significantly reduces liver fat comma gamma glutamyl transpeptidase

GLP-1 RAs also reduce visceral and subcutaneous fat which is not seen with insulin which only affects glycemic control. The reduction in liver fat content with liraglutide was higher than that of pioglitazone[56-60]. Similarly, exenatide 1.8 mg/d reduced liver fat with a higher reduction in body weight, visceral fat, and liver enzymes as compared to insulin glargine[61].

Semaglutide is longer acting GLP-1 RA (half-life 165 h as compared to 13-15 h with liraglutide) which is used as a once-weekly subcutaneous injection (2.4 mg) and is as effective as once-daily liraglutide (3 mg) for weight loss[51,52,65]. However, daily semaglutide with lower doses (up to 0.4 mg) has been used in NASH trials which have shown significant dose-dependent NASH resolution (most pronounced with 0.4 mg) without worsening fibrosis [50,51].

Future directions and implications for NASH management

The tremendous redundancy of the downstream pathways leading to NASH progression warrants combination therapies rather than using single agents. Blocking a single pathway was not shown to be very effective. The best effects are seen with agents that target the underlying metabolic dysregulation with pleiotropic effects such as incretins.

One such approach is combining semaglutide (GLP-1 RA) with small molecules such as firsocostat (acetyl-CoA carboxylase- rate-controlling enzyme for de novo lipogenesis) and cilofexor (second generation farnesoid X receptor agonist). The combination was safe and led to a greater reduction in liver fat compared to semaglutide or a combination of two agents.

GIP reduces ectopic fat accumulation and has anti-inflammatory effects (reduced macrophage polarization and cytokine production). More importantly, GIP ameliorates nausea induced by higher doses of GLP-1 allowing the latter to be used at higher doses. Dual GIP/GLP-1 receptor agonist tirzepatide is being studied in phase III trials for NASH (SYNERGY-NASH). Initial data in T2D and NAFLD showed a significantly higher decrease in liver, subcutaneous, and visceral fat as compared to insulin degludec[63]. Once weekly semaglutide (0.5 mg/wk) is being studied with sodium glucose co-transporter 2 inhibitors (Luseogliflozin 2.5 mg/d) for T2D with NASH[62].

GLP-1 is also being combined with glucagon for NASH. Glucagon has direct liver receptors whereas effects of GLP-1 RAs are based on extra-hepatic effects. Glucagon has hepatic receptors where it has anti-inflammatory effects, reduces bile acid production/hepatic stellate cell activation, inhibits transforming growth factor signaling, reduces collagen production, and increases turnover. Dulaglutide, a GLP-1 RA, is being studied in NASH. Earlier studies in T2D and NAFLD showed a reduction in liver fat[64]. GLP-1/glucagon receptor co-agonist efinopegdutide 10 mg weekly resulted in a greater reduction in liver fat than weekly semaglutide (1 mg) in a phase IIa study[52]. Histology-based trials in NASH are warranted.

Challenges and promise of incretin-based therapies for NASH

Selecting the right category of patients for incretin-based therapy needs further exploration. Disease activity (inflammation), stage (fibrosis), and comorbidities (e.g., T2D) need to be considered. The long-term safety of these therapies in NASH needs further study. The holistic approach of combination treatment including incretin-based therapies beyond glycemic control is a potential paradigm shift in NASH management and is an area of ongoing research. Collaborative efforts among healthcare providers such as primary care doctors, hepatologists, and endocrinologists are warranted.

CONCLUSION

In conclusion, specific targeted interventions are essential for NAFLD in T2D. This is because of the alarmingly high prevalence of NAFLD in T2D, their synergistic effect on hepatic and extra-hepatic complications, and the bidirectional link driven by alterations in glucose/Lipid metabolism along with insulin resistance. PPAR agonists such as pioglitazone have been shown to improve liver outcomes by regulating lipid metabolism, glucose homeostasis, and inflammation. Newer dual and pan-PPAR agonists show promising results with minimized side effects in NAFLD/NASH with T2D. Incretin-based therapies, (GLP-1RAs and DPP-4 inhibitors) act in T2D-related NAFLD/NASH by glycemic control as well their exhibit pleiotropic effects such as anti-inflammatory and anti-fibrotic actions. Liraglutide and semaglutide have demonstrated remarkable efficacy in resolving NASH without worsening fibrosis apart from improving metabolic parameters. Given the multifactorial nature of NASH pathogenesis, a combination of incretin-based therapies with small molecules targeting de novo lipogenesis and sodium glucose co-transporter 2 inhibitors are upcoming approaches for treating NASH. Dual GLP-1/glucagon receptor agonists with direct hepatic effects of glucagon affecting fibrosis could indicate a paradigm shift in NASH management. The current challenges include the proper selection of patients for incretin-based therapies, concerns regarding long-term safety, and the need for a multifaceted approach. As we better understand the intricate interplay between T2D and NAFLD, PPAR agonists and incretin-based therapies offer hope for more effective and targeted management of NAFLD/NASH in T2D.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country/Territory of origin: India

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Geng TY, China S-Editor: Liu JH L-Editor: A P-Editor: Zhao S

References
1.  Younossi ZM, Golabi P, de Avila L, Paik JM, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 773]  [Cited by in F6Publishing: 1150]  [Article Influence: 230.0]  [Reference Citation Analysis (0)]
2.  Mantovani A, Byrne CD, Scorletti E, Mantzoros CS, Targher G. Efficacy and safety of anti-hyperglycaemic drugs in patients with non-alcoholic fatty liver disease with or without diabetes: An updated systematic review of randomized controlled trials. Diabetes Metab. 2020;46:427-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 70]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
3.  Targher G, Corey KE, Byrne CD, Roden M. The complex link between NAFLD and type 2 diabetes mellitus - mechanisms and treatments. Nat Rev Gastroenterol Hepatol. 2021;18:599-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 329]  [Article Influence: 109.7]  [Reference Citation Analysis (0)]
4.  Gastaldelli A, Cusi K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Rep. 2019;1:312-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 221]  [Article Influence: 44.2]  [Reference Citation Analysis (0)]
5.  Marra F, Gastaldelli A, Svegliati Baroni G, Tell G, Tiribelli C. Molecular basis and mechanisms of progression of non-alcoholic steatohepatitis. Trends Mol Med. 2008;14:72-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 305]  [Cited by in F6Publishing: 322]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
6.  Dela Peña A, Leclercq I, Field J, George J, Jones B, Farrell G. NF-kappaB activation, rather than TNF, mediates hepatic inflammation in a murine dietary model of steatohepatitis. Gastroenterology. 2005;129:1663-1674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 232]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
7.  Luukkonen PK, Sädevirta S, Zhou Y, Kayser B, Ali A, Ahonen L, Lallukka S, Pelloux V, Gaggini M, Jian C, Hakkarainen A, Lundbom N, Gylling H, Salonen A, Orešič M, Hyötyläinen T, Orho-Melander M, Rissanen A, Gastaldelli A, Clément K, Hodson L, Yki-Järvinen H. Saturated Fat Is More Metabolically Harmful for the Human Liver Than Unsaturated Fat or Simple Sugars. Diabetes Care. 2018;41:1732-1739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 191]  [Article Influence: 31.8]  [Reference Citation Analysis (1)]
8.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 224]  [Article Influence: 112.0]  [Reference Citation Analysis (0)]
9.  Björkström K, Franzén S, Eliasson B, Miftaraj M, Gudbjörnsdottir S, Trolle-Lagerros Y, Svensson AM, Hagström H. Risk Factors for Severe Liver Disease in Patients With Type 2 Diabetes. Clin Gastroenterol Hepatol. 2019;17:2769-2775.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
10.  Ajmera V, Cepin S, Tesfai K, Hofflich H, Cadman K, Lopez S, Madamba E, Bettencourt R, Richards L, Behling C, Sirlin CB, Loomba R. A prospective study on the prevalence of NAFLD, advanced fibrosis, cirrhosis and hepatocellular carcinoma in people with type 2 diabetes. J Hepatol. 2023;78:471-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 53]  [Article Influence: 53.0]  [Reference Citation Analysis (0)]
11.  Jarvis H, Craig D, Barker R, Spiers G, Stow D, Anstee QM, Hanratty B. Metabolic risk factors and incident advanced liver disease in non-alcoholic fatty liver disease (NAFLD): A systematic review and meta-analysis of population-based observational studies. PLoS Med. 2020;17:e1003100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 197]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
12.  Huang DQ, Noureddin N, Ajmera V, Amangurbanova M, Bettencourt R, Truong E, Gidener T, Siddiqi H, Majzoub AM, Nayfeh T, Tamaki N, Izumi N, Yoneda M, Nakajima A, Idilman R, Gumussoy M, Oz DK, Erden A, Allen AM, Noureddin M, Loomba R. Type 2 diabetes, hepatic decompensation, and hepatocellular carcinoma in patients with non-alcoholic fatty liver disease: an individual participant-level data meta-analysis. Lancet Gastroenterol Hepatol. 2023;8:829-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 21]  [Article Influence: 21.0]  [Reference Citation Analysis (1)]
13.  Singh S, Allen AM, Wang Z, Prokop LJ, Murad MH, Loomba R. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: a systematic review and meta-analysis of paired-biopsy studies. Clin Gastroenterol Hepatol. 2015;13:643-54.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 932]  [Cited by in F6Publishing: 1045]  [Article Influence: 116.1]  [Reference Citation Analysis (0)]
14.  Huang DQ, Wilson LA, Behling C, Kleiner DE, Kowdley KV, Dasarathy S, Amangurbanova M, Terrault NA, Diehl AM, Chalasani N, Neuschwander-Tetri BA, Sanyal AJ, Tonascia J; NASH Clinical Research Network, Loomba R. Fibrosis Progression Rate in Biopsy-Proven Nonalcoholic Fatty Liver Disease Among People With Diabetes Versus People Without Diabetes: A Multicenter Study. Gastroenterology. 2023;165:463-472.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 27]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
15.  Castera L, Laouenan C, Vallet-Pichard A, Vidal-Trécan T, Manchon P, Paradis V, Roulot D, Gault N, Boitard C, Terris B, Bihan H, Julla JB, Radu A, Poynard T, Brzustowsky A, Larger E, Czernichow S, Pol S, Bedossa P, Valla D, Gautier JF; QUID-NASH investigators. High Prevalence of NASH and Advanced Fibrosis in Type 2 Diabetes: A Prospective Study of 330 Outpatients Undergoing Liver Biopsies for Elevated ALT, Using a Low Threshold. Diabetes Care. 2023;46:1354-1362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 15]  [Reference Citation Analysis (0)]
16.  Choudhary NS, Kumar N, Duseja A. Peroxisome Proliferator-Activated Receptors and Their Agonists in Nonalcoholic Fatty Liver Disease. J Clin Exp Hepatol. 2019;9:731-739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
17.  Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications--a review. Nutr J. 2014;13:17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 811]  [Cited by in F6Publishing: 771]  [Article Influence: 77.1]  [Reference Citation Analysis (0)]
18.  Schoeb TR. Klossiella sp infection in a galago. J Am Vet Med Assoc. 1984;185:1381-1382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 654]  [Cited by in F6Publishing: 699]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
19.  Cardella JF, Lukens ML, Fox PS. Fibrin sheath entrapment of peripherally inserted central catheters. J Vasc Interv Radiol. 1994;5:439-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 133]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
20.  Zhong X, Liu H. Honokiol attenuates diet-induced non-alcoholic steatohepatitis by regulating macrophage polarization through activating peroxisome proliferator-activated receptor γ. J Gastroenterol Hepatol. 2018;33:524-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
21.  Fernández-Miranda C, Pérez-Carreras M, Colina F, López-Alonso G, Vargas C, Solís-Herruzo JA. A pilot trial of fenofibrate for the treatment of non-alcoholic fatty liver disease. Dig Liver Dis. 2008;40:200-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 184]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
22.  Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, Rakela J, McGill DB. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology. 1996;23:1464-1467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 396]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
23.  Peters JM, Gonzalez FJ, Müller R. Establishing the Role of PPARβ/δ in Carcinogenesis. Trends Endocrinol Metab. 2015;26:595-607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 52]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
24.  Bays HE, Schwartz S, Littlejohn T 3rd, Kerzner B, Krauss RM, Karpf DB, Choi YJ, Wang X, Naim S, Roberts BK. MBX-8025, a novel peroxisome proliferator receptor-delta agonist: lipid and other metabolic effects in dyslipidemic overweight patients treated with and without atorvastatin. J Clin Endocrinol Metab. 2011;96:2889-2897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 122]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
25.  Ratziu V, Giral P, Jacqueminet S, Charlotte F, Hartemann-Heurtier A, Serfaty L, Podevin P, Lacorte JM, Bernhardt C, Bruckert E, Grimaldi A, Poynard T; LIDO Study Group. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the randomized placebo-controlled Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology. 2008;135:100-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 458]  [Cited by in F6Publishing: 443]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
26.  Aithal GP, Thomas JA, Kaye PV, Lawson A, Ryder SD, Spendlove I, Austin AS, Freeman JG, Morgan L, Webber J. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology. 2008;135:1176-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 522]  [Cited by in F6Publishing: 506]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
27.  Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, Neuschwander-Tetri BA, Lavine JE, Tonascia J, Unalp A, Van Natta M, Clark J, Brunt EM, Kleiner DE, Hoofnagle JH, Robuck PR; NASH CRN. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 2010;362:1675-1685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2215]  [Cited by in F6Publishing: 2217]  [Article Influence: 158.4]  [Reference Citation Analysis (2)]
28.  Ishibashi S, Yamashita S, Arai H, Araki E, Yokote K, Suganami H, Fruchart JC, Kodama T; K-877-04 Study Group. Effects of K-877, a novel selective PPARα modulator (SPPARMα), in dyslipidaemic patients: A randomized, double blind, active- and placebo-controlled, phase 2 trial. Atherosclerosis. 2016;249:36-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 131]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
29.  Araújo S, Soares E Silva A, Gomes F, Ribeiro E, Oliveira W, Oliveira A, Lima I, Lima MDC, Pitta I, Peixoto C. Effects of the new thiazolidine derivative LPSF/GQ-02 on hepatic lipid metabolism pathways in non-alcoholic fatty liver disease (NAFLD). Eur J Pharmacol. 2016;788:306-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
30.  Boettcher E, Csako G, Pucino F, Wesley R, Loomba R. Meta-analysis: pioglitazone improves liver histology and fibrosis in patients with non-alcoholic steatohepatitis. Aliment Pharmacol Ther. 2012;35:66-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 196]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
31.  Cusi K, Isaacs S, Barb D, Basu R, Caprio S, Garvey WT, Kashyap S, Mechanick JI, Mouzaki M, Nadolsky K, Rinella ME, Vos MB, Younossi Z. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 300]  [Article Influence: 150.0]  [Reference Citation Analysis (1)]
32.  Duseja A, Singh SP, De A, Madan K, Rao PN, Shukla A, Choudhuri G, Saigal S, Shalimar, Arora A, Anand AC, Das A, Kumar A, Eapen CE, Devadas K, Shenoy KT, Panigrahi M, Wadhawan M, Rathi M, Kumar M, Choudhary NS, Saraf N, Nath P, Kar S, Alam S, Shah S, Nijhawan S, Acharya SK, Aggarwal V, Saraswat VA, Chawla YK. Indian National Association for Study of the Liver (INASL) Guidance Paper on Nomenclature, Diagnosis and Treatment of Nonalcoholic Fatty Liver Disease (NAFLD). J Clin Exp Hepatol. 2023;13:273-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
33.  Ratziu V, Harrison SA, Francque S, Bedossa P, Lehert P, Serfaty L, Romero-Gomez M, Boursier J, Abdelmalek M, Caldwell S, Drenth J, Anstee QM, Hum D, Hanf R, Roudot A, Megnien S, Staels B, Sanyal A; GOLDEN-505 Investigator Study Group. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-α and -δ, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology. 2016;150:1147-1159.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 677]  [Cited by in F6Publishing: 725]  [Article Influence: 90.6]  [Reference Citation Analysis (0)]
34.  Hamrén B, Ohman KP, Svensson MK, Karlsson MO. Pharmacokinetic-pharmacodynamic assessment of the interrelationships between tesaglitazar exposure and renal function in patients with type 2 diabetes mellitus. J Clin Pharmacol. 2012;52:1317-1327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
35.  Lincoff AM, Tardif JC, Schwartz GG, Nicholls SJ, Rydén L, Neal B, Malmberg K, Wedel H, Buse JB, Henry RR, Weichert A, Cannata R, Svensson A, Volz D, Grobbee DE; AleCardio Investigators. Effect of aleglitazar on cardiovascular outcomes after acute coronary syndrome in patients with type 2 diabetes mellitus: the AleCardio randomized clinical trial. JAMA. 2014;311:1515-1525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 184]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
36.  Nissen SE, Wolski K, Topol EJ. Effect of muraglitazar on death and major adverse cardiovascular events in patients with type 2 diabetes mellitus. JAMA. 2005;294:2581-2586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 424]  [Cited by in F6Publishing: 399]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
37.  Gawrieh S, Noureddin M, Loo N, Mohseni R, Awasty V, Cusi K, Kowdley KV, Lai M, Schiff E, Parmar D, Patel P, Chalasani N. Saroglitazar, a PPAR-α/γ Agonist, for Treatment of NAFLD: A Randomized Controlled Double-Blind Phase 2 Trial. Hepatology. 2021;74:1809-1824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 159]  [Article Influence: 53.0]  [Reference Citation Analysis (0)]
38.  Rajesh NA, Drishya L, Ambati MMR, Narayanan AL, Alex M, R KK, Abraham JJ, Vijayakumar TM. Safety and Efficacy of Saroglitazar in Nonalcoholic Fatty Liver Patients With Diabetic Dyslipidemia-A Prospective, Interventional, Pilot Study. J Clin Exp Hepatol. 2022;12:61-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
39.  Tenenbaum A, Fisman EZ. Balanced pan-PPAR activator bezafibrate in combination with statin: comprehensive lipids control and diabetes prevention? Cardiovasc Diabetol. 2012;11:140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
40.  Wettstein G, Luccarini JM, Poekes L, Faye P, Kupkowski F, Adarbes V, Defrêne E, Estivalet C, Gawronski X, Jantzen I, Philippot A, Tessier J, Tuyaa-Boustugue P, Oakley F, Mann DA, Leclercq I, Francque S, Konstantinova I, Broqua P, Junien JL. The new-generation pan-peroxisome proliferator-activated receptor agonist IVA337 protects the liver from metabolic disorders and fibrosis. Hepatol Commun. 2017;1:524-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 100]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
41.  Jain MR, Giri SR, Bhoi B, Trivedi C, Rath A, Rathod R, Ranvir R, Kadam S, Patel H, Swain P, Roy SS, Das N, Karmakar E, Wahli W, Patel PR. Dual PPARα/γ agonist saroglitazar improves liver histopathology and biochemistry in experimental NASH models. Liver Int. 2018;38:1084-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 141]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
42.  Bhosle D, Bhosle V, Bobde J, Bhagat A, Shaikh H, Kadam R. Study of Saroglitazar in Treatment Of Pre-diabetes with Dyslipidemia: STOP-D. J Assoc Physicians India. 2018;66:14-17.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013;17:819-837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 918]  [Cited by in F6Publishing: 955]  [Article Influence: 86.8]  [Reference Citation Analysis (0)]
44.  Andreasen CR, Andersen A, Vilsbøll T. The future of incretins in the treatment of obesity and non-alcoholic fatty liver disease. Diabetologia. 2023;66:1846-1858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
45.  Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M. From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol. 2019;16:411-428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 814]  [Cited by in F6Publishing: 805]  [Article Influence: 161.0]  [Reference Citation Analysis (0)]
46.  Muthiah MD, Sanyal AJ. Current management of non-alcoholic steatohepatitis. Liver Int. 2020;40 Suppl 1:89-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 47]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
47.  Wang XC, Gusdon AM, Liu H, Qu S. Effects of glucagon-like peptide-1 receptor agonists on non-alcoholic fatty liver disease and inflammation. World J Gastroenterol. 2014;20:14821-14830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 61]  [Cited by in F6Publishing: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
48.  Lee J, Hong SW, Rhee EJ, Lee WY. GLP-1 Receptor Agonist and Non-Alcoholic Fatty Liver Disease. Diabetes Metab J. 2012;36:262-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
49.  O'Neil PM, Birkenfeld AL, McGowan B, Mosenzon O, Pedersen SD, Wharton S, Carson CG, Jepsen CH, Kabisch M, Wilding JPH. Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial. Lancet. 2018;392:637-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 378]  [Article Influence: 63.0]  [Reference Citation Analysis (0)]
50.  Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, Sanyal AJ, Sejling AS, Harrison SA; NN9931-4296 Investigators. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N Engl J Med. 2021;384:1113-1124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 513]  [Cited by in F6Publishing: 780]  [Article Influence: 260.0]  [Reference Citation Analysis (0)]
51.  Loomba R, Abdelmalek MF, Armstrong MJ, Jara M, Kjær MS, Krarup N, Lawitz E, Ratziu V, Sanyal AJ, Schattenberg JM, Newsome PN; NN9931-4492 investigators. Semaglutide 2·4 mg once weekly in patients with non-alcoholic steatohepatitis-related cirrhosis: a randomised, placebo-controlled phase 2 trial. Lancet Gastroenterol Hepatol. 2023;8:511-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 96]  [Article Influence: 96.0]  [Reference Citation Analysis (0)]
52.  Romero-Gómez M, Lawitz E, Shankar RR, Chaudhri E, Liu J, Lam RLH, Kaufman KD, Engel SS; MK-6024 P001 Study Group. A phase IIa active-comparator-controlled study to evaluate the efficacy and safety of efinopegdutide in patients with non-alcoholic fatty liver disease. J Hepatol. 2023;79:888-897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 31]  [Reference Citation Analysis (0)]
53.  Alkhouri N, Herring R, Kabler H, Kayali Z, Hassanein T, Kohli A, Huss RS, Zhu Y, Billin AN, Damgaard LH, Buchholtz K, Kjær MS, Balendran C, Myers RP, Loomba R, Noureddin M. Safety and efficacy of combination therapy with semaglutide, cilofexor and firsocostat in patients with non-alcoholic steatohepatitis: A randomised, open-label phase II trial. J Hepatol. 2022;77:607-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 85]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
54.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 66]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
55.  Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, Hazlehurst JM, Guo K; LEAN trial team, Abouda G, Aldersley MA, Stocken D, Gough SC, Tomlinson JW, Brown RM, Hübscher SG, Newsome PN. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1100]  [Cited by in F6Publishing: 1209]  [Article Influence: 151.1]  [Reference Citation Analysis (1)]
56.  Khoo J, Hsiang J, Taneja R, Law NM, Ang TL. Comparative effects of liraglutide 3 mg vs structured lifestyle modification on body weight, liver fat and liver function in obese patients with non-alcoholic fatty liver disease: A pilot randomized trial. Diabetes Obes Metab. 2017;19:1814-1817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
57.  Yan J, Yao B, Kuang H, Yang X, Huang Q, Hong T, Li Y, Dou J, Yang W, Qin G, Yuan H, Xiao X, Luo S, Shan Z, Deng H, Tan Y, Xu F, Xu W, Zeng L, Kang Z, Weng J. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 151]  [Article Influence: 30.2]  [Reference Citation Analysis (0)]
58.  Bizino MB, Jazet IM, de Heer P, van Eyk HJ, Dekkers IA, Rensen PCN, Paiman EHM, Lamb HJ, Smit JW. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 62]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
59.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
60.  Zhang LY, Qu XN, Sun ZY, Zhang Y. Effect of liraglutide therapy on serum fetuin A in patients with type 2 diabetes and non-alcoholic fatty liver disease. Clin Res Hepatol Gastroenterol. 2020;44:674-680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
61.  Liu L, Yan H, Xia M, Zhao L, Lv M, Zhao N, Rao S, Yao X, Wu W, Pan B, Bian H, Gao X. Efficacy of exenatide and insulin glargine on nonalcoholic fatty liver disease in patients with type 2 diabetes. Diabetes Metab Res Rev. 2020;36:e3292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 68]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
62.  Miyake T, Yoshida O, Matsuura B, Furukawa S, Hirooka M, Abe M, Tokumoto Y, Koizumi Y, Watanabe T, Takeshita E, Sunago K, Yukimoto A, Watanabe K, Miyazaki M, Kanzaki S, Nakaguchi H, Koizumu M, Yamamoto Y, Kumagi T, Hiasa Y. Additional Effect of Luseogliflozin on Semaglutide in Nonalcoholic Steatohepatitis Complicated by Type 2 Diabetes Mellitus: An Open-Label, Randomized, Parallel-Group Study. Diabetes Ther. 2022;13:1083-1096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
63.  Gastaldelli A, Cusi K, Fernández Landó L, Bray R, Brouwers B, Rodríguez Á. Effect of tirzepatide versus insulin degludec on liver fat content and abdominal adipose tissue in people with type 2 diabetes (SURPASS-3 MRI): a substudy of the randomised, open-label, parallel-group, phase 3 SURPASS-3 trial. Lancet Diabetes Endocrinol. 2022;10:393-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 166]  [Article Influence: 83.0]  [Reference Citation Analysis (0)]
64.  Kuchay MS, Krishan S, Mishra SK, Choudhary NS, Singh MK, Wasir JS, Kaur P, Gill HK, Bano T, Farooqui KJ, Mithal A. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 101]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
65.  Rubino DM, Greenway FL, Khalid U, O'Neil PM, Rosenstock J, Sørrig R, Wadden TA, Wizert A, Garvey WT; STEP 8 Investigators. Effect of Weekly Subcutaneous Semaglutide vs Daily Liraglutide on Body Weight in Adults With Overweight or Obesity Without Diabetes: The STEP 8 Randomized Clinical Trial. JAMA. 2022;327:138-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 271]  [Article Influence: 135.5]  [Reference Citation Analysis (0)]