Skip to main content
Journal of Clinical and Experimental Hepatology logoLink to Journal of Clinical and Experimental Hepatology
. 2019 Jul 2;9(6):731–739. doi: 10.1016/j.jceh.2019.06.004

Peroxisome Proliferator-Activated Receptors and Their Agonists in Nonalcoholic Fatty Liver Disease

Narendra S Choudhary , Naveen Kumar , Ajay Duseja ‡,
PMCID: PMC6926194  PMID: 31889755

Abstract

Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide. In addition to the liver-related morbidity and mortality, NAFLD is now also associated with various extrahepatic diseases. Pathogenesis of NAFLD is multifactorial with limited pharmacotherapy options for the treatment of patients with NAFLD. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that are involved in the transcriptional regulation of lipid metabolism, glucose homeostasis, energy balance, inflammation, and atherosclerosis. PPAR agonists are attractive options for treatment of NAFLD as they can act at multiple targets involved in the pathogenesis of NAFLD. We reviewed the available literature on the pathophysiological role of PPARs and use of PPAR agonists in the treatment of NAFLD. Original studies and review articles available on PubMed regarding the role of PPARs in the pathogenesis and utility of PPAR agonists in the treatment of NAFLD were included in this review article. ClinicalTrials.gov and Clinical Trials Registry-India sites were searched for ongoing studies on saroglitazar. The available literature suggests that PPARs play an important role in the pathogenesis of NAFLD. Use of PPAR gamma agonists is associated with histological improvement in NAFLD. Dual PPAR agonists with no or minimal PPAR gamma activity are being explored in the treatment of NAFLD. Because of the pathophysiological role of PPARs in NAFLD, PPAR agonists are attractive options for the treatment of patients with NAFLD. Dual PPAR agonists without significant gamma activity appear promising for the treatment of NAFLD.

Keywords: nonalcoholic steatohepatitis, hepatic steatosis, fatty liver, metabolic syndrome

Abbreviations: CPT-1, Carnitine palmitoyltransferase-1; MPC, mitochondrial pyruvate carrier; NAFLD, nonalcoholic fatty liver disease; NASH, Nonalcoholic steatohepatitis; NF-κB, nuclear factor-kappa beta; OR, odds ratio; PPAR, Peroxisome proliferator-activated receptors; RXR, retinoid X receptor; TZDs, thiazolidinediones


Peroxisome proliferator-activated receptors (PPARs) are a ligand-activated transcription factor belonging to a nuclear receptor subfamily that is involved in the transcriptional regulation of lipid metabolism, glucose homeostasis, energy balance, inflammation, and atherosclerosis. There are three PPAR isoforms, alpha (α), beta (β)/delta (δ), and gamma (γ), which are differentially expressed in various tissues.1, 2 PPARα is expressed ubiquitously but is largely present in the liver. PPARβ/δ is expressed mainly in skeletal muscles and to a lesser degree in adipose tissue and skin. PPARγ is highly expressed in adipose tissue (Table 1).3, 4, 5, 6, 7, 8

Table 1.

Types and Actions of PPARs (Based on Grygiel-Górniak et al3, Pawlak et al4, Tailleux et al5, Corton6, Fisher et al7, and Fisher et al8).

Type Tissue distributiona Gene targets Natural ligand Synthetic agonists Effect
PPARα Liver
Muscle
Heart
Kidney
Β oxidation
Fatty acid transport protein
Fatty acid translocase
Lipoprotein lipase
Apolipoprotein A-I and A-II
Sterol 12-hydroxylase
Unsaturated fatty acids
Leukotriene B4
8-Hydroxyeicosatetraenoic acid
Gemfibrozil
Fenofibrate
Clofibrate
Fatty acid oxidation
Anti-inflammatory
PPARβ/δ Ubiquitous, muscle, gastrointestinal, adipose tissue
Macrophages
Heart
Genes involved in lipid uptake, metabolism, and efflux (repressed by PPARs) Unsaturated fatty acids
Carbaprostacyclin
Components of VLDL
GW501516
GW0742
MBX-8025
Glucose homeostasis, insulin sensitivity
PPARγ Adipose tissue
Liver, kidney, intestine
Fatty acid–binding protein (aP2)
Fatty acid transport protein
Fatty acid translocase
Unsaturated fatty acids, 15-hydroxyeicosatetraenoic acid, 9- and 13- hydroxyoctadecadienoic acid, 15-deoxy-12,14-prostaglandin J2, prostaglandin PGJ2 Thiazolidinediones Adipogenesis
Insulin sensitization, glucose homeostasis
Fatty acid oxidation

PPAR, peroxisome proliferator-activated receptor; VLDL, very low-density lipoprotein.

bAll tissues not shown.

a

Tissues in bold fonts indicate main tissue expression.

Free fatty acids (FAs), eicosanoids, and various complex lipids are considered endogenous PPAR ligands. Exogenous ligands are environmental and pharmaceutical molecules that can activate various PPAR family receptors to varying degrees.6, 7, 8, 9 PPARs form a heterodimer with the retinoid X receptor (RXR) after binding to the ligand and bind to response elements that regulate the expression of genes encoding proteins involved in beta oxidation, FA uptake, adipogenesis, and adipocyte differentiation.3, 10, 11, 12 PPAR ligands have emerged as potential therapeutics for nonalcoholic fatty liver disease (NAFLD) (Table 1).

NAFLD has emerged as the most common liver disease in the world, including Asia Pacific, and is responsible for significant liver disease burden.13, 14 The spectrum of NAFLD ranges from nonalcoholic fatty liver to nonalcoholic steatohepatitis (NASH), which has the propensity to progress on to cirrhosis of the liver and hepatocellular carcinoma.15 NAFLD is characterized by presence of insulin resistance, dyslipidemia, and a proinflammatory state. The mainstay of current treatment of NAFLD is weight loss by lifestyle modification, which is difficult to achieve and sustain for most of the patients. There are limited pharmacotherapy options, and the focus of treatment has largely been on patients with progressive NASH.15 PPARs affect glucose homeostasis (insulin-sensitizing properties), inflammation, and atherogenesis and control dyslipidemia. Thus, these agents should act at multiple levels in NAFLD pathogenesis, which makes them an attractive target for drug development.

PPARα and its agonists

PPARα is expressed in many mammalian cells and tissues such as the liver, kidney, heart, muscle, adipose tissue, and others including immune cells (e.g., macrophages).16 PPARα plays a role in multiple regulatory functions. In the liver, it plays a crucial role in FA oxidation, which provides energy for peripheral tissues, and has a potential role in the oxidant/antioxidant pathway.17 PPARα also has anti-inflammatory effects through complex regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB).18 The activation of PPARα occurs after dimerization with the RXR, resulting in formation of a multiprotein complex with protein coactivators. After activation, PPARα binds to responsive elements in DNA, resulting in the transcription of various anti-inflammatory proteins, such as the kB-α inhibitor.19 Carnitine palmitoyltransferase 1 (CPT-1) is a pivotal enzyme that allows the FA to go through the inner mitochondrial membrane and reach the mitochondrial matrix for further metabolism.20 Reduction of PPARα expression in the liver causes impairment of the transcription of its target gene CPT-1, and excessive FAs tend to accumulate in the form of triglycerides.21, 22

PPARα acts as a nutritional sensor, which allows adaptation of the rates of FA oxidation, lipogenesis, and ketone body synthesis, in response to feeding and fasting.23 During excessive dietary intake of lipids, hepatic PPARα expression decreases.24, 25 Studies in mice with genetic deletion of PPARα (PPARα−/−) and high-fat diet resulted in accumulation of more hepatic triglycerides with a significantly higher NAFLD activity score (NAS) compared with wild-type (WT) controls.26, 27 Mice with the PPARα gene knockout, fed with a high-fat diet, also showed increased markers of oxidative stress, inflammation, and cell death.28

Natural agonists of PPARs include FAs, eicosanoids, and phospholipids derived from cellular FA metabolism or from dietary lipids. The synthetic ligands include fibrates, thiazolidinediones (TZDs), glitazars, elafibranor, and several others.29 In the mice model, the PPARα agonist Wy-14643 prevented methionine and choline deficient (MCD) diet-induced hepatic triglyceride accumulation in WT mice, but it had no effect on PPARα−/− mice.30

Fibrates are considered PPARα agonists, although less potent than the PPARα agonist Wy-14643. Several studies have shown improvement of biochemical or histological parameters with fibrates in patients with NAFLD.31, 32, 33 Fibrates, however, have limited clinical efficacy because they are weak PPARα agonists and have significant adverse events. Fernandez-Miranda et al used 200 mg/day fenofibrate for 48 weeks in a series of 16 patients with NAFLD. The authors noted a significant decrease in levels of triglycerides, glucose, and gamma-glutamyl transpeptidase and proportion of patients with raised transaminases or metabolic syndrome and a trend of decreasing insulin resistance. The repeat biopsy at the end of the study showed decreased ballooning, but other histologic parameters did not improve significantly.31 El-Haggar et al compared fenofibrate with fenofibrate plus pentoxifylline for 24 weeks. The addition of pentoxifylline to fenofibrate did not improve lipid parameters, but a beneficial effect on indirect markers of hepatic fibrosis, inflammation, insulin resistance, and liver stiffness was present.33 Laurin et al34 compared clofibrate with ursodeoxycholic acid in a 1-year study. The authors could not find any advantage of clofibrate in patients with NAFLD. Newer selective PPARα-specific agonists, known as selective PPARα modulators, are in different phases of development.35, 36

PPARβ/δ and its agonists

PPARβ/δ is the least studied among all the PPAR isotypes, although it has significant expression in tissues controlling lipid metabolism, such as adipocytes, heart, skeletal muscle, liver, and macrophages.37 In the liver, PPARβ/δ is well expressed in hepatocytes but is also expressed in Kupffer cells and hepatic stellate cells, suggesting a potential role in inflammation and fibrosis.38 The PPARβ/δ agonist, GW501516, has been shown to ameliorate obesity and insulin resistance in rats.39 In a small 2-week pilot trial on six subjects, it was found by magnetic resonance imaging that GW501516 resulted in reduction of liver fat along with reduction in serum triglycerides and low-density lipoprotein cholesterol.40 However, clinical development of GW501516 was abandoned owing to development of cancer in preclinical models.41 Another PPARβ/δ agonist, GW0742, improved insulin signaling and reduced hepatic steatosis in a rat model.42

In a study using GW0742, the activation of PPARβ/δ was found to inhibit CCl4-induced liver toxicity through the PPARβ/δ-dependent downregulation of proinflammatory signaling through interactions between PPARβ/δ and NF-κB.43 A novel PPARβ/δ agonist, MBX-8025, was evaluated in a randomized, double-blind, placebo-controlled study. The study included overweight subjects with dyslipidemia and found that treatment with MBX-8025 (seladelpar) resulted in favorable lipid profiles and decreased liver enzymes.44 In a randomized study of MBX-8025 versus vehicle (1% methylcellulose), MBX-8025 normalized hyperglycemia, hyperinsulinemia, and glucose disposal in foz/foz mice. MBX-8025 reduced alanine aminotransferase and normalized serum lipids. There was significantly less steatosis, inflammation, ballooning, apoptosis, and fibrosis in the MBX-8025 arm.45

At this time, data regarding effectiveness of PPARβ/δ in treatment of NAFLD are limited to derive any conclusion.

PPARγ and its agonists

PPARγ is most highly expressed in adipose tissue, where it serves an important role in the regulation of adipocyte differentiation, adipogenesis, and lipid metabolism.46 In an animal model of NASH, Zhong and Liu47 showed that the activation of PPARγ regulates the polarization of the macrophages to M2 subtype. Kupffer cells have proinflammatory (M1) and anti-inflammatory (M2) subtypes; thus, change toward the M2 subtype may prevent development of NAFLD.47

TZDs are the most widely investigated PPARγ agonists. TZDs represent a class of clinically used insulin-sensitizing drugs, which currently include rosiglitazone and pioglitazone. TZDs promote the uptake and storage of FAs in adipose tissue, increasing adipose tissue mass while sparing the skeletal muscle and the liver.48 In an animal model of NASH induced with a choline–methionine–deficient diet, rosiglitazone prevented the development of NASH.49 In another animal model, Deng et al50 demonstrated that pioglitazone has anti-inflammatory and antifibrotic effects by repressing the expression level of platelet-derived growth factor and tissue inhibitor of metalloproteinase-2.

Caldwell et al reported data of troglitazone treatment in 10 female patients with histological NASH including 3 with compensated cirrhosis. Troglitazone was given for ≤6 months. A repeat biopsy in responders (normal alanine aminotransferase (ALT) at the end of treatment) showed persistence of NASH in all; four patients had one-point improvement of necroinflammation.51 Troglitazone was associated with serious hepatotoxicity and was banned later.52 Ratziu et al53 studied the role of rosiglitazone in a randomized controlled trial (RCT) called the FLIRT trial. Thirty-two patients on rosiglitazone were compared with 31 patients on placebo. The rosiglitazone arm had improved steatosis (47% vs 16%) and transminases (38% vs 7%) at the end of 1 year as compared with the placebo arm. There was no improvement in fibrosis and the NAFLD activity score. Weight gain happened significantly in the rosiglitazone arm, and dose reduction/discontinuation happened in some patients owing to painful swollen legs.53

In the PIVENS trial involving patients with NASH randomized to receive pioglitazone, vitamin E, or a placebo for 96 weeks, pioglitazone demonstrated a reduction in hepatic steatosis, lobular inflammation, and hepatic enzymes aspartate aminotransferase (AST) and ALT.54 A meta-analysis evaluated randomized placebo-controlled trials using TZDs in the treatment of patients with NASH. In the identified four high-quality randomized trials, treatment with TZDs resulted in significant decrease in serum ALT along with improvement in steatosis, inflammation, and hepatocyte ballooning, but the change in fibrosis was not statistically significant.55 But when three studies evaluating the effectiveness of pioglitazone were included, the improvement in fibrosis became statistically significant, suggesting that pioglitazone has superior effects on reversing hepatic fibrosis compared with rosiglitazone.55

The use of PPARγ agonists is associated with pedal edema, weight gain, cardiac adverse events, and risk of bladder cancer. A recent meta-analysis of 26 studies including 15,332 patients with type 2 diabetes mellitus (T2DM) showed that the odds ratio (OR) for edema with TZDs was 2.26 (95% confidence interval [CI], 2.02–2.53).55 TZDs are also associated with risk of cardiac adverse events. A meta-analysis of 3 RCTs showed an OR of 2.1, and 4 observation studies showed an OR of 1.55 for heart failure with TZDs.56 Rosiglitazone is associated with more cardiac adverse events than pioglitazone.57 The use of pioglitazone was found to be associated with risk of bladder cancer in RCTs (OR, 1.84; 95% CI, 0.99–3.42) and in observational studies (OR, 1.13; 95% CI, 1.03–1.25). The risk of bladder cancer with pioglitazone increased in a dose- and time-dependent manner.58

Table 2 summarizes histology-based studies of PPAR agonists in patients with NAFLD.31, 34, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 68

Table 2.

Summary of Studies of PPAR Agonists in NAFLD.

Author N, treatment duration Results
Biochemical response Histological response
Fernandez-Miranda et al31 16, fenofibrate for 48 weeks, no comparative group Significant decrease in triglyceride, glucose, liver enzymes, MS Decreased ballooning, grade steatosis, inflammation/fibrosis—no change
Laurin et al34 16 clofibrate arm, 24 UDCA arm, 1 year No change in the clofibrate arm other than decreased ALP No change in the clofibrate arm
Neuschwander-Tetri et al60 N = 30, 22 had paired biopsies, rosiglitazone for 48 weeks, no comparative group Decreased AST, ALT, and GGT Improved steatosis and ballooning, weight gain occurred in 67% of patients, and the median weight increase was 7.3%. Within 6 months of completing treatment, liver enzyme levels had increased to near-pretreatment levels
Ratziu et al53 RCT, 32 rosiglitazone
31 placebo
Normalized transaminase levels (38% vs 7%, P = .005) Improved steatosis (47% vs 16%; P = .014), although only half of the patients responded, no change of other histologic parameters, weight gain in rosiglitazone
Torres et al61 RCT, n = 137, rosiglitazone and metformin versus rosiglitazone and losartan versus rosiglitazone alone, 48 weeks Decreased transaminases in all groups 108 completed the study, overall improvement of all histologic parameters, no added benefit of metformin (did not prevent weight gain) or losartan
Belfort et al62 RCT, 6 months of low-calorie diet with pioglitazone (n = 26) or diet and placebo (n = 21) Decreased transaminases in pioglitazone arm Improved steatosis, inflammation, and ballooning, no change of fibrosis, weight gain despite low-calorie diet in the pioglitazone arm
Aithal et al63 RCT, 12 months of diet, exercise, and either placebo or pioglitazone, 30 in placebo and 31 in the pioglitazone arm had paired biopsies Improvement of ALT and GGT Hepatocellular injury (P = .005), Mallory–Denk bodies (P = .004), and fibrosis (P = .05) were reduced in patients treated with pioglitazone, weight gain
Sanyal et al54 RCT, pioglitazone (n = 80), vitamin E (n = 83), placebo (n = 84), 96 weeks Improvement of transaminases in the vitamin E and pioglitazone arm Improvement in NASH as compared with placebo (vitamin E, P = .001), with (pioglitazone P = .04), both vitamin E and pioglitazone associated with significant reductions in steatosis, lobular inflammation. Improvement in fibrosis, weight gain in pioglitazone
Cusi et al64 RCT, 18 months, followed by an 18-month open-label phase with pioglitazone (n = 50) or placebo (n = 51) More normalization in the pioglitazome arm Pioglitazone is associated with a better NAS reduction and resolution of NASH, steatosis, inflammation, ballooning, no improvement in fibrosis, weight gain
Ratziu et al68 Elafibranor 120 mg, elafibranor 80 mg, and placebo Liver enzymes, lipids, glucose profiles, and markers of systemic inflammation significantly reduced in the elafibranor 120-mg group Elafibranor 120 mg superior to placebo, NASH resolution without worsening of fibrosis in 19% versus 12% in the placebo group (P = .045), based on a post hoc analysis for the modified definition

PPAR, peroxisome proliferator-activated receptor; NAFLD, nonalcoholic fatty liver disease; RCT, randomized controlled trial; NASH, nonalcoholic steatohepatitis; NAS, NAFLD activity score; MS, metabolic syndrome; AST, aspartate aminotransferase; ALT, alanine aminotransferase; UDCA, Ursodeoxycholic Acid; GGT, gamma-glutamyl transferase.

The mitochondrial target of TZD modulators bind and modulate activity of the mitochondrial pyruvate carrier (MPC). MSDC-0602 is the next-generation TZD, which has diminished ability to activate PPARγ.65 Improved insulin sensitivity by these compounds decreases lipolysis from adipose tissue and de novo lipogenesis. In addition, blocking pyruvate entry into the mitochondria should normalize the tricarboxylic acid cycle, which is increased in NAFLD. This normalization of the tricarboxylic acid cycle leads to decreased reactive oxygen species and cell damage signals, which should reduce inflammation and stellate cell activation.66 Genetic (selective knockout) or pharmacologic targeting of the MPC by MSDC-0602K has been shown to increase insulin sensitivity and to prevent or reverse NASH pathology in a mouse model.67

Dual and pan-PPAR agonists

Combining the role of PPARs can result in multiple actions useful to treat NAFLD. If a compound holds desired potency for different PPARs, it can be maximally beneficial with minimal undesired effects. Thus, dual or pan-PPAR agonists can produce antihyperlipidemic (PPARα) effect with insulin sensitization (PPARγ) and increase β-oxidation in the liver and skeletal muscle (α and β/δ) while limiting side effects of singular agonists such as weight gain/cardiac events with TZDs or improving limited clinical efficacy of fibrates. However, development of several agents has been terminated owing to safety concerns, and only few are in later phases of development.29

  • a)

    PPARα/δ and their agonists

In recent years, agonists of PPARs have arisen with affinity for binding to multiple isoforms owing to relatively nonselective ligand-binding pockets, known as dual agonists, and represent interesting therapeutic targets. A novel dual PPARα/δ agonist, GFT505, also known as elafibranor, has been studied in treating NASH. Using various rodent models of NASH, treatment with GFT505 (elafibranor) demonstrated improvement in histologic features of NASH and decreased hepatic triglyceride content, along with reduced expression of inflammatory cytokines and fibrosis markers.69

Elafibranor is an agonist of PPARα and PPARδ. Thus, it works on insulin sensitivity, glucose homeostasis, and lipid metabolism. The RCT by Ratziu et al68 (GOLDEN trial) included the following arms: elafibranor 80 mg (n = 93), elafibranor 120 mg (n = 91), and placebo (n = 92). The study was conducted in Europe and the United States, and the study duration was 52 weeks. The primary outcome was resolution of NASH without worsening of fibrosis. While there was no difference between elafibranor and placebo groups in the primary outcome in intention-to-treat analysis, NASH resolved without worsening of fibrosis in a higher proportion of patients in the 120-mg elafibranor group (19%) versus the placebo group (12%, P = .045), based on a post hoc analysis for the modified definition. The modified definition defined resolution of NASH as disappearance of ballooning with either disappearance of lobular inflammation or persistence of mild inflammation only. Thus, the modified target was pathologic diagnosis of steatosis alone or steatosis with mild inflammation only. The authors found improvement in the histological score after removing patients with mild steatohepatitis from analysis. The outcomes in the 80-mg dose arm were not better than those of placebo. In addition, liver enzymes, lipids, glucose profiles, and markers of systemic inflammation were significantly reduced in the elafibranor 120-mg group versus the placebo group. Elafibranor was well tolerated and did not result in weight gain or cardiac events, the authors noted a mild and reversible increase in serum creatinine (4.31 ± 1.19 mmol/L).68 A phase III study (RESOLVE-IT) is presently recruiting a large number of patients with NASH to study the efficacy of elafibranor in comparison with placebo (ClinicalTrials.gov. NCT02704403).

  • b)

    PPARα/γ and their agonists

Compounds working as PPARα/γ agonists are called glitazars. These compounds improve dyslipidemia, which is α-action, and glycemic parameters/insulin sensitivity, which is γ-action. Thus, theoretically, these compounds address 2 important issues of NAFLD, dyslipidemia and insulin resistance and thus are the area of interest. Several glitazars were initially tried, but their clinical development was stopped later owing to adverse events. Tesaglitazar was the first dual PPARα/γ agonist; a large dose was needed owing to weak action, and clinical development was stopped owing to nephrotoxicity.70 Muraglitazar produced better lipid changes, but increase in cardiovascular events was observed.71, 72 AleCardio was a phase 3 multicenter randomized placebo-controlled trial that was conducted across 26 countries. A total of 7226 patients hospitalized for myocardial infarction or unstable angina with type 2 diabetes received either aleglitazar 150 μg or placebo. The trial was terminated after an interim analysis owing to serious adverse events that included statistically significant gastrointestinal hemorrhages and renal dysfunction in the aleglitazar arm.73 Most of these glitazar compounds had significant PPARγ action (than PPARα), which contributes to adverse events. Saroglitazar is the first glitazar class compound approved as a therapeutic agent. Saroglitazar has a different structure from glitazones, other glitazars, and fibrates. It is an aryl alkoxy propionic acid class molecule, which contains a unique pyrol moiety and lacks the glitazone ring. Saroglitazar was designed as a dual PPAR agonist having predominant PPARα effect with moderate PPARγ effect; thus, it provides antilipid effect with insulin sensitization, without being associated with typical glitazone side effects.74, 75 Although the data on the use of saroglitazar in NAFLD are still evolving, based on the results of various studies (PRESSV, VI), Drug Controller General of India has already approved use of saroglitazar for patients with diabetic dyslipidemia, uncontrolled by statins.76, 77, 78, 79

In NAFLD, saroglitazar has been shown to be more effective for reduction of the histological NAS than pioglitazone and fenofibrate in a mice model.80 C57BL/6 mice that were maintained on choline-deficient, L-amino acid–defined, high-fat diet for 8 weeks were treated with saroglitazar (3 mg/kg), fenofibrate (100 mg/kg), pioglitazone (30 mg/kg), or vehicle for 12 weeks. Saroglitazar reduced hepatic steatosis, inflammation, and ballooning and prevented development of fibrosis. It also reduced serum ALT and AST levels and expression of inflammatory and fibrosis biomarkers. Pioglitazone and fenofibrate did not show any improvement in steatosis but partially improved inflammation and liver function. In the same study, antifibrotic effect of saroglitazar (4 mg/kg) was also observed in the carbon tetrachloride–induced fibrosis model.80 Use of saroglitazar in patients with type 2 diabetes mellitus and dyslipidemia or those with type 2 diabetes mellitus and without dyslipidemia has been shown to improve serum transaminase levels.81, 82

A phase II study has also evaluated the improvement in serum ALT levels with saroglitazar in patients with biopsy-proven NASH (CTRI/2010/091/000108).83 Another phase II study in the USA is evaluating the efficacy of saroglitazar in improving serum liver enzymes and other serum biomarkers of inflammation and fibrosis with improvement in hepatic steatosis on MR imaging-estimated proton density fat fraction (MR-PDFF) in patients with NAFLD/NASH diagnosed on imaging or histology (NCT03061721).84 In addition, a recent phase III study in India is evaluating the histological efficacy of saroglitazar in comparison with placebo in patients with biopsy-proven NASH (CTRI/2015/10/006236).85 The results of these studies would clarify the role of saroglitazar in patients with NAFLD.

  • c)

    Pan-PPAR agonists

Several pan-PPAR agonists are in various phases of development, with many studies reported in animal models.26, 86, 87, 88, 89, 90 Bezafibrate has predominantly alpha action and has been shown to improve glycated hemoglobin in patients with diabetes mellitus.91, 92 Bezafibrate has also been shown to improve atherogenic dyslipidemia and insulin resistance without causing overweight.91, 92, 93 Although human studies are lacking, bezafibrate has been shown to improve NAFLD and diabetes in mice models.94, 95 Lanifibranor (IV1337) is another pan-PPAR agonist, which in addition to improving insulin sensitivity, has been shown to have antilipid, anti-inflammatory, and antifibrosis properties.96, 97, 98 In animal models, lanifibranor has been shown to improve insulin sensitivity and decrease hepatic steatosis, inflammation, ballooning, and fibrosis in liver tissue.96 An ongoing study is evaluating the efficacy of lanifibranor in patients with diabetes and NAFLD (ClinicalTrials.gov: NCT03459079).

PPAR agonists are attractive targets for the treatment of patients with NAFLD, given multiple actions of the PPAR on lipid metabolism, oxidation of FAs, glucose homeostasis, and inflammation. This becomes all the more important in absence of any recommended pharmacotherapy for these patients. Of all the PPAR agonists, the PPARγ agonist pioglitazone is the most extensively evaluated and has been found to be useful in patients with NAFLD but is limited by its side effect profile. Emerging data of dual PPAR agonists and pan-PPAR agonists appear encouraging and may hold promise for patients with NAFLD.

Conflicts of interest

The authors have none to declare.

References

  • 1.Ahmed W., Ziouzenkova O., Brown J. PPARs and their metabolic modulation: new mechanisms for transcriptional regulation? J Intern Med. 2007;262:184–198. doi: 10.1111/j.1365-2796.2007.01825.x. [DOI] [PubMed] [Google Scholar]
  • 2.Poulsen L., Siersbaek M., Mandrup S. PPARs: fatty acid sensors controlling metabolism. Semin Cell Dev Biol. 2012;23:631–639. doi: 10.1016/j.semcdb.2012.01.003. [DOI] [PubMed] [Google Scholar]
  • 3.Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications--a review. Nutr J. 2014 14;13:17. doi: 10.1186/1475-2891-13-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pawlak M., Lefebvre P., Staels B. Molecular mechanism of PPARalpha action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 2015;62:720–733. doi: 10.1016/j.jhep.2014.10.039. [DOI] [PubMed] [Google Scholar]
  • 5.Tailleux A., Wouters K., Staels B. Roles of PPARs in NAFLD: potential therapeutic targets. Biochim Biophys Acta. 2012;1821:809–818. doi: 10.1016/j.bbalip.2011.10.016. [DOI] [PubMed] [Google Scholar]
  • 6.Corton J.C. Evaluation of the role of peroxisome proliferator-activated receptor alpha (PPARalpha) in mouse liver tumor induction by trichloroethylene and metabolites. Crit Rev Toxicol. 2008;38:857–875. doi: 10.1080/10408440802209796. [DOI] [PubMed] [Google Scholar]
  • 7.Fisher C.D., Jackson J.P., Lickteig A.J., Augustine L.M., Cherrington N.J. Drug metabolizing enzyme induction pathways in experimental non-alcoholic steatohepatitis. Arch Toxicol. 2008;82:959–964. doi: 10.1007/s00204-008-0312-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Takeuchi S., Matsuda T., Kobayashi S., Takahashi T., Kojima H. In vitro screening of 200 pesticides for agonistic activity via mouse peroxisome proliferator-activated receptor (PPAR)alpha and PPARgamma and quantitative analysis of in vivo induction pathway. Toxicol Appl Pharmacol. 2006;217:235–244. doi: 10.1016/j.taap.2006.08.011. [DOI] [PubMed] [Google Scholar]
  • 9.Laughter A.R., Dunn C.S., Swanson C.L., Howroyd P., Cattley R.C., Corton J.C. Role of the peroxisome proliferator-activated receptor alpha (PPARalpha) in responses to trichloroethylene and metabolites, trichloroacetate and dichloroacetate in mouse liver. Toxicology. 2004;203:83–98. doi: 10.1016/j.tox.2004.06.014. [DOI] [PubMed] [Google Scholar]
  • 10.Evans R.M., Barish G.D., Wang Y.X. PPARs and the complex journey to obesity. Nat Med. 2004;10:355–361. doi: 10.1038/nm1025. [DOI] [PubMed] [Google Scholar]
  • 11.Desvergne B., Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev. 1999;20:649–688. doi: 10.1210/edrv.20.5.0380. [DOI] [PubMed] [Google Scholar]
  • 12.Issemann I., Green S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature. 1990;347:645–650. doi: 10.1038/347645a0. [DOI] [PubMed] [Google Scholar]
  • 13.Duseja A., Najmy S., Sachdev S. High prevalence of non-alcoholic fatty liver disease among healthy male blood donors of urban India. JGH Open. 2019;3:133–139. doi: 10.1002/jgh3.12117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Duseja A., Sharma B., Kumar A. Nonalcoholic fatty liver in a developing country is responsible for significant liver disease. Hepatology. 2010;52:2248–2249. doi: 10.1002/hep.23838. [DOI] [PubMed] [Google Scholar]
  • 15.Duseja A., Singh S.P., Saraswat V.A. Non-alcoholic fatty liver disease and metabolic syndrome-position paper of the Indian national association for the study of the liver, endocrine society of India, Indian college of cardiology and Indian society of gastroenterology. J. Clin. Exp. Hepatol. 2015;5:51–68. doi: 10.1016/j.jceh.2015.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Tyagi S., Gupta P., Saini A.S., Kaushal C., Sharma S. The peroxisome proliferator-activated receptor: a family of nuclear receptors role in various diseases. "J Adv Pharm Technol Research"" (JAPTR)". 2011;2:236–240. doi: 10.4103/2231-4040.90879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lefebvre P., Chinetti G., Fruchart J.C., Staels B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J Clin Investig. 2006;116:571–580. doi: 10.1172/JCI27989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Vanden Berghe W., Vermeulen L., Delerive P., De Bosscher K., Staels B., Haegeman G. A paradigm for gene regulation: inflammation, NF-kappaB and PPAR. Adv Exp Med Biol. 2003;544:181–196. doi: 10.1007/978-1-4419-9072-3_22. [DOI] [PubMed] [Google Scholar]
  • 19.Delerive P., Gervois P., Fruchart J.C., Staels B. Induction of IkappaBalpha expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferator-activated receptor-alpha activators. J Biol Chem. 2000;275:36703–36707. doi: 10.1074/jbc.M004045200. [DOI] [PubMed] [Google Scholar]
  • 20.Serviddio G., Giudetti A.M., Bellanti F. Oxidation of hepatic carnitine palmitoyl transferase-I (CPT-I) impairs fatty acid beta-oxidation in rats fed a methionine-choline deficient diet. PLoS One. 2011;6:e24084. doi: 10.1371/journal.pone.0024084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yu G.S., Lu Y.C., Gulick T. Co-regulation of tissue-specific alternative human carnitine palmitoyltransferase Ibeta gene promoters by fatty acid enzyme substrate. J Biol Chem. 1998;273:32901–32909. doi: 10.1074/jbc.273.49.32901. [DOI] [PubMed] [Google Scholar]
  • 22.Brandt J.M., Djouadi F., Kelly D.P. Fatty acids activate transcription of the muscle carnitine palmitoyltransferase I gene in cardiac myocytes via the peroxisome proliferator-activated receptor alpha. J Biol Chem. 1998;273:23786–23792. doi: 10.1074/jbc.273.37.23786. [DOI] [PubMed] [Google Scholar]
  • 23.Hashimoto T., Cook W.S., Qi C., Yeldandi A.V., Reddy J.K., Rao M.S. Defect in peroxisome proliferator-activated receptor alpha-inducible fatty acid oxidation determines the severity of hepatic steatosis in response to fasting. J Biol Chem. 2000;275:28918–28928. doi: 10.1074/jbc.M910350199. [DOI] [PubMed] [Google Scholar]
  • 24.Fraulob J.C., Souza-Mello V., Aguila M.B., Mandarim-de-Lacerda C.A. Beneficial effects of rosuvastatin on insulin resistance, adiposity, inflammatory markers and non-alcoholic fatty liver disease in mice fed on a high-fat diet. Clin Sci. 2012;123:259–270. doi: 10.1042/CS20110373. [DOI] [PubMed] [Google Scholar]
  • 25.Souza-Mello V., Gregorio B.M., Cardoso-de-Lemos F.S., de Carvalho L., Aguila M.B., Mandarim-de-Lacerda C.A. Comparative effects of telmisartan, sitagliptin and metformin alone or in combination on obesity, insulin resistance, and liver and pancreas remodelling in C57BL/6 mice fed on a very high-fat diet. Clin Sci (Lond) 2010;119:239–250. doi: 10.1042/CS20100061. [DOI] [PubMed] [Google Scholar]
  • 26.Stienstra R., Mandard S., Patsouris D., Maass C., Kersten S., Muller M. Peroxisome proliferator-activated receptor alpha protects against obesity-induced hepatic inflammation. Endocrinology. 2007;148:2753–2763. doi: 10.1210/en.2007-0014. [DOI] [PubMed] [Google Scholar]
  • 27.Patsouris D., Reddy J.K., Muller M., Kersten S. Peroxisome proliferator-activated receptor alpha mediates the effects of high-fat diet on hepatic gene expression. Endocrinology. 2006;147:1508–1516. doi: 10.1210/en.2005-1132. [DOI] [PubMed] [Google Scholar]
  • 28.Abdelmegeed M.A., Yoo S.H., Henderson L.E., Gonzalez F.J., Woodcroft K.J., Song B.J. PPARalpha expression protects male mice from high fat-induced nonalcoholic fatty liver. J Nutr. 2011;141:603–610. doi: 10.3945/jn.110.135210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tan C.K., Zhuang Y., Wahli W. Synthetic and natural Peroxisome Proliferator-Activated Receptor (PPAR) agonists as candidates for the therapy of the metabolic syndrome. Expert Opin Ther Targets. 2017;21:333–348. doi: 10.1080/14728222.2017.1280467. [DOI] [PubMed] [Google Scholar]
  • 30.Ip E., Farrell G.C., Robertson G., Hall P., Kirsch R., Leclercq I. Central role of PPARalpha-dependent hepatic lipid turnover in dietary steatohepatitis in mice. Hepatology. 2003;38:123–132. doi: 10.1053/jhep.2003.50307. [DOI] [PubMed] [Google Scholar]
  • 31.Fernandez-Miranda C., Perez-Carreras M., Colina F., Lopez-Alonso G., Vargas C., Solis-Herruzo J.A. A pilot trial of fenofibrate for the treatment of non-alcoholic fatty liver disease. Dig Liver Dis. 2008;40:200–205. doi: 10.1016/j.dld.2007.10.002. [DOI] [PubMed] [Google Scholar]
  • 32.Basaranoglu M., Acbay O., Sonsuz A. A controlled trial of gemfibrozil in the treatment of patients with nonalcoholic steatohepatitis. J Hepatol. 1999;31:384. doi: 10.1016/s0168-8278(99)80243-8. [DOI] [PubMed] [Google Scholar]
  • 33.El-Haggar S.M., Mostafa T.M. Comparative clinical study between the effect of fenofibrate alone and its combination with pentoxifylline on biochemical parameters and liver stiffness in patients with non-alcoholic fatty liver disease. Hepatol Int. 2015;9:471–479. doi: 10.1007/s12072-015-9633-1. [DOI] [PubMed] [Google Scholar]
  • 34.Laurin J., Lindor K.D., Crippin J.S. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology. 1996;23:1464–1467. doi: 10.1002/hep.510230624. [DOI] [PubMed] [Google Scholar]
  • 35.Ishibashi S., Yamashita S., Arai H. Effects of K-877, a novel selective PPARalpha modulator (SPPARMalpha), in dyslipidaemic patients: a randomized, double blind, active- and placebo-controlled, phase 2 trial. Atherosclerosis. 2016;249:36–43. doi: 10.1016/j.atherosclerosis.2016.02.029. [DOI] [PubMed] [Google Scholar]
  • 36.Araujo S., Soares E.S.A., Gomes F. 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. doi: 10.1016/j.ejphar.2016.06.043. [DOI] [PubMed] [Google Scholar]
  • 37.Gross B., Staels B. PPAR agonists: multimodal drugs for the treatment of type-2 diabetes. Best Pract Res Clin Endocrinol Metabol. 2007;21:687–710. doi: 10.1016/j.beem.2007.09.004. [DOI] [PubMed] [Google Scholar]
  • 38.Odegaard J.I., Ricardo-Gonzalez R.R., Goforth M.H. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007;447:1116–1120. doi: 10.1038/nature05894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tanaka T., Yamamoto J., Iwasaki S. Activation of peroxisome proliferator-activated receptor delta induces fatty acid beta-oxidation in skeletal muscle and attenuates metabolic syndrome. Proc Natl Acad Sci U S A. 2003;100:15924–15929. doi: 10.1073/pnas.0306981100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Riserus U., Sprecher D., Johnson T. Activation of peroxisome proliferator-activated receptor (PPAR) delta promotes reversal of multiple metabolic abnormalities, reduces oxidative stress, and increases fatty acid oxidation in moderately obese men. Diabetes. 2008;57:332–339. doi: 10.2337/db07-1318. [DOI] [PubMed] [Google Scholar]
  • 41.Peters J.M., Gonzalez F.J., Muller R. Establishing the role of PPARbeta/delta in carcinogenesis. Trends Endocrinol Metabol. 2015;26:595–607. doi: 10.1016/j.tem.2015.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lee M.Y., Choi R., Kim H.M. Peroxisome proliferator-activated receptor δ agonist attenuates hepatic steatosis by anti-inflammatory mechanism. Exp Mol Med. 2012;44:578–585. doi: 10.3858/emm.2012.44.10.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shan W., Palkar P.S., Murray I.A. Ligand activation of peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) attenuates carbon tetrachloride hepatotoxicity by downregulating proinflammatory gene expression. Toxicol Sci. 2008;105:418–428. doi: 10.1093/toxsci/kfn142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bays H.E., Schwartz S., Littlejohn T., 3rd 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. doi: 10.1210/jc.2011-1061. [DOI] [PubMed] [Google Scholar]
  • 45.Haczeyni F., Wang H., Barn V. The selective peroxisome proliferator-activated receptor-delta agonist seladelpar reverses nonalcoholic steatohepatitis pathology by abrogating lipotoxicity in diabetic obese mice. Hepatol Commun. 2017;1:663–674. doi: 10.1002/hep4.1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhu Y., Alvares K., Huang Q., Rao M.S., Reddy J.K. Cloning of a new member of the peroxisome proliferator-activated receptor gene family from mouse liver. J Biol Chem. 1993;268:26817–26820. [PubMed] [Google Scholar]
  • 47.Zhong X., Liu H. Honokiol attenuates diet-induced non-alcoholic steatohepatitis by regulating macrophage polarization through activating peroxisome proliferator-activated receptor gamma. J Gastroenterol Hepatol. 2018;33:524–532. doi: 10.1111/jgh.13853. [DOI] [PubMed] [Google Scholar]
  • 48.Yki-Jarvinen H. Thiazolidinediones. N Engl J Med. 2004;351:1106–1118. doi: 10.1056/NEJMra041001. [DOI] [PubMed] [Google Scholar]
  • 49.Nan Y.M., Fu N., Wu W.J. Rosiglitazone prevents nutritional fibrosis and steatohepatitis in mice. Scand J Gastroenterol. 2009;44:358–365. doi: 10.1080/00365520802530861. [DOI] [PubMed] [Google Scholar]
  • 50.Deng W., Meng Z., Sun A., Yang Z. Pioglitazone suppresses inflammation and fibrosis in nonalcoholic fatty liver disease by down-regulating PDGF and TIMP-2: evidence from in vitro study. Cancer Biomark. 2017;20:411–415. doi: 10.3233/CBM-170157. [DOI] [PubMed] [Google Scholar]
  • 51.Caldwell S.H., Hespenheide E.E., Redick J.A., Iezzoni J.C., Battle E.H., Sheppard B.L. A pilot study of a thiazolidinedione, troglitazone, in nonalcoholic steatohepatitis. Am J Gastroenterol. 2001;96:519–525. doi: 10.1111/j.1572-0241.2001.03553.x. [DOI] [PubMed] [Google Scholar]
  • 52.Masubuchi Y. Metabolic and non-metabolic factors determining troglitazone hepatotoxicity: a review. Drug Metab Pharmacokinet. 2006;21:347–356. doi: 10.2133/dmpk.21.347. [DOI] [PubMed] [Google Scholar]
  • 53.Ratziu V., Giral P., Jacqueminet S. 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. doi: 10.1053/j.gastro.2008.03.078. [DOI] [PubMed] [Google Scholar]
  • 54.Sanyal A.J., Chalasani N., Kowdley K.V. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 2010;362:1675–1685. doi: 10.1056/NEJMoa0907929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.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. doi: 10.1111/j.1365-2036.2011.04912.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Berlie H.D., Kalus J.S., Jaber L.A. Thiazolidinediones and the risk of edema: a meta-analysis. Diabetes Res Clin Pract. 2007;76:279–289. doi: 10.1016/j.diabres.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 57.Singh S., Loke Y.K., Furberg C.D. Thiazolidinediones and heart failure: a teleo-analysis. Diabetes Care. 2007;30:2148–2153. doi: 10.2337/dc07-0141. [DOI] [PubMed] [Google Scholar]
  • 58.Loke Y.K., Kwok C.S., Singh S. Comparative cardiovascular effects of thiazolidinediones: systematic review and meta-analysis of observational studies. BMJ. 2011;342:d1309. doi: 10.1136/bmj.d1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tang H., Shi W., Fu S. Pioglitazone and bladder cancer risk: a systematic review and meta-analysis. Cancer Med. 2018;7:1070–1080. doi: 10.1002/cam4.1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Neuschwander-Tetri B.A., Brunt E.M., Wehmeier K.R., Oliver D., Bacon B.R. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand rosiglitazone. Hepatology. 2003;38:1008–1017. doi: 10.1053/jhep.2003.50420. [DOI] [PubMed] [Google Scholar]
  • 61.Torres D.M., Jones F.J., Shaw J.C., Williams C.D., Ward J.A., Harrison S.A. Rosiglitazone versus rosiglitazone and metformin versus rosiglitazone and losartan in the treatment of nonalcoholic steatohepatitis in humans: a 12-month randomized, prospective, open- label trial. Hepatology. 2011;54:1631–1639. doi: 10.1002/hep.24558. [DOI] [PubMed] [Google Scholar]
  • 62.Belfort R., Harrison S.A., Brown K. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med. 2006;355:2297–2307. doi: 10.1056/NEJMoa060326. [DOI] [PubMed] [Google Scholar]
  • 63.Aithal G.P., Thomas J.A., Kaye P.V. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology. 2008;135:1176–1184. doi: 10.1053/j.gastro.2008.06.047. [DOI] [PubMed] [Google Scholar]
  • 64.Cusi K., Orsak B., Bril F. Long-term pioglitazone treatment for patients with nonalcoholic steatohepatitis and prediabetes or type 2 diabetes mellitus: a randomized trial. Ann Intern Med. 2016;165:305–315. doi: 10.7326/M15-1774. [DOI] [PubMed] [Google Scholar]
  • 65.Colca J.R., McDonald W.G., McCommis K.S., Finck B.N. Treating fatty liver disease by modulating mitochondrial pyruvate metabolism. Hepatol Commun. 2017;1:193–197. doi: 10.1002/hep4.1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.McCommis K.S., Finck B.N. Treating hepatic steatosis and fibrosis by modulating mitochondrial pyruvate metabolism. Cell Mol Gastroenterol Hepatol. 2019;7:275–284. doi: 10.1016/j.jcmgh.2018.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.McCommis K.S., Hodges W.T., Brunt E.M. Targeting the mitochondrial pyruvate carrier attenuates fibrosis in a mouse model of nonalcoholic steatohepatitis. Hepatology. 2017;65:1543–1556. doi: 10.1002/hep.29025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ratziu V., Harrison S.A., Francque S. 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. doi: 10.1053/j.gastro.2016.01.038. e5. [DOI] [PubMed] [Google Scholar]
  • 69.Staels B., Rubenstrunk A., Noel B. Hepatoprotective effects of the dual peroxisome proliferator-activated receptor alpha/delta agonist, GFT505, in rodent models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology. 2013;58:1941–1952. doi: 10.1002/hep.26461. [DOI] [PubMed] [Google Scholar]
  • 70.Hamren B., Ohman K.P., Svensson M.K., Karlsson M.O. 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. doi: 10.1177/0091270011416937. [DOI] [PubMed] [Google Scholar]
  • 71.Nissen S.E., Wolski K., Topol E.J. Effect of muraglitazar on death and major adverse cardiovascular events in patients with type 2 diabetes mellitus. J Am Med Assoc. 2005;294:2581–2586. doi: 10.1001/jama.294.20.joc50147. [DOI] [PubMed] [Google Scholar]
  • 72.Kendall D.M., Rubin C.J., Mohideen P. Improvement of glycemic control, triglycerides, and HDL cholesterol levels with muraglitazar, a dual (alpha/gamma) peroxisome proliferator-activated receptor activator, in patients with type 2 diabetes inadequately controlled with metformin monotherapy: a double-blind, randomized, pioglitazone-comparative study. Diabetes Care. 2006;29:1016–1023. doi: 10.2337/diacare.2951016. [DOI] [PubMed] [Google Scholar]
  • 73.Lincoff A.M., Tardif J.C., Schwartz G.G. Effect of aleglitazar on cardiovascular outcomes after acute coronary syndrome in patients with type 2 diabetes mellitus: the AleCardio randomized clinical trial. J Am Med Assoc. 2014;16:1515–1525. doi: 10.1001/jama.2014.3321. [DOI] [PubMed] [Google Scholar]
  • 74.Krishna Vaseem A., Sah R.K., Ali M. Saroglitazar: a novel dual acting peroxisome proliferator activated receptor (PPAR) in dyslipidemia associated with T2DM. Ejpmr. 2017;4:680–684. [Google Scholar]
  • 75.Joshi S.R. Saroglitazar for the treatment of dyslipidemia in diabetic patients. Expert Opin Pharmacother. 2015;16:597-606. doi: 10.1517/14656566.2015.1009894. [DOI] [PubMed] [Google Scholar]
  • 76.Agrawal R. The first approved agent in the Glitazar's Class: Saroglitazar. Curr Drug Targets. 2014;15:151–155. doi: 10.2174/13894501113149990199. [DOI] [PubMed] [Google Scholar]
  • 77.Jani R.H., Pai V., Jha P. A multicenter, prospective, randomized, double-blind study to evaluate the safety and efficacy of Saroglitazar 2 and 4 mg compared with placebo in type 2 diabetes mellitus patients having hypertriglyceridemia not controlled with atorvastatin therapy (PRESS VI) Diabetes Technol Ther. 2014;16:63–71. doi: 10.1089/dia.2013.0253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Pai V., Paneerselvam A., Mukhopadhyay S. A multicenter, prospective, randomized, double-blind study to evaluate the safety and efficacy of saroglitazar 2 and 4 mg compared to pioglitazone 45 mg in diabetic dyslipidemia (PRESS V) J Diabetes Sci Technol. 2014;8:132–141. doi: 10.1177/1932296813518680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Shetty S.R., Kumar S., Mathur R.P., Sharma K.H., Jaiswal A.D. Observational study to evaluate the safety and efficacy of saroglitazar in Indian diabetic dyslipidemia patients. Indian Heart J. 2015;67:23–26. doi: 10.1016/j.ihj.2015.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jain M.R., Giri S.R., Bhio B. Dual PPARα/γ agonist saroglitazar improves liver histopathology and biochemistry in experimental NASH models. Liver Int. 2018;38:1084–1094. doi: 10.1111/liv.13634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bhosle D., Bhosle V., Bobde J., Shaikh H., Kadam R. Study of saroglitazar in treatment of pre-diabetes with dyslipidemia: STOP-D. J Assoc Phys India. 2018;66:14–17. [PubMed] [Google Scholar]
  • 82.Chatterjee S., Majumder A., Ray S. Observational study of effects of saroglitazar on glycaemic and lipid parameters on Indian patients with type 2 diabetes. Sci Rep. 2015;5:7706. doi: 10.1038/srep07706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.ClinicalTrials.gov Identifier: CTRI/2010/091/000108, http://ctri.nic.in/Clinicaltrials/advsearch.php.
  • 84.ClinicalTrials.gov Identifier: NCT03061721 (https://clinicaltrials.gov/ct2/show/NCT03061721).
  • 85.CTRI/2015/10/006236,http://ctri.nic.in/Clinicaltrials/pdf_generate.php?trialid=11280&EncHid=&modid=&compid=%27,%2711280det%27.
  • 86.Penna-de-Carvalho A., Graus-Nunes F., Rabelo-Andrade J., Mandarim-de-Lacerda C.A., Souza-Mello V. Enhanced pan-peroxisome proliferator-activated receptor gene and protein expression in adipose tissue of diet-induced obese mice treated with telmisartan. Exp Physiol. 2014;99:1663–1678. doi: 10.1113/expphysiol.2014.081596. [DOI] [PubMed] [Google Scholar]
  • 87.Sadasivuni M.K., Reddy B.M., Singh J. CNX-013-B2, a unique pan tissue acting rexinoid, modulates several nuclear receptors and controls multiple risk factors of the metabolic syndrome without risk of hypertriglyceridemia, hepatomegaly and body weight gain in animal models. Diabetol Metab Syndrome. 2014;6:83. doi: 10.1186/1758-5996-6-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chen W., Fan S., Xie X., Xue N., Jin X., Wang L. Novel PPAR pan agonist, ZBH ameliorates hyperlipidemia and insulin resistance in high fat diet induced hyperlipidemic hamster. PLoS One. 2014;9:e96056. doi: 10.1371/journal.pone.0096056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Harrington W.W., S Britt C., G Wilson J. The effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan agonists on body weight, body mass, and serum lipid profiles in diet-induced obese AKR/J mice. PPAR Res. 2007;2007:97125. doi: 10.1155/2007/97125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Feng L., Luo H., Xu Z. Bavachinin, as a novel natural pan-PPAR agonist, exhibits unique synergistic effects with synthetic PPAR-gamma and PPAR-alpha agonists on carbohydrate and lipid metabolism in db/db and diet-induced obese mice. Diabetologia. 2016;59:1276–1286. doi: 10.1007/s00125-016-3912-9. [DOI] [PubMed] [Google Scholar]
  • 91.Tenenbaum A., Fisman E.Z. Balanced pan-PPAR activator bezafibrate in combination with statin: comprehensive lipids control and diabetes prevention? Cardiovasc Diabetol. 2012;11:140. doi: 10.1186/1475-2840-11-140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ogawa S., Takeuchi K., Sugimura K. Bezafibrate reduces blood glucose in type 2 diabetes mellitus. Metabolism. 2000;49:331–334. doi: 10.1016/s0026-0495(00)90176-8. [DOI] [PubMed] [Google Scholar]
  • 93.The BIP Study Group Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease: the Bezafibrate Infarction Prevention (BIP) study. Circulation. 2000;102:21–27. doi: 10.1161/01.cir.102.1.21. [DOI] [PubMed] [Google Scholar]
  • 94.Franko A., Neschen S., Rozman J. Bezafibrate ameliorates diabetes via reduced steatosis and improved hepatic insulin sensitivity in diabetic TallyHo mice. Mol Metab. 2017;6:256–266. doi: 10.1016/j.molmet.2016.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Sasaki Y., Shimada T., Iizuka S. Effects of bezafibrate in nonalcoholic steatohepatitis model mice with monosodium glutamate-induced metabolic syndrome. Eur J Pharmacol. 2011;662:1–8. doi: 10.1016/j.ejphar.2011.04.051. [DOI] [PubMed] [Google Scholar]
  • 96.Ruzehaji N., Frantz C., Ponsoye M. Pan PPAR agonist IVA337 is effective in prevention and treatment of experimental skin fibrosis. Ann Rheum Dis. 2016;75:2175–2183. doi: 10.1136/annrheumdis-2015-208029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Avouac J., Konstantinova I., Guignabert C. Pan-PPAR agonist IVA337 is effective in experimental lung fibrosis and pulmonary hypertension. Ann Rheum Dis. 2017;76:1931–1940. doi: 10.1136/annrheumdis-2016-210821. [DOI] [PubMed] [Google Scholar]
  • 98.Wettstein G., Luccarini J.M., Poekes L. The new-generation pan-peroxisome proliferator-activated receptor agonist IVA337 protects the liver from metabolic disorders and fibrosis. Hepatol Commun. 2017;1:524–537. doi: 10.1002/hep4.1057. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Clinical and Experimental Hepatology are provided here courtesy of Elsevier

RESOURCES