Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2020 Jul 8;52(4):619–636. doi: 10.1111/apt.15935

Review article: the impact of liver‐directed therapies on the atherogenic risk profile in non‐alcoholic steatohepatitis

Margery A Connelly 1,, Jonathan Velez Rivera 2, John R Guyton 2, Mohammad Shadab Siddiqui 3, Arun J Sanyal 3
PMCID: PMC7497003  PMID: 32638417

Summary

Background

Patients with non‐alcoholic fatty liver disease (NAFLD), the most common cause of chronic liver disease, are at higher risk of cardiovascular disease (CVD) and associated mortality. Therefore, it is important to understand how new therapies for non‐alcoholic steatohepatitis (NASH) may impact CVD risk factors in these patients.

Aims

To summarise the effects of drug therapies on lipid and lipoprotein levels in patients with NASH and provide insight into the potential mechanisms for the observed changes.

Methods

PubMed searches of the literature were performed and results were compiled.

Results

Recent clinical trials have highlighted the safety and efficacy of drug candidates for the treatment of NASH. Several agents have shown improvements in the histological features of NASH and liver function. Pioglitazone, a drug that is currently available for type 2 diabetes and may be useful for NASH, exhibits beneficial effects on lipids. However, agents such as farnesoid X receptor agonists, which are in development for NASH, may adversely affect circulating lipids and lipoproteins.

Conclusions

NASH is a multi‐system disease with a disproportionate CVD burden. Current and future drugs for NASH have had variable impact on the atherogenic risk profile. Potential co‐administration of a statin may help mitigate the negative impact of some of these therapies on lipid and lipoprotein levels.

1. INTRODUCTION

Nonalcoholic fatty liver disease (NAFLD) encompasses a histopathologic spectrum ranging from hepatic steatosis alone (fat content >5%), to steatosis accompanied by liver inflammation with or without fibrosis (nonalcoholic steatohepatitis or NASH), to cirrhosis of the liver. 1 NAFLD is a common cause of liver disease in developed countries, largely due to the increased prevalence of comorbidities such as obesity and type 2 diabetes (T2D). 1 , 2 Similar to obesity and T2D, patients with NAFLD have a higher risk of atherosclerotic cardiovascular disease (ASCVD, CVD), and CVD is the leading cause of death among patients with NAFLD. 2 , 3 , 4 While weight loss and exercise regimens have demonstrated short‐term success in treatment of hepatic steatosis and NASH, these are only achievable in <10% of patients. 5 Therefore, drug therapies for NASH have been proposed. Due to the higher risk of ASCVD in these patients, however, it is important to understand how new therapeutics, currently used or in development, may favourably or unfavourably impact CVD risk factors in NASH patients.

Recent published literature have demonstrated that many pharmaceutical agents including diuretics, anti‐diabetic agents, hormone therapies, steroids, anti‐psychotics, and anti‐inflammatory agents, elicit direct effects on lipoprotein metabolism thereby affecting circulating lipid and lipoprotein levels. 6 , 7 Many of these medications have favourable effects, however, there are a number of drug classes that adversely impact circulating lipids and lipoproteins—for example, some raise low‐density lipoprotein cholesterol (LDL‐C) levels, which are considered to be a risk marker and causative agent for ASCVD. There has been concern that these agents may thereby, augment ASCVD progression. While several publications have chronicled the many agents that are currently in development for NASH, 8 , 9 this review focuses on the effects of leading anti‐NASH drug therapies on lipids and lipoproteins in NASH patients and provides mechanistic insight on lipoprotein metabolism. Moreover evidence will be presented that these drug‐induced alterations may not necessarily lead to increased CVD risk in NASH patients, if current best practices in management of CVD risk are followed.

2. DYSLIPIDAEMIA IN PATIENTS WITH NASH AND/OR TYPE 2 DIABETES

2.1. What is atherogenic dyslipidaemia?

It is well known that a lipid profile with low high density lipoprotein cholesterol (HDL‐C) (<40 mg/dL) and high triglycerides (TG) (>200 mg/dL) is associated with increased risk of T2D, CVD and CVD‐related events. 10 , 11 These particular changes are referred to as diabetic or atherogenic dyslipidaemia and are most notable in patients with T2D, insulin resistance and/or NAFLD. 12 , 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 LDL particles contribute to CVD risk, and in clinical practice monitoring LDL‐C in the general population is sufficient for assessing a patient's CVD risk. 21 However, atherogenic dyslipidaemia is often associated with LDL‐C levels within the normal or near normal range making it hard to assess a patient's CVD risk using LDL‐C alone (Figure 1). 17 , 22 , 23 , 24 , 25 This occurs because the majority of the LDL particles in patients with atherogenic dyslipidaemia are small and dense and carry more TG and less cholesteryl esters and free cholesterol than large LDL particles (Figure 1). 17 , 22 , 26 Small‐dense LDL are highly atherogenic due to several factors including their propensity to traverse the endothelium and accumulate in atherosclerotic lesions as well as their longer plasma half‐lives accounting for accumulation of oxidation products. 22 , 27 , 28 , 29 , 30 , 31 , 32 , 33 Therefore, an increase in small‐dense LDL particles is indicative of metabolic dysregulation of lipoprotein metabolism and can contribute to the higher CVD risk observed in patients with T2D, hepatic steatosis, insulin resistance and/or NASH. 17 , 33 Consequently, having LDL‐C levels within the normal range, or somewhat raised, on a background of atherogenic dyslipidaemia, obesity, NAFLD and/or T2D, where the LDL particles are smaller, more numerous and highly atherogenic, is undesirable. Hence, measurement of total LDL and small‐dense LDL particle concentrations along with a standard lipid panel [total cholesterol (TC), TG, LDL‐C and HDL‐C] may provide a clearer picture of CVD risk in patients with metabolic disease. 22 , 31 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41

FIGURE 1.

FIGURE 1

Comparison of aspects of normal lipoprotein metabolism and atherogenic dyslipidaemia. HDL, high‐density lipoprotein particles; HDL‐C, high‐density lipoprotein cholesterol; IDL, intermediate density lipoprotein particles; LDL, low‐density lipoprotein particles; LDL‐C, low‐density lipoprotein cholesterol; TG, triglycerides; VLDL, very low density lipoprotein

2.2. Mechanism behind atherogenic dyslipidaemia

Insulin resistance is central to atherogenic dyslipidaemia and leads to an increase in fatty acid flux from adipose tissue to the liver and upregulation of hepatic synthesis of TG, free cholesterol and cholesteryl esters, which are secreted from the liver in large VLDL particles (Figure 1). 17 , 18 , 33 These large TG‐enriched VLDL are metabolised slowly in the periphery due to a reduction in lipoprotein lipase activity and contribute to the formation of TG‐enriched LDL, which in turn leads to generation of small‐dense LDL by hepatic lipase. 17 , 26 , 42 In contrast, metabolism of cholesteryl ester‐enriched small VLDL results in the formation of large‐buoyant LDL, both being associated with a less‐atherogenic lipoprotein profile. 17 Additionally, TG in VLDL particles can exchange with cholesteryl ester in LDL and HDL via the activity of cholesteryl ester transfer protein (CETP), culminating in altered metabolism and further generation of the atherogenic lipoprotein profile (Figure 1). 17 Overall, the changes in lipoprotein class and subclass levels (lipoprotein profile) that occur with insulin resistance and metabolic diseases such as NASH and T2D are: (a) increased particle concentrations for large VLDL and small LDL, (b) increased mean VLDL size, (c) decreased large HDL particles and (d) decrease in mean LDL and HDL size (Figure 1). 17 , 43

2.3. Reversal of dyslipidaemia

Some therapies for T2D, and potentially for NASH, lead to improvements in insulin sensitivity and to a reversal in the atherogenic lipoprotein phenotype. Because atherogenic dyslipidaemia results in an increase in small‐dense LDL and a decrease in larger more buoyant LDL particles that carry more cholesteryl ester and free cholesterol, the reversal of this lipoprotein phenotype may lead to increased LDL‐C, but not necessarily to an increase in the total number of LDL particles. The American Association of Clinical Endocrinologists is aware of this phenomenon and ha recommended the evaluation of drug treatments on circulating lipoprotein and lipid levels. 44 This evaluation would include a standard lipid panel along with apoB or a more in‐depth analysis using a comprehensive lipoprotein profile (total VLDL, LDL, and HDL particle concentrations and mean sizes as well as concentrations of the small, medium and large sub‐fractions). 45 Understanding whether alterations in the lipoprotein particle profile are due to direct (mechanism‐of‐action of the drug) or indirect (consequence of improving insulin resistance and reversing atherogenic dyslipidaemia) drug actions is key to understanding the drug's potential to elicit an increase or decrease in CVD risk with long‐term treatment.

3. LIPID AND LIPOPROTEIN EFFECTS OF POTENTIAL NASH THERAPIES

3.1. Diet and exercise

Because NAFLD is considered the hepatic manifestation of metabolic disease, the same risk factors for obesity and T2D, such as sedentary lifestyle, excess dietary fat and carbohydrates, including excess fructose and glucose consumption, hold true for NAFLD as well. 46 At present, there are no drug therapies that have been approved for the treatment of NAFLD or NASH, therefore lifestyle recommendations remain the standard of care. 47 , 48 , 49 Improvements in nutrition with or without weight loss can have a positive impact on fatty liver. 49 For example, a short‐term isocaloric diet that was low in carbohydrates and high in protein in subjects with NAFLD led to a dramatic reduction in liver fat content, decreased hepatic de novo lipogenesis, and increased fatty acid oxidation reflected by increased circulating β‐hydroxybutyrate concentrations. 50 In another study, a low carbohydrate high protein diet resulted in improvements in HbA1c and liver fat content even though the subjects did not lose weight. 51 Hypocaloric diets, especially those that elicit weight loss, have shown beneficial effects on hepatic and metabolic end points in subjects with NAFLD/NASH. 49 Hypocaloric very low carbohydrate diets in particular tend to increase ketone bodies (ketogenic diets) and decrease liver fat content, insulin resistance and features of metabolic syndrome. 52 In a recent study of 262 subjects who were placed on a diet that restricted carbohydrates and allowed for the consumption of protein and fat to satiety, subjects had reduced liver fat content and exhibited lipoprotein changes that were reminiscent of a reduction in insulin resistance and a reversal of atherogenic dyslipidaemia (Figure 1). 53 At 1 year, patients in the continuous care intervention group were in a state of nutritional ketosis and exhibited improvements in markers of CVD risk. 53 While they experienced an increase in LDL‐C, the increase appeared to be limited to the large LDL sub‐fraction. LDL size increased, accompanied by a decrease in small LDL, while total LDL particle concentration and apoB remained the same. Additionally, HDL‐C and apoA‐I levels increased, while blood pressure, TG and inflammation decreased. 53 These lipoprotein changes have been noted previously and are thought to be due to the reduction in carbohydrate and enhancement in fat utilisation leading to an increase in fatty acid oxidation. 53 , 54 , 55 , 56 Taken together, these studies suggest that ketogenic diets may be good for reducing the accumulation of liver fat potentially leading to a reduction in progression to NASH or liver cirrhosis. Caution needs to be taken, however, because human observational data has associated hepatic steatosis with high fat intake (as employed in some ketogenic diets), and rodent studies of high‐fat diets often show increased liver fat, insulin resistance and oxidative stress. 57 , 58

Combining weight loss with an exercise regimen is the most beneficial in terms of improving the histological features of NASH. 47 , 48 , 49 A systematic review and a recent meta‐analysis of a number of interventions suggested that weight loss of 5% in NAFLD or 7%‐10% in NASH was beneficial and could be achieved by a combination of a hypocaloric diet and 30‐60 minutes of moderate intensity exercise for 3‐5 days per week. 47 , 49 Combined diet and exercise interventions tended to show improvements in serum levels of liver enzymes as well as liver histology. 48 Dietary modifications such as reduction in carbohydrate intake as well as increased physical activity may lead to improvements in liver fat content, however, lifestyle changes including both diet and exercise are necessary to show improvement in hepatic inflammation and fibrosis. 47 , 48 , 49 Most physicians realize, however, that achieving these results in real‐life settings can be challenging. 47 , 49

With or without weight loss, the majority of studies have shown that lifestyle changes can be accompanied by alterations in CVD risk factors, such as circulating lipid levels and insulin resistance. 49 Low fat diets tend to reduce TC and LDL‐C levels while very low carbohydrate diets tend to increase LDL‐C, which can be a result of a potential increase in the percentage of fat in the diet and/or due to the reversal of insulin resistance. 49 , 53 It appears that regardless of the diet (low fat or low carbohydrate) or exercise regimen (high vs moderate intensity), reduced caloric intake accompanied by increased physical activity is likely to lead to weight loss as well as improvements in hepatic fat content, insulin resistance and serum lipid levels, all of which are thought to be anti‐atherogenic.

3.2. Statins

The safety of statins in patients with NAFLD/NASH has achieved increasing consensus. Initially regulatory agencies such as the US Food and Drug Administration (FDA) recommended monitoring hepatic transaminases, which are commonly elevated in NAFLD/NASH, and stopping statins for elevations ≥3 times the upper limit of normal (ULN). The 2006 National Lipid Association (NLA) Statin Safety Task Force Report suggested that baseline liver function tests should be obtained prior to starting statin therapy, but periodic monitoring was not recommended unless clinically indicated. 59 In 2012, the FDA decided to change statin labelling to reflect the 2006 NLA statement. The 2014 NLA Statin Safety Task Force Report commended this decision and encouraged the use of statins in NAFLD/NASH patients. 60 , 61 Furthermore, a recent study reported that statin therapy was safely tolerated in patients with decompensated cirrhosis, thereby, adding further support for its safety in patients with chronic and advanced liver disease. 62

Over the years, studies have shown that alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels may normalise on statin treatment, but randomised trials have not specifically addressed whether statins lead to improved biochemical and histological outcomes. Since patients with NAFLD/NASH are at higher risk of ASCVD, most experts assume that statins should improve ASCVD outcomes. Tikkanen et al conducted a post‐hoc subgroup analysis of a large CV outcomes trial (IDEAL) and found that treatment with atorvastatin 80 mg or simvastatin 20‐40 mg daily in patients with baseline normal vs moderately high ALT (>1 and <3 × ULN) was safe and resulted in similar lipoprotein changes irrespective of ALT elevation. 63 Moreover patients with high ALT had significantly better prevention of major coronary events. The GREACE study aimed at demonstrating ASCVD risk reduction among 1600 participants randomized to receive statin therapy in a university clinic vs usual care elsewhere. A post‐hoc subgroup analysis among 437 patients with moderately abnormal liver function tests, mostly attributable to NAFLD, showed that 227 receiving statins had favourable lipoprotein changes as well as 35% mean reduction in ALT and 47% reduction in AST. 64 Interestingly, 89% of participants in the statin group with abnormal liver tests at baseline had ALT and AST concentrations within the normal range by the end of the study. 64 No patient had more than minor increases in bilirubin or alkaline phosphatase. More importantly, patients treated with a statin had a significantly reduced risk of ASCVD events. Similar outcomes in ultrasound features, ALT and AST changes, and lipoprotein changes were seen in other post‐hoc analyses (IDEAL and ATTEMPT). 65 , 66

Besides studies reporting changes in liver enzymes with statin treatment, others have looked at potential changes in liver fat content and liver histology. For example, Athyros et. al. assessed changes in liver fat content by ultrasound in patients with metabolic syndrome, LDL‐C >130 mg/dL, hepatic steatosis and elevated AST and/or ALT. 67 The authors showed that intensive lifestyle modifications, in addition to treatment of hypertension, impaired fasting glucose, obesity and dyslipidaemia, led to reduced liver fat when assessed via ultrasound. In fact, 67% of the participants taking atorvastatin no longer had evidence of fatty liver. 67 A recent Cochrane review demonstrated no significant effects of statins on ALT, AST, or liver fat by ultrasonography and showed mixed outcomes with regard to histological improvement in NASH parameters on liver biopsy. 68 Therefore, the therapeutic benefits of statins in NAFLD and NASH patients, besides the known reductions in LDL‐C, remain elusive. 68 However, despite the cardiovascular benefit of statin therapy, in patients with NAFLD or suspected NAFLD, statin therapy is often stopped. 69

In summary, evidence suggests that statins are safe and effective for LDL lowering in patients with NAFLD/NASH, at least when baseline transaminase elevations are less than 3 times ULN. They should be considered in patients meeting criteria per the ASCVD risk calculation. However, improvements in overall NAFLD/NASH parameters and hepatic steatosis by ultrasound have been inconsistent. Further, we lack evidence to suggest improvements in outcomes such as biopsy‐proven histological and fibrotic scores with statin treatment. 65 , 68

3.3. Vitamin E

Prior to 2000, the recognition of vitamin E's antioxidant role and new evidence for lipoprotein oxidation as a proatherogenic mechanism generated enthusiasm for vitamin E as a natural antiatherogenic strategy. This led to large cardiovascular outcome trials, which demonstrated neither beneficial nor harmful effects of vitamin E in doses averaging 300‐400 IU per day on cardiovascular events. 70 , 71 Likewise, in the Pioglitazone vs Vitamin E vs Placebo for the Treatment of Nondiabetic Patients with Nonalcoholic Steatohepatitis (PIVENS) trial, only treatment with vitamin E (800 IU daily) met the primary therapeutic endpoint. 72 Based on these results, guidelines suggest the use of vitamin E in nondiabetic subjects with biopsy‐proven NASH. 73 Besides PIVENS, a number of clinical studies have explored the use of vitamin E in adult or paediatric subjects with NAFLD, NASH and/or T2D. 74 , 75 , 76 Many of these studies reported reductions in ALT and/or improvements in hepatic steatosis or NASH (alone or in combination therapy) which have been confirmed in a meta‐analysis. 75 , 76 , 77 To date, however, no significant changes in lipids or lipoprotein levels have been noted with vitamin E treatment (Table 1).

TABLE 1.

Drugs on the market or in clinical development for NASH and the reported effects on lipids and the lipoprotein profile in patients with NASH

Name of medication or drug candidate Drug class/mechanism Significant changes in lipids and lipoproteins Potential mitigation Shown to have CV benefit or no harm References
Vitamin E Nutritional Supp. No significant changes in lipids or lipoproteins observed. 72, 74, 75, 76
Pioglitazone PPARγ agonist ↓TG, ↑HDL‐C, ↑LDL‐C, ↑large LDL‐P, ↓small LDL‐P Statin treatment Yes 72, 83, 84, 85, 86, 87, 88, 89
Elafibranor PPARα/δ agonist Potential changes have not been reported to date. 94
Seladelpar (MBX‐8025) PPARδ agonist ↓TG, ↑HDL‐C, ↓LDL‐C, ↓small LDL‐P Statin treatment 95, 96
Saroglitazar PPARα/γ agonist ↓TG, ↓TC, ↑HDL‐C, ↓nonHDL‐C 97, 98
Liraglutide, exenatide GLP‐1 agonists When given alone, no significant changes observed. Yes 99, 100, 101, 102, 103, 104, 105, 106
Canagliflozin SGLT2 inhibitor ↓TG, ↑HDL‐C, ↑LDL‐C Statin treatment Yes 107, 112, 117, 118, 119
Dapagliflozin SGLT2 inhibitor ↓TG, ↑HDL‐C, ↑LDL‐C, ↓small LDL‐P, ↑large LDL‐P, ↑large HDL‐P Statin treatment Yes 109, 110, 113, 116
Empagliflozin SGLT2 inhibitor ↔TG, ↑HDL‐C, ↑LDL‐C Statin treatment Yes 108, 111, 115, 120, 121, 122
Obeticholic acid (OCA) FXR agonist ↑TC, ↑LDL‐C, ↓HDL‐C, ↓large VLDL‐P, ↑small VLDL‐P, ↑large and small LDL‐P, ↓total, large and medium HDL‐P Statin treatment 128, 129, 130, 131, 132, 133, 134, 135, 136, 137
NGM282 FGF19 analogue ↑TC, ↑LDL‐C, ↑HDL‐C, ↓large VLDL‐P, ↑total and large LDL‐P, ↓small HDL‐P Statin treatment 138, 139, 140, 141
Pegbelfermin FGF21 analogue ↓TG, ↑HDL‐C 142, 143
GS‐0976 ACC1/2 inhibitor ↑TG, ↑VLDL‐P Fish oil 144, 145, 146
MK‐4702 ACC1/2 inhibitor ↑TG, ↑VLDL‐P 147
Cenicriviroc CCR2/5 antagonist Has not yet been studied or reported. 149, 150
Selonsertib (GS‐4997) ASK1 inhibitor Has not yet been studied or reported. 151, 152
Emricasan (IDN‐6556) Caspase inhibitor Has not yet been studied or reported. 153, 154, 155, 156
Resmetirom (MGL‐3196) THRβ agonist ↓TG, trend ↓LDL‐C 157, 158, 159
Aramchol SCD1 inhibitor Has not yet been studied or reported. 160
MSDC‐0602K MPC activator Has not yet been studied or reported. 161, 162, 163

Abbreviations: ACC1/2, acetyl‐CoA carboxylase 1 and 2; ASK1, apoptosis signal‐regulating kinase 1; CCR2/5, chemokine receptor type 2/5; CV, cardiovascular; FGF, fibroblast growth factor; FXR, farnesoid X receptor; GLP‐1, glucagon‐like peptide 1; HDL‐C, high‐density lipoprotein cholesterol; HDL‐P, high‐density lipoprotein particles; LDL‐C, low density lipoprotein cholesterol; LDL‐P, low density lipoprotein particles; MPC, mitochondrial pyruvate carrier; NASH, non‐alcoholic steatohepatitis; PPAR, peroxisome proliferator‐activated receptor; SCD1, stearoyl‐CoA desaturase‐1; SGLT2, sodium glucose co‐transporter 2; Supp., supplement; T2D, type 2 diabetes; TC, total cholesterol; TG, triglycerides; THRβ, thyroid hormone receptor β; VLDL‐P, very large density lipoprotein particles.

The story with vitamin E treatment in patients with NASH and/or T2D, however, is not finished. Recently it was shown that the haptoglobin 2 allele is associated with a histologic response to vitamin E in subjects with NASH, suggesting that genotyping of the haptoglobin allele may be a way of identifying patients who are more likely to respond to vitamin E treatment. 78 Moreover the haptoglobin 2 allele was shown to be associated with a reduction in CVD in subjects with T2D treated with vitamin E. 79 This was shown in the Heart Outcome Prevention Evaluation (HOPE) study, the Women's Health Study and the Israel Cardiovascular Vitamin E (ICARE) study. 79 The authors suggest that the protection afforded by the haptoglobin 2 allele may be related to oxidative stress and improvements in HDL functionality. 79 Additional large prospective studies are needed to confirm these observations, however, it is intriguing to think that there may be a way to identify NASH and/or T2D patients who would realize both hepatic and CVD benefits with vitamin E treatment.

3.4. Peroxisome proliferator‐activated receptor agonists

Peroxisome proliferator‐activated receptors (PPARs) are a group of nuclear receptors that are expressed in liver, adipose tissue, heart, skeletal muscle and kidney and are responsible for the transcriptional regulation of processes such as fatty acid oxidation, lipid transport and gluconeogenesis. 80 There are three PPARs (α, β/δ and γ) that bind the same target DNA sequence but differ in tissue distribution. A few PPAR agonists are on the market for treatment of T2D (pioglitazone) and hypertriglyceridaemia (gemfibrozil, fenofibrate). 81 PPARα agonists or fibrates reduce TG but have not been shown to provide beneficial effects on liver histological endpoints in patients with NAFLD. 82 Several PPAR single or dual receptor agonists have been evaluated or are currently being tested in clinical trials in NAFLD/NASH patients.

Pioglitazone is a PPARγ agonist that has been shown to be efficacious in dyslipidaemia and T2D. 81 In the PIVENS trial, while it did not meet the primary liver endpoint, pioglitazone treatment did lead to a significant reduction in ALT, AST, hepatic steatosis, and lobular inflammation in NASH patients. 72 A meta‐analysis of four clinical studies confirmed these results. 83 In a longer term study in patients with biopsy‐proven NASH and either prediabetes or T2D, pioglitazone treatment, compared to placebo, led to improvements in steatosis, inflammation and ballooning with 58% of the patients achieving the primary outcome after 18 months. 84 Moreover there was a reduction in the mean fibrosis score but not the number of patients with improvement in fibrosis stage ≥1 in the pioglitazone treated group compared to the placebo treated group. 84

Analysis of lipoprotein sub‐fraction concentrations in the PIVENS trial revealed that NASH resolution was associated with favourable changes in a subset of lipoproteins, suggesting a reduction in the atherogenic profile, which may translate to a reduction in CVD risk (Table 1). 85 These positive effects of pioglitazone on the lipoprotein profile had been previously noted. 86 , 87 While small LDL particles were reduced, pioglitazone treatment elicited an increase in large LDL particles and a small but significant increase in LDL‐C levels (as the label for pioglitazone reflects). 86 , 87 These lipid changes may be related to the drug's ability to reduce insulin resistance and reverse atherogenic dyslipidaemia (Figure 1). Regardless of the mechanism for the lipoprotein changes observed with pioglitazone treatment, a prospective 3‐year study, called the PROspective pioglitAzone Clinical Trial In macroVascular Events (PROactive), revealed that pioglitazone reduced the composite of all‐cause mortality, non‐fatal myocardial infarction and stroke in T2D patients at high CVD risk. 88 Although there was no statistically significant difference between pioglitazone and placebo treatment in the incidence of a first event in the composite end‐point, meaning that pioglitazone did not have a mortality or CVD risk benefit, there was no increase in mortality or in total CVD events. 88 A 10‐year follow up confirmed that there were no increases in all‐cause mortality or CVD events between pioglitazone and placebo treatment. 89 It should be noted, however, that fluid retention and weight gain are common side effects of pioglitazone which may limit its use. In addition, pioglitazone is not widely recommended for routine T2D treatment because of concerns about reduced bone density and questionable risk of bladder cancer. Nevertheless, the overall results illustrate that a small increase in LDL‐C accompanied by favourable effects on TG and HDL‐C may not lead to an increase in CVD events, at least in patients with T2D. This remains to be investigated in patients with NASH. Of course, due to the pleiotropic effects of therapeutic agents, each drug that raises LDL‐C, or has what appears to be a non‐beneficial effect on CVD risk factors, needs to be studied mechanistically so that a full picture of the relative benefit‐risk profile can be understood and monitored over time.

The PPARγ agonist rosiglitazone that was approved for the treatment of T2D has been shown to increase LDL‐C, nonHDL‐C and triglyceride levels. 86 As a result of a meta‐analysis of forty‐two clinical studies showing untoward effects of rosiglitazone on CVD outcomes and plausible concerns over a potential increase in ASCVD, rosiglitazone was removed from the market. 90 After multiple studies revealed that there was no clear increase in CVD events with rosiglitazone treatment in T2D patients, the FDA allowed it to be marketed with a black box warning on the label. 91 , 92 , 93 This warning includes language stating that while the meta‐analysis showed rosiglitazone was associated with an increased risk of myocardial ischemic events, three large, long‐term, prospective, randomised, controlled clinical trials have not confirmed or excluded this risk. It is because of lingering concerns over the relationship between LDL‐C, oedema and the potential for increased CVD events, however, that rosiglitazone is not recommended in guidelines for use in patients with T2D, nor has it been tested in NASH trials.

Elafibranor is a dual PPARα/δ agonist that showed efficacy in treatment of NASH in the Phase 2b GOLDEN505 trial in patients with NASH without cirrhosis. 94 Although it did not meet the primary endpoint, in the post‐hoc analysis elafibranor resolved NASH in a larger proportion of patients with NAFLD activity score ≥4, than placebo. In addition, elafibranor treatment led to improvements in insulin sensitivity, glucose homeostasis, and lipid metabolism and reduced liver enzymes and systemic inflammation. 94 However, results from the Phase 3 RESOLVE‐IT trial (NCT02704403) were recently revealed and the data showed that elafibranor treatment did not meet its primary endpoint of NASH resolution without worsening of fibrosis. The response rate was 19.2% for patients who received elafibranor 120 mg compared to 14.7% for patients in the placebo arm. In addition, the other key secondary endpoint related to metabolic parameters did not achieve statistical significance. To date, no comprehensive lipoprotein profile results have been published with elafibranor (Table 1).

In an early study in patients with obesity and atherogenic dyslipidaemia, seladelpar (MBX‐8025), a selective PPARδ agonist, was tested with and without atorvastatin. 95 Results revealed that treatment with seladelpar led to a reduction in apoB, LDL‐C, TG, non‐HDL‐C and high sensitivity C‐reactive protein (hsCRP), and an increase in HDL‐C. 95 After seladelpar treatment, there was a lower percentage of study participants with a preponderance of small LDL particles. These results were extended by analysing concentrations of lipoprotein. 96 In short, seladelpar treatment led to a decrease in the atherogenic lipoprotein phenotype and seladelpar combined with atorvastatin led to a further reduction across all apoB‐containing lipoproteins (Table 1). These effects may have been due to the reduced waist circumference and improved insulin sensitivity that were observed in these subjects. Unfortunately, the Phase 2b proof‐of‐concept study in patients with biopsy‐proven NASH (NCT03551522) was placed on hold after 52 weeks of treatment due to unexpected histological changes suggestive of immune mediated injury.

Lipaglyn (saroglitazar) is a PPARα/γ agonist used in India for the treatment of dyslipidaemia in T2D patients. The results of EVIDENCES IV, a Phase 2 trial in 106 patients with NAFLD and/or NASH, revealed that saroglitazar treatment led to an improvement in ALT, liver fat content, insulin resistance and dyslipidemia. 97 Previously it was shown that saroglitazar treatment reduced TG, TC, non‐HDL‐C, LDL‐C and HbA1c as well as increased HDL‐C in patients with NAFLD and diabetic dyslipidaemia. 98 However, overall safety of saroglitazar has yet to be studied in a large randomised controlled trial. IVA337 is a pan PPAR agonist that is in a Phase 2 trial in patients with NAFLD and/or NASH (NCT03008070). It will be interesting to see if or how the lipoprotein profiles differ in NAFLD/NASH patients treated with these varied PPAR agonists.

3.5. Glucagon‐like peptide 1 agonists

Glucagon‐like peptide 1 (GLP‐1) is an incretin hormone that is secreted from the gut in response to food intake. GLP‐1 peptidomimetic receptor agonists, or GLP‐1 peptides that have been modified to increase half‐life, have been shown to improve glucose control in T2D patients via several mechanisms including enhanced insulin secretion, delayed gastric emptying, preserved β‐cell function, and inhibition of glucagon secretion. 99 Clinically, treatment with GLP‐1 agonists results in weight loss and improvement systolic blood pressure and HbA1c. Given the improvements in multiple CVD risk factors, it is not surprising that GLP‐1 receptor agonists have shown beneficial effects, or at least no harm, in CVD outcomes trials to date. 100 , 101 , 102 For example, results of CVD outcome trials in T2D patients with increased CVD risk demonstrated a significant reduction in time to first major adverse cardiovascular event (MACE) with liraglutide (LEADER), semaglutide (SUSTAIN‐6) and dulaglutide (REWIND), while a CVD outcome trial examining lixisenatide (ELIXA) demonstrated no difference in CVD events. 100 , 101 , 102 While GLP‐1 receptor agonists have been shown to elicit weight loss, few studies have reported a significant improvement in insulin sensitivity. 103 In addition, when given alone (eg without a concomitant diet regimen) GLP‐1 receptor agonists did not lead to the types of changes in lipids and lipoprotein parameters that would suggest an improvement or reversal of atherogenic dyslipidaemia (Table 1). 103 , 104 , 105 Therefore, the mechanism for the CVD benefits with GLP‐1 receptor agonism do not seem related to modifications in lipids, but may be related to amelioration of other CVD risk factors such as reducing blood pressure and weight. 103

In LEAN, a Phase 2 study in patients with biopsy proven NASH, a larger percentage of subjects treated with liraglutide exhibited resolution of NASH compared to placebo. 106 Larger clinical studies testing liraglutide (NCT02654665), semaglutide (NCT03884075, NCT03987451, NCT02970942, NCT03987074) or dulaglutide (NCT03648554, NCT03590626), in patients with NAFLD or NASH are currently in progress. These studies should reveal whether the same beneficial effects observed on blood pressure and CVD outcomes in T2D patients are recapitulated in larger studies in NASH patients.

3.6. Sodium glucose co‐transporter 2 inhibitors

Several sodium glucose co‐transporter 2 (SGLT2) inhibitors have been approved for treatment of T2D (canagliflozin, dapagliflozin, empagliflozin, ertugliflozin). SGLT2 inhibitors lower plasma glucose by inhibiting renal reabsorption of glucose thereby increasing urinary glucose excretion. SGLT2 inhibitors, such as canagliflozin, have been shown to enhance insulin sensitivity, probably due to a reduction in body weight, and also to reduce HbA1c, systolic and diastolic blood pressure, and liver enzymes. They also cause a decrease in circulating TG and small but statistically significant 2%‐5% increases in LDL‐C and HDL‐C. 107 , 108 Dapagliflozin was also shown to decrease small‐dense LDL‐C and increase large‐buoyant LDL and HDL2 cholesterol, suggesting that the lipoprotein changes observed with SGLT2 inhibition may be related to decreased insulin resistance and a reversal of atherogenic dyslipidaemia (Table 1). 109 , 110 Furthermore, the pattern of lipoprotein changes observed in clinical trials with various SGLT2 inhibitors is reminiscent of metabolic switching from glucose utilisation as a fuel source (glucose oxidation) to use of fat as a fuel source (fatty acid oxidation), which occurs largely in liver and muscle tissue (Figure 2). This is reflected by the increases in ketone bodies that have been observed in T2D subjects treated with SGLT2 inhibitors. These changes are thought to be due to a reduction in carbohydrate and enhancement in fat utilisation which then leads to an increase in fatty acid oxidation, similar to the effects of a ketogenic diet. In the case of SGLT2 inhibition, the increased urinary glucose excretion forces a switch in energy utilisation from glucose to fatty acids. The liver responds by secreting more VLDL‐TG, which are catabolized by muscle to produce energy (lipoprotein lipase‐mediated lipolysis and fatty acid oxidation), creating cholesteryl ester‐enriched LDL and HDL particles (increased LDL‐C and HDL‐C). In addition, the reduced body weight is consistent with the loss of calories due to increased urinary glucose excretion and fatty acid oxidation in adipose tissue. Therefore, one possible interpretation of the clinical data with SGLT2 inhibition is that the overall metabolic changes observed (favourable changes in blood pressure, HDL‐C, TG and weight, but unfavourable changes in LDL‐C) are partly the result of a switch in fuel source from glucose to fatty acids and a reversal of the atherogenic dyslipidaemia (Figure 2).

FIGURE 2.

FIGURE 2

Pleiotropic effects of sodium glucose co‐transporter 2 (SGLT2) inhibition. HDL‐C, high‐density lipoprotein cholesterol; LDL‐C, low‐density lipoprotein cholesterol; NASH, non‐alcoholic steatohepatitis; TG, triglycerides; VLDL, very low density lipoprotein

Despite the increases in LDL‐C that have been observed with SGLT2 inhibitors and are reflected in their product labels, this class of therapeutic agents has shown signs of providing benefits with respect to CVD outcomes (Table 1). 100 , 111 , 112 The results of the EMPA‐REG OUTCOME trial revealed that in patients with T2D and high CVD risk empagliflozin treatment led to a significantly lower rate of death from cardiovascular causes compared to placebo as well as a reduction in hospitalisation for heart failure and all‐cause mortality. 111 The CANVAS trials revealed that canagliflozin treatment in patients with T2D and high CVD risk led to a lower risk of CVD events. 112 In addition, the DECLARE‐TIMI trial reported that dapagliflozin treatment in a similar population of patients led to a lower rate of CVD death or hospitalisation for heart failure. 113 Subsequently, multiple meta‐analyses, post‐registration investigations and independent clinical studies have corroborated these findings. 100 , 114 While the exact mechanisms for the improvements in CVD outcomes have not yet been fully delineated, it appears that the pleiotropic effects of SGLT2 inhibition may work in concert to elicit both CVD and renal protective effects (Figure 2). 114 , 115 It is therefore clear that one must examine the spectrum of changes in CVD risk factors (positive or negative) before hypothesising whether long‐term treatment with a particular drug class might result in benefit or harm with respect to CVD outcomes (Figure 2).

SGLT2 inhibitors have shown benefits for reduction of hepatic steatosis and liver enzymes and show promise for treatment of NASH. 116 Treatment with canagliflozin for 24 weeks in a small number of patients with NASH and T2D resulted in improvements in the histological features of NASH. 117 In addition, canagliflozin treatment led to a reduction in hepatic steatosis and an improvement in insulin sensitivity and insulin secretion in patients with T2D and NAFLD. 118 , 119 Analysis of multiple trials with empagliflozin showed improvements in liver enzymes that might be a reflection of weight loss and reduced liver fat. 120 , 121 Additionally, empagliflozin elicited improvements in steatosis, ballooning and fibrosis in a small pilot study of subjects with NASH. 122 Dapagliflozin, luseogliflozin and ipragliflozin treatment showed similar benefits in patients with T2D or NAFLD/NASH. 116 It will be essential, however, to see if these effects are recapitulated by SGLT2 inhibition in large Phase 3 trials in patients with NASH, especially since these patients would benefit from the improvements in renal function and CVD outcomes.

3.7. Farnesoid X receptor agonists and fibroblast growth factor 19 peptidomimetics

Farnesoid X receptor (FXR) is an intracellular nuclear receptor that binds bile acids and elicits transcriptional signalling leading to the expression of a number of target genes relevant for bile acid, cholesterol, lipid and carbohydrate metabolism as well as inflammation, fibrosis and carcinogenesis. 123 Specifically, FXR agonists decrease bile acid synthesis, bile acid uptake, lipogenesis, gluconeogenesis and liver inflammation, and increase hepatic bile acid export. 123 , 124 Many of these effects are mediated by FGF19, a peptide hormone synthesised by gut epithelial cells and hepatocytes in response to FXR signalling. FXR activation reduces VLDL levels and circulating TG via repression of hepatic sterol responsive element binding protein 1c (SREBP1c), microsomal triglyceride transfer protein, and apoB gene expression. 124 Moreover FXR amplifies lipoprotein lipase activity by increasing apoCII (activator of lipoprotein lipase) and reducing apoCIII expression (inhibitor of lipoprotein lipase), thereby promoting peripheral VLDL‐TG lipolysis. 124 , 125 , 126 Based on these effects, FXR agonism would appear to be an attractive therapeutic target for the treatment of liver disease. 127 However, since FXR inhibits the synthesis of bile acids from cholesterol, thereby increasing an active pool of hepatic cholesterol, FXR agonism may lead to reduced LDL receptor cell surface expression. A reduction in LDL receptors, coupled with increased lipoprotein lipase activity, may result in reduced LDL clearance and elevated concentrations of circulating LDL particles and LDL‐C (Figure 3). Additionally, FXR agonism has been shown to increase HDL‐C clearance by increasing scavenger receptor‐B1 (SR‐B1) and to decrease HDL production by reducing apoA‐I. 128 , 129 , 130 , 131 FXR agonists in development for treatment of NASH have exhibited both the positive and negative aspects of FXR agonism, and clinical studies have been undertaken to understand the effects on disease activity as well as CVD risk.

FIGURE 3.

FIGURE 3

Pleiotropic effects of farnesoid X receptor (FXR) agonism. C4, 7‐alpha‐hydroxy‐4‐cholesten‐3‐one; FGF, fibroblast growth factor; LDL, low‐density lipoprotein particles; LDL‐C, low‐density lipoprotein cholesterol; NASH, non‐alcoholic steatohepatitis; TC, total cholesterol; TG, triglycerides; VLDL, very low density lipoprotein

Obeticholic acid (OCA) is a potent FXR agonist that was recently shown to improve disease‐related factors in patients with NASH. In a small proof‐of‐concept study, OCA treatment for 6 weeks led to an increase in serum FGF19 and insulin sensitivity, and a decrease in bile acids, the bile acid precursor C4, ALT and γ‐glutamyl transferase (GGT). 132 In the Farnesoid X Receptor Ligand Obeticholic Acid in NASH Treatment (FLINT) trial, OCA elicited improvements in the histological features of NASH. In both studies, OCA vs placebo treatment led to 16%‐24% increases in LDL‐C. 132 , 133 To gain a better understanding of OCA's effects, nuclear magnetic resonance analysis of lipoprotein particle concentrations was obtained before and after OCA treatment. 134 OCA treatment elicited increases in large‐buoyant and small‐dense LDL particles. The increases trended higher at 12 weeks than 72 weeks, which may be attributed to increased use of statins in patients whose LDL‐C levels were increased by OCA, and tended to be much higher for large than small LDL. OCA treatment elicited a reduction in large VLDL and an increase in small VLDL without any change in total VLDL concentration. 134 Thus, the net effect of FXR agonism is a shift to what some lipidologists believe is a less atherogenic lipoprotein profile. OCA vs placebo treatment also led to 2.6%‐4.5% reductions in HDL‐C. Total HDL particles were similarly reduced including both large and medium HDL particle concentrations, most likely due to increased clearance. 128 , 129 , 130 , 131 This is likely due to an increase in scavenger receptor B1 (SR‐B1) which would lead to an increase in the reverse cholesterol transport pathway and would be anti‐atherogenic. However, kinetic studies are warranted to fully understand the mechanism. All of the lipoprotein changes reverted to baseline 12 weeks after OCA discontinuation. 134 A study in healthy volunteers also revealed an increase in LDL‐C, confirming OCA's direct effects on LDL‐C, presumably via increased intrahepatic cholesterol content and decreased LDL receptor cell surface expression (Figure 3). 135 In this study, HDL particles decreased largely due to a reduction in medium and small HDL, and total LDL particles increased along with an increase in large LDL and LDL‐C. 135 In order to evaluate whether OCA has long‐term detrimental effects on CVD, it would be important to determine if concurrent statin treatment can reduce LDL‐C in OCA‐treated subjects. To this end, the recently conducted CONTROL Phase 2 study investigated the effects of atorvastatin treatment on OCA‐induced changes in LDL‐C in patients with NASH. 136 Results revealed that OCA increased mean LDL‐C, mostly attributable to an increase in large LDL. 136 More importantly, the study showed that the lowest available dose of atorvastatin was able to reverse the OCA‐mediated increase in LDL‐C to below baseline levels. However, atorvastatin did not reverse the OCA‐mediated decrease in HDL‐C. 136 While not meeting the endpoint of NASH resolution, the 18‐month interim analysis of the Phase 3 REGENERATE trial (NCT02548351) revealed that OCA treatment met the primary endpoint of reduction in liver fibrosis (23% OCA vs 12% placebo) with no worsening of NASH. 137 More will be understood about the effects of OCA on hepatic function and long‐term liver‐related outcomes, regardless of alterations in lipid and lipoprotein parameters, when the final results of the REGENERATE trial are reported. Furthermore, additional large clinical studies need to be conducted to monitor the safety of OCA treatment with regard to long‐term CVD outcomes. While the strategies employed in clinical trials of co‐administration of statin therapy in patients with NASH lead to improvement in lipid profile, the “true‐world” clinical practice remains anchored in multi‐society guidelines that recommend monitoring lipid profiles and adjust statin therapy accordingly. Besides the indirect and direct effects on LDL‐C, subjects treated with OCA also experienced pruritis which led to discontinuation during each of the NASH trials and may affect long term use.

Several selective, non‐bile acid FXR agonists are currently in development. While their mechanism of action through FXR and fibroblast growth factor 19 (FGF19) may still raise LDL‐C, they may not elicit other adverse effects of OCA such as pruritus. Recent results from the Phase 2 ATLAS trial with the FXR agonist cilofexor (GS‐09674) revealed that neither monotherapy nor combined therapy reached the pre‐specified endpoint of significant improvement in liver fibrosis without worsening of NASH, leaving questions regarding the potential efficacy of FXR agonists that are not bile acids. The field will watch carefully the results of ongoing Phase 2 NASH studies with the other FXR agonists, such as EDP‐305, LJN452 and LMB763, and lipid and lipoprotein levels are being closely monitored in these trials.

Aldafermin (NGM282) is an engineered analogue of the gut hormone FGF19 that potently inhibits de novo bile acid synthesis and reduces liver fat content, ALT and AST, while eliminating the FGF19 associated tumorigenicity. 138 , 139 More importantly, NGM282 treatment has been shown to improve the histological features of NASH in only 12 weeks with significant reductions in the NAFLD activity score of two or more points without worsening in liver fibrosis. 140 All components of NAFLD activity score (steatosis, hepatocellular ballooning and lobular inflammation) improved significantly from baseline with NGM282 treatment. 140 A significant portion of the NASH patients treated with NGM282 (42%) exhibited a decrease in the fibrosis score from baseline of at least one stage in only 12 weeks. In addition, NGM282 treatment led to the reduction in the liver fibrosis marker N‐terminal type III collagen propeptide (Pro‐C3) and the Enhanced Liver Fibrosis (ELF) score. 140 Along with the impressive efficacy, NGM282 suppresses hepatic bile acid synthesis from cholesterol which leads to an increase in serum TC and LDL‐C (Figure 3). 139 A comprehensive lipoprotein class/subclass analysis of a recent Phase 2 trial revealed that NGM282 treatment resulted in significant elevation in total LDL particle concentrations, driven mainly by an increase in large‐buoyant LDL particles. 141 Also consistent with its mechanism‐of‐action of reducing insulin resistance and hepatic lipogenesis, NGM282 elicited reductions in large VLDL, mean VLDL size and small HDL particles. Importantly, this study showed that initiation of a statin in NGM282‐treated patients resulted in a rapid decline in plasma lipid levels (Figure 3), showing that the increase in LDL‐C is related to the LDL receptor and that statins can reduce the increased LDL‐C observed with NGM‐282 treatment. 141 At 12 weeks, there were significant reductions in TC, LDL‐C, TG, total LDL and VLDL particle concentrations, and elevations in HDL‐C. 141 The reduction in LDL was driven mainly by a reduction in large LDL. 141 It can be concluded that statin treatment leads to an increase in LDL receptor expression on the hepatocyte surface, thereby reducing LDL‐C and LDL particle concentrations in NASH patients treated with NGM282 (Figure 3). The fact that NASH patients may receive a benefit from NGM282 treatment for their liver disease simultaneously with a statin‐mediated reduction in their CVD risk factors suggests that Aldafermin (NGM282) coupled with a statin may be useful for the treatment of NASH despite initial concerns regarding increased LDL‐C (Figure 3).

3.8. Fibroblast growth factor 21 analogues

Pegbelfermin (BMS‐986036) is a pegylated human fibroblast growth factor 21 (FGF21) analogue that was recently tested in patients with either obesity, T2D or NASH. FGF21 is a key regulator of energy metabolism that is produced by the liver, adipose tissue and pancreas. Pegbelfermin treatment for 12 weeks increased HDL‐C, adiponectin levels and insulin sensitivity, and reduced weight, ALT, AST, TG and Pro‐C3 in subjects who were obese with T2D. 142 In patients with NASH, daily and weekly treatment for 16 weeks led to a significant decrease in hepatic fat fraction as well as an increase in adiponectin and HDL‐C. 143 There was also a reduction in LDL‐C in the once daily arm of the study suggesting that pegbelfermin treatment may lead to beneficial changes in lipid levels. 143 Future studies with larger numbers of subjects will need to be done to fully understand the effects of this FGF21 analogue on lipids and CVD risk. Phase 2b study (FALCON 1) is underway in patients with biopsy‐proven NASH in order to evaluate potential changes in histological endpoints (NCT03486899). Another FGF21 “mimic,” a bispecific antibody agonist to the receptor complex for FGF21 (FGFR1c and βKlotho) called BFKB8488A, is also in clinical development for NAFLD (NCT03060538). Because these agonists may interact with the receptors in very different ways, it will be interesting to see if their effects on lipoproteins differ as well.

3.9. Acetyl‐CoA carboxylase inhibitors

Acetyl‐CoA carboxylase (ACC) catalyses the first committed step of de novo fatty acid synthesis. Therefore, it has been hypothesised that ACC inhibition may be a target for reducing hepatic steatosis and treating patients with insulin resistance, T2D and NAFLD. Currently there are ACC inhibitors in clinical development that may provide answers to these questions. Firsocostat (GS‐0976) is a liver‐targeted, dual ACC1/ACC2 inhibitor, that was tested in a Phase 2 trial in patients with NASH. After 12 weeks administration of GS‐0976, patients with NASH exhibited reduced hepatic de novo lipogenesis, hepatic steatosis and markers of liver injury. 144 , 145 However, median increases of 11%‐13% in TG were observed. Sixteen of 100 patients treated with GS‐0976 developed substantial hypertriglyceridaemia (>500 mg/dL), which was associated with baseline TG of ≥250 mg/dL. Four of these subjects received fibrates or fish oil, which reduced their TG levels to <500 mg/dL by week 12. 145 In order to gain a more comprehensive view of the changes in lipid‐related CVD risk factors, a complete lipoprotein class/subclass profile was obtained. Treatment with GS‐0976 led to an increase in VLDL particles and VLDL‐TG that peaked at 1 week. 145 Additionally, there was no change in TC, HDL‐C, LDL‐C, total LDL or small LDL particle concentrations (Table 1). 145 Therefore, GS‐0976 treatment did not elicit alterations suggestive of an atherogenic lipoprotein phenotype. The mechanisms for the increased TG are thought to be due to a combination of increased VLDL synthesis and secretion 146 and a reduction in TG clearance due to a decrease in peripheral lipoprotein lipase activity. Similar findings were observed with another ACC inhibitor, MK‐4704. 147 Because elevated serum TG may increase risk of CVD and, in extreme cases, pancreatitis, treatment with ACC1/ACC2 inhibitors would likely require lipid monitoring and sometimes addition of TG lowering therapy. 145 PF‐05221304 is a liver‐targeted ACC1/ACC2 inhibitor that was evaluated in a Phase 1 clinical study in healthy subjects. 148 PF‐05221304 demonstrated robust inhibition of de novo lipogenesis in NASH patients, with minimal effects on platelet count or TG. 148 Given the effects of previously tested ACC inhibitors, a comprehensive lipid and lipoprotein profile should be conducted in future studies in NASH patients.

3.10. Anti‐chemokine receptor therapeutic agents

The chemokine receptor type 2/5 (CCR2/5) antagonist, cenicriviroc, has been shown to have anti‐inflammatory and anti‐fibrotic properties in preclinical and clinical studies. 149 By virtue of its antagonism of CCR2, cenicriviroc is expected to impair the recruitment, migration, and infiltration of pro‐inflammatory monocytes and macrophages at the site of liver injury. In a recent Phase 2b study, cenicriviroc achieved the fibrosis endpoint (improvement in fibrosis ≥1 with no worsening of NASH) in a larger percentage of subjects than placebo. 149 , 150 While biomarkers of systemic inflammation were reduced with cenicriviroc treatment, no changes in lipid or lipoprotein levels were reported.

3.11. Apoptosis inhibitors

Selonsertib (GS‐4997) is a selective inhibitor of apoptosis signal‐regulating kinase (ASK1), a ubiquitously expressed serine/threonine kinase that is activated by oxidative stress to promote hepatocellular apoptosis, inflammation and fibrosis. 151 Recently selonsertib was tested in a Phase 2 trial in NASH patients with or without addition of simtuzumab (GS‐6624), a humanized monoclonal antibody to LOXL2 that failed to show efficacy for reducing fibrosis in a NASH trial. 151 , 152 After 24 weeks of treatment in patients with biopsy‐proven NASH, the proportion of patients with ≥1 stage reduction in fibrosis was: 43% with selonsertib + simtuzumab, 30% with selonsertib + simtuzumab and 20% with simtuzumab‐alone. Improvements in fibrosis were associated with reductions in liver stiffness by magnetic resonance elastography, collagen content and lobular inflammation on liver biopsy. 151 Unfortunately, two Phase 3 studies with selonsertib (STELLAR‐3 in subjects with NASH and bridging fibrosis and STELLAR‐4 in subjects with NASH and compensated cirrhosis) did not meet the primary endpoint of ≥1‐stage histologic improvement in fibrosis without worsening of NASH, suggesting that a apoptosis signal‐regulating kinase (ASK1) inhibition alone may not be sufficient to reduce fibrotic disease in the liver. No lipid or lipoprotein results were reported for any of these trials.

Emricasan (IDN‐6556) is an irreversible pan‐caspase inhibitor that has been tested recently in patients with various liver diseases. 153 , 154 , 155 Emricasan treatment lowered portal pressure in patients with compensated cirrhosis and severe portal hypertension 154 and improved hepatic function in patients with cirrhosis. 155 In a pilot study in patients with NAFLD, emricasan treatment led to a reduction in ALT and cytokeratin 18 (CK18), suggesting that emricasan should be tested in larger trials in patients with NASH. 153 Unfortunately in a recent Phase 2b trial, emricasan treatment did not improve histological endpoints in patients with NASH and liver fibrosis and may have even worsened fibrosis and ballooning. 156 To date, no effects on lipoproteins, lipids or other CVD risk factors have been reported.

3.12. Thyroid hormone receptor β agonist

Resmetirom (MGL‐3196) is a highly selective thyroid hormone receptor β (THRβ) agonist. 157 THRβ‐selective agonism is hypothesised to elicit the beneficial effects on dyslipidaemia without eliciting the adverse effects of thyroid hormone receptor α agonism in heart and bone. In a Phase 1 study, MGL‐3196 dosed at 50 mg per day or higher for 2 weeks significantly reduced LDL‐C by 30% and showed a trend for decreased TG. 158 A Phase 2 trial in patients with biopsy‐proven NASH revealed that resmetirom treatment resulted in a significant reduction in hepatic fat content as well as in multiple pro‐atherogenic proteins and lipids including LDL‐C, apoB, TG, apolipoprotein CIII, lipoprotein (a), small dense LDL particles and large VLDL. 159 Notably, resmetirom is one of the few potential therapeutics for NASH that has been shown to reduce lipoprotein (a), a lipoprotein closely associated with atherogenic risk and CVD.

3.13. Stearoyl‐CoA desaturase‐1 inhibitor

Aramchol is a synthetic lipid inhibitor of stearoyl‐CoA desaturase‐1 (SCD1) that was tested for efficacy in patients with biopsy‐proven NAFLD. 160 Aramchol treatment (300 mg) elicited a reduction in liver fat content and is currently being evaluated in a Phase 2b trial in NASH patients (NCT02279524). Because SCD1 is an enzyme that catalyses the synthesis of monounsaturated FA from saturated FA, there may be an effect of SCD1 inhibition on the lipid species carried by lipoprotein particles. However, effects of SCD1 inhibition on the overall lipoprotein profile are not known at this time. Therefore, it is difficult to predict if SCD1 inhibition will affect CVD risk with long‐term treatment.

3.14. Mitochondrial pyruvate carrier modulator

MSDC‐0602K is a next‐generation insulin sensitizer that modulates the mitochondrial pyruvate carrier (MPC), but avoids direct PPARγ activation. 161 In mice fed a diet rich in trans‐fatty acid, fructose and cholesterol, MSDC‐0602 prevented and reversed liver fibrosis and reduced hepatic markers of stellate cell activation presumably by modulating pyruvate metabolism since reduction of MPC expression in hepatocytes led to similar effects. 162 In the one‐year Phase 2 trial in NASH subjects with and without type 2 diabetes (EMMINENCE), MSDC‐0602K treatment produced dose‐dependent reductions in glycaemia and fasting insulin, and while there no changes in overall lipids, there were shifts in particles that could be consistent with a reduction in atherogenic dyslipidaemia secondary to increased insulin sensitivity. 163 Treatment led to an increase in VLDL particles, largely due to an increase in small VLDL that was not accompanied by a significant increase in TG. 163 MSDC‐0602K treatment also led to an increase in large LDL and a reduction in small dense LDL particles. 163 In addition, there was an increase in medium HDL and a decrease in small HDL, which led to an overall increase in total HDL particles. 163

4. CONCLUSIONS

Recently physicians have begun to appreciate the fact that many therapeutics on the market, or currently in development, elicit effects on lipoprotein metabolism leading to direct or indirect effects on circulating lipid and lipoprotein levels. Many of these medications have overall positive effects lipid profiles, however, there are a number of drug classes that negatively impact circulating lipids and lipoproteins, leading to concern that these agents may augment CVD progression. This is especially true for anti‐diabetic agents, some of which are in development for treatment of NAFLD/NASH. Some of these agents have direct effects (increased hepatic cholesterol content leading to increased serum LDL‐C), indirect effects (enhanced insulin sensitivity and reversal of atherogenic dyslipidaemia) or both. Moreover the mechanisms for the alterations in the lipoprotein profile often differ. Evidence to date suggests that these drug‐induced alterations may not necessarily lead to increased CVD risk in NASH patients. Co‐administration of lipid modifying therapies that reduce CVD risk factors such as statins may help reduce CVD risk, however, larger clinical studies are needed in order to prove this to be the case for each of the potential therapeutics in question. It is clear, however, that in order to fully understand the overall impact of these agents on both hepatic and CVD outcomes, it may be necessary to consider all of the benefits and risks that are associated with each of these drugs or drug candidates.

AUTHORSHIP

Guarantor of the article: None.

Authorship contributions: MAC wrote the first draft of the article. JVR, MSS, JRG and AS critically reviewed and revised the manuscript. All authors approved the final version for publication.

ACKNOWLEDGEMENTS

The authors thank Drs. Damiano and Garcia for their careful review of the manuscript and valuable input as well as Sarah Perry for her assistance with Figure 1.

Declaration of personal interest: Dr Connelly is an employee of and owns stock in LabCorp. Dr Velez Rivera has no conflicts to disclose. Dr Guyton has received research support from Sanofi, Regeneron, Amgen and Amarin. Dr Siddiqui is a member of an advisory board at Pfizer and receives research grant support from Zydus. Dr Sanyal is the President of Sanyal Biotechnology and has stock options in Genfit, Akarna, Tiziana, Indalo, Durect Inversago and Galmed. Has served as a consultant to Astra Zeneca, Nitto Denko, Conatus, Nimbus, Salix, Tobira, Takeda, Jannsen, Gilead, Terns, Birdrock, Merck, Valeant, Boehringer‐Ingelheim, Bristol Myers Squibb, Lilly, Hemoshear, Zafgen, Novartis, Novo Nordisk, Pfizer, Exhalenz and Genfit. He has been an unpaid consultant to Intercept, Echosens, Immuron, Galectin, Fractyl, Syntlogic, Affimune, Chemomab, Zydus, Nordic Bioscience, Albireo, Prosciento, Surrozen. His institution has received grant support from Gilead, Salix, Tobira, Bristol Myers, Shire, Intercept, Merck, Astra Zeneca, Malinckrodt, Cumberland and Novartis. He receives royalties from Elsevier and UptoDate. Virginia Commonwealth University, where he is a full time employee, also has ownership interests in Sanyal Biotechnology. Dr Sanyal also acknowledges funding from NIH project R01 DK 105961.

Connelly MA, Velez Rivera J, Guyton JR, Siddiqui MS, Sanyal AJ. Review article: the impact of liver‐directed therapies on the atherogenic risk profile in non‐alcoholic steatohepatitis. Aliment Pharmacol Ther. 2020;52:619–636. 10.1111/apt.15935

The Handling Editor for this article was Dr Stephen Ryder, and this uncommissioned review was accepted for publication after full peer‐review.

REFERENCES

  • 1. Sanyal AJ. Past, present and future perspectives in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol. 2019;16:377‐386. [DOI] [PubMed] [Google Scholar]
  • 2. Younossi ZM. Non‐alcoholic fatty liver disease ‐ a global public health perspective. J Hepatol. 2019;70:531‐544. [DOI] [PubMed] [Google Scholar]
  • 3. Targher G, Day CP, Bonora E. Risk of cardiovascular disease in patients with nonalcoholic fatty liver disease. N Engl J Med. 2010;363:1341‐1350. [DOI] [PubMed] [Google Scholar]
  • 4. Di Costanzo A, D'Erasmo L, Polimeni L, et al. Non‐alcoholic fatty liver disease and subclinical atherosclerosis: a comparison of metabolically‐ versus genetically‐driven excess fat hepatic storage. Atherosclerosis. 2017;257:232‐239. [DOI] [PubMed] [Google Scholar]
  • 5. Vilar‐Gomez E, Martinez‐Perez Y, Calzadilla‐Bertot L, et al. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology. 2015;149:367‐378.e5; quiz e314–365. [DOI] [PubMed] [Google Scholar]
  • 6. Herink M, Ito MK. Medication induced changes in lipid and lipoproteins In:Feingold KR, Anawalt B, Boyce A, eds. Endotext. South Dartmouth, MA: 2000–2018 May 10. [PubMed] [Google Scholar]
  • 7. Rosenblit PD. Common medications used by patients with type 2 diabetes mellitus: what are their effects on the lipid profile? Cardiovasc Diabetol. 2016;15:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Friedman SL, Neuschwander‐Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 2018;24:908‐922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Noureddin M, Muthiah MD, Sanyal AJ. Drug discovery and treatment paradigms in nonalcoholic steatohepatitis. Endocrinol Diab Metab. 2019;e00105:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Turner RC, Millns H, Neil HAW, et al. Risk factors for coronary artery disease in non‐insulin dependent diabetes mellitus: United Kingdom Prospective Diabetes Study (UKPDS: 23). BMJ. 1998;316:823‐828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Miller M, Stone NJ, Ballantyne C, et al. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2011;123:2292‐2333. [DOI] [PubMed] [Google Scholar]
  • 12. Ginsberg HN. Lipoprotein physiology in nondiabetic and diabetic states. Relationship to atherogenesis. Diabetes Care. 1991;14:839‐855. [DOI] [PubMed] [Google Scholar]
  • 13. Saydah SH, Fradkin J, Cowie CC. Poor control of risk factors for vascular disease among adults with previously diagnosed diabetes. JAMA. 2004;291:335‐342. [DOI] [PubMed] [Google Scholar]
  • 14. Adiels M, Olofsson SO, Taskinen MR, Boren J. Overproduction of very low‐density lipoproteins is the hallmark of the dyslipidemia in the metabolic syndrome. Arterioscler Thromb Vasc Biol. 2008;28:1225‐1236. [DOI] [PubMed] [Google Scholar]
  • 15. DeFilippis AP, Blaha MJ, Martin SS, et al. Nonalcoholic fatty liver disease and serum lipoproteins: the Multi‐Ethnic Study of Atherosclerosis. Atherosclerosis. 2013;227:429‐436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Siddiqui MS, Sterling RK, Luketic VA, et al. Association between high‐normal levels of alanine aminotransferase and risk factors for atherogenesis. Gastroenterology. 2013;145:1271‐1279.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Verges B. Pathophysiology of diabetic dyslipidaemia: where are we? Diabetologia. 2015;58:886‐899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Siddiqui MS, Fuchs M, Idowu MO, et al. Severity of nonalcoholic fatty liver disease and progression to cirrhosis are associated with atherogenic lipoprotein profile. Clin Gastroenterol Hepatol. 2015;13:1000‐1008.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Katsiki N, Mikhailidis DP, Mantzoros CS. Non‐alcoholic fatty liver disease and dyslipidemia: an update. Metabolism. 2016;65:1109‐1123. [DOI] [PubMed] [Google Scholar]
  • 20. Nemes K, Aberg F. Interpreting lipoproteins in nonalcoholic fatty liver disease. Curr Opin Lipidol. 2017;28:355‐360. [DOI] [PubMed] [Google Scholar]
  • 21. Cromwell WC, Triffon DW. Clinical utility of LDL particle number to optimize management of LDL‐related cardiovascular risk. J Fam Pract. 2016;65:1‐8. [Google Scholar]
  • 22. Berneis KK, Krauss RM. Metabolic origins and clinical significance of LDL heterogeneity. J Lipid Res. 2002;43:1363‐1379. [DOI] [PubMed] [Google Scholar]
  • 23. Tabas I. Cholesterol in health and disease. J Clin Invest. 2002;110:583‐590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Goff DC Jr, D'Agostino RB Jr, Haffner SM, Otvos JD. Insulin resistance and adiposity influence lipoprotein size and subclass concentrations. Results from the Insulin Resistance Atherosclerosis Study. Metabolism. 2005;54:264‐270. [DOI] [PubMed] [Google Scholar]
  • 25. Arai H, Ishibashi S, Bujo H, et al. Management of type IIb dyslipidemia. J Atheroscler Thromb. 2012;19:105‐114. [DOI] [PubMed] [Google Scholar]
  • 26. Packard CJ. Triacylglycerol‐rich lipoproteins and the generation of small, dense low‐density lipoprotein. Biochem Soc Trans. 2003;31:1066‐1069. [DOI] [PubMed] [Google Scholar]
  • 27. Gardner CD, Fortmann SP, Krauss RM. Association of small low‐density lipoprotein particles with the incidence of coronary artery disease in men and women. JAMA. 1996;276:875‐881. [PubMed] [Google Scholar]
  • 28. Howard BV, Robbins DC, Sievers ML, et al. LDL cholesterol as a strong predictor of coronary heart disease in diabetic individuals with insulin resistance and low LDL: the Strong Heart Study. Arterioscler Thromb Vasc Biol. 2000;20:830‐835. [DOI] [PubMed] [Google Scholar]
  • 29. Blake GJ, Otvos JD, Rifai N, Ridker PM. Low‐density lipoprotein particle concentration and size as determined by nuclear magnetic resonance spectroscopy as predictors of cardiovascular disease in women. Circulation. 2002;106:1930‐1937. [DOI] [PubMed] [Google Scholar]
  • 30. Carmena R, Duriez P, Fruchart JC. Atherogenic lipoprotein particles in atherosclerosis. Circulation. 2004;109:III‐2‐III‐7. [DOI] [PubMed] [Google Scholar]
  • 31. Cromwell WC, Otvos JD, Keyes MJ, et al. LDL particle number and risk of future cardiovascular disease in the Framingham Offspring Study ‐ implications for LDL management. J Clin Lipidol. 2007;1:583‐592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Mora S, Szklo M, Otvos JD, et al. LDL particle subclasses, LDL particle size, and carotid atherosclerosis in the Multi‐Ethnic Study of Atherosclerosis (MESA). Atherosclerosis. 2007;192:211‐217. [DOI] [PubMed] [Google Scholar]
  • 33. Arca M, Pigna G, Favoccia C. Mechanisms of diabetic dyslipidemia: relevance for atherogenesis. Curr Vasc Pharmacol. 2012;10:684‐686. [DOI] [PubMed] [Google Scholar]
  • 34. Lamarche B, Tchernof A, Moorjani S, et al. Small, dense low‐density lipoprotein particles as a predictor of the risk of ischemic heart disease in men. Prospective results from the Quebec Cardiovascular Study. Circulation. 1997;95:69‐75. [DOI] [PubMed] [Google Scholar]
  • 35. Rosenson RS, Otvos JD, Freedman DS. Relations of lipoprotein subclass levels and low‐density lipoprotein size to progression of coronary artery disease in the Pravastatin Limitation of Atherosclerosis in the Coronary Arteries (PLAC‐I) trial. Am J Cardiol. 2002;90:89‐94. [DOI] [PubMed] [Google Scholar]
  • 36. Otvos JD, Collins D, Freedman DS, et al. Low‐density lipoprotein and high‐density lipoprotein particle subclasses predict coronary events and are favorably changed by gemfibrozil therapy in the Veterans Affairs High‐Density Lipoprotein Intervention Trial. Circulation. 2006;113:1556‐1563. [DOI] [PubMed] [Google Scholar]
  • 37. El Harchaoui K, van der Steeg WA, Stroes ESG, et al. Value of low‐density lipoprotein particle number and size as predictors of coronary artery disease in apparently healthy men and women: the EPIC‐Norfolk Prospective Population Study. J Am Coll Cardiol. 2007;49:547‐553. [DOI] [PubMed] [Google Scholar]
  • 38. Brunzell JD, Davidson M, Furberg CD, et al. Lipoprotein management in patients with cardiometabolic risk: consensus conference report from the American Diabetes Association and the American College of Cardiology Foundation. J Am Coll Cardiol. 2008;51:1512‐1524. [DOI] [PubMed] [Google Scholar]
  • 39. Mora S, Otvos JD, Rifai N, Rosenson RS, Buring JE, Ridker PM. Lipoprotein particle profiles by nuclear magnetic resonance compared with standard lipids and apolipoproteins in predicting incident cardiovascular disease in women. Circulation. 2009;119:931‐939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Arsenault BJ, Després J‐P, Stroes ESG, et al. Lipid assessment, metabolic syndrome and coronary heart disease risk. Eur J Clin Invest. 2010;40:1081‐1093. [DOI] [PubMed] [Google Scholar]
  • 41. Master SR, Rader DJ. Beyond LDL cholesterol in assessing cardiovascular risk: apo B or LDL‐P? Clin Chem. 2013;59:723‐725. [DOI] [PubMed] [Google Scholar]
  • 42. Packard CJ, Demant T, Stewart JP, et al. Apolipoprotein B metabolism and the distribution of VLDL and LDL subfractions. J Lipid Res. 2000;41:305‐318. [PubMed] [Google Scholar]
  • 43. Shalaurova I, Connelly MA, Garvey WT, Otvos JD. Lipoprotein insulin resistance index: a lipoprotein particle‐derived measure of insulin resistance. Metab Syndr Relat Disord. 2014;12:422‐429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Garber A, Abrahamson M, Barzilay J, et al. AACE comprehensive diabetes management algorithm 2013. Endocr Pract. 2013;19:327‐336. [DOI] [PubMed] [Google Scholar]
  • 45. Jeyarajah EJ, Cromwell WC, Otvos JD. Lipoprotein particle analysis by nuclear magnetic resonance spectroscopy. Clin Lab Med. 2006;26:847‐870. [DOI] [PubMed] [Google Scholar]
  • 46. Chakravarthy MV, Neuschwander‐Tetri, BA . The meatabolic basis of nonalcoholic steatohepatitis. Endocrinol Diab Metab. 2020;00:e00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Kenneally S, Sier JH, Moore JB. Efficacy of dietary and physical activity intervention in non‐alcoholic fatty liver disease: a systematic review. BMJ Open Gastroenterol. 2017;4:e000139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Katsagoni CN, Georgoulis M, Papatheodoridis GV, Panagiotakos DB, Kontogianni MD. Effects of lifestyle interventions on clinical characteristics of patients with non‐alcoholic fatty liver disease: a meta‐analysis. Metabolism. 2017;68:119‐132. [DOI] [PubMed] [Google Scholar]
  • 49. Hallsworth K, Adams LA. Lifestyle modification in NAFLD/NASH: facts and figures. J Hepatol Rep. 2019;1:468‐479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Mardinoglu A, Wu H, Bjornson E, et al. An integrated understanding of the rapid metabolic benefits of a carbohydrate‐restricted diet on hepatic steatosis in humans. Cell Metab. 2018;27(3):559‐571.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Skytte MJ, Samkani A, Petersen AD, et al. A carbohydrate‐reduced high‐protein diet improves HbA1c and liver fat content in weight stable participants with type 2 diabetes: a randomised controlled trial. Diabetologia. 2019;62:2066‐2078. [DOI] [PubMed] [Google Scholar]
  • 52. Yki‐Jarvinen H. Nutritional modulation of non‐alcoholic fatty liver disease and insulin resistance. Nutrients. 2015;7:9127‐9138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Bhanpuri NH, Hallberg SJ, Williams PT, et al. Cardiovascular disease risk factor responses to a type 2 diabetes care model including nutritional ketosis induced by sustained carbohydrate restriction at 1 year: an open label, non‐randomized, controlled study. Cardiovasc Diabetol. 2018;17:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Seshadri P, Iqbal N, Stern L, et al. A randomized study comparing the effects of a low‐carbohydrate diet and a conventional diet on lipoprotein subfractions and C‐reactive protein levels in patients with severe obesity. Am J Med. 2004;117:398‐405. [DOI] [PubMed] [Google Scholar]
  • 55. Siri PW, Krauss RM. Influence of dietary carbohydrate and fat on LDL and HDL particle distributions. Curr Atheroscler Rep. 2005;7:455‐459. [DOI] [PubMed] [Google Scholar]
  • 56. Volek JS, Fernandez ML, Feinman RD, Phinney SD. Dietary carbohydrate restriction induces a unique metabolic state positively affecting atherogenic dyslipidemia, fatty acid partitioning, and metabolic syndrome. Prog Lipid Res. 2008;47:307‐318. [DOI] [PubMed] [Google Scholar]
  • 57. Mollard RC, Senechal M, MacIntosh AC, et al. Dietary determinants of hepatic steatosis and visceral adiposity in overweight and obese youth at risk of type 2 diabetes. Am J Clin Nutr. 2014;99:804‐812. [DOI] [PubMed] [Google Scholar]
  • 58. Basaranoglu M, Kayacetin S, Yilmaz N, Kayacetin E, Tarcin O, Sonsuz A. Understanding mechanisms of the pathogenesis of nonalcoholic fatty liver disease. World J Gastroenterol. 2010;16:2223‐2226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Cohen DE, Anania FA, Chalasani N. National Lipid Association Statin Safety Task Force Liver Expert Panel. An assessment of statin safety by hepatologists. Am J Cardiol. 2006;97:77C‐81C. [DOI] [PubMed] [Google Scholar]
  • 60. Jacobson TA. NLA task force on statin safety–2014 update. J Clin Lipidol. 2014;8:S1‐S4. [DOI] [PubMed] [Google Scholar]
  • 61. Bays H, Cohen DE, Chalasani N, Harrison SA. The National Lipid Association's Statin Safety Task Force. An assessment by the statin liver safety task force: 2014 update. J Clin Lipidol. 2014;8:S47‐S57. [DOI] [PubMed] [Google Scholar]
  • 62. Patel SS, Guzman LA, Lin F‐P, et al. Utilization of aspirin and statin in management of coronary artery disease in patients with cirrhosis undergoing liver transplant evaluation. Liver Transpl. 2018;24:872‐880. [DOI] [PubMed] [Google Scholar]
  • 63. Tikkanen MJ, Fayyad R, Faergeman O, et al. Effect of intensive lipid lowering with atorvastatin on cardiovascular outcomes in coronary heart disease patients with mild‐to‐moderate baseline elevations in alanine aminotransferase levels. Int J Cardiol. 2013;168:3846‐3852. [DOI] [PubMed] [Google Scholar]
  • 64. Athyros VG, Tziomalos K, Gossios TD, et al. Safety and efficacy of long‐term statin treatment for cardiovascular events in patients with coronary heart disease and abnormal liver tests in the Greek Atorvastatin and Coronary Heart Disease Evaluation (GREACE) study: a post‐hoc analysis. Lancet. 2010;376:1916‐1922. [DOI] [PubMed] [Google Scholar]
  • 65. Athyros VG, Alexandrides TK, Bilianou H, et al. The use of statins alone, or in combination with pioglitazone and other drugs, for the treatment of non‐alcoholic fatty liver disease/non‐alcoholic steatohepatitis and related cardiovascular risk. An Expert Panel Statement. Metabolism. 2017;71:17‐32. [DOI] [PubMed] [Google Scholar]
  • 66. Tikkanen MJ, Szarek M, Fayyad R, et al. Total cardiovascular disease burden: comparing intensive with moderate statin therapy insights from the IDEAL (Incremental Decrease in End Points through Aggressive Lipid Lowering) trial. J Am Coll Cardiol. 2009;54:2353‐2357. [DOI] [PubMed] [Google Scholar]
  • 67. Athyros VG, Mikhailidis DP, Didangelos TP, et al. Effect of multifactorial treatment on non‐alcoholic fatty liver disease in metabolic syndrome: a randomised study. Curr Med Res Opin. 2006;22:873‐883. [DOI] [PubMed] [Google Scholar]
  • 68. Eslami L, Merat S, Malekzadeh R, Nasseri‐Moghaddam S, Aramin H. Statins for non‐alcoholic fatty liver disease and non‐alcoholic steatohepatitis. Cochrane Database Syst Rev. 2013;12:CD008623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Blais P, Lin M, Kramer JR, El‐Serag HB, Kanwal F. Statins are underutilized in patients with nonalcoholic fatty liver disease and dyslipidemia. Dig Dis Sci. 2016;61:1714‐1720. [DOI] [PubMed] [Google Scholar]
  • 70. Lee I‐M, Cook NR, Gaziano JM, et al. Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women's Health Study: a randomized controlled trial. JAMA. 2005;294:56‐65. [DOI] [PubMed] [Google Scholar]
  • 71. Heart Outcomes Prevention Evaluation Study Investigators ; Yusuf S, Dagenais G, Pogue J, Bosch J, Sleight P. Vitamin E supplementation and cardiovascular events in high‐risk patients. N Engl J Med. 2000;342:154‐160. [DOI] [PubMed] [Google Scholar]
  • 72. Sanyal AJ, Chalasani N, Kowdley KV, et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 2010;362:1675‐1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of non‐alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology. 2012;55:2005‐2023. [DOI] [PubMed] [Google Scholar]
  • 74. Lavine JE, Schwimmer JB, Van Natta ML, et al. Effect of vitamin E or metformin for treatment of nonalcoholic fatty liver disease in children and adolescents: the TONIC randomized controlled trial. JAMA. 2011;305:1659‐1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Perumpail B, Li A, John N, et al. The role of vitamin E in the treatment of NAFLD. Diseases. 2018;6:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Bril F, Biernacki DM, Kalavalapalli S, et al. Role of vitamin E for nonalcoholic steatohepatitis in patients with type 2 diabetes: a randomized controlled trial. Diab Care. 2019;42:1481‐1488. [DOI] [PubMed] [Google Scholar]
  • 77. Sato K, Gosho M, Yamamoto T, et al. Vitamin E has a beneficial effect on nonalcoholic fatty liver disease: a meta‐analysis of randomized controlled trials. Nutrition. 2015;31:923‐930. [DOI] [PubMed] [Google Scholar]
  • 78. Banini BA, Cazanave SC, Yates KP, et al. Haptoglobin 2 allele is associated with histologic response to vitamin E in subjects with nonalcoholic steatohepatitis. J Clin Gastroenterol. 2019;53:750‐758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Hochberg I, Berinstein EM, Milman U, Shapira C, Levy AP. Interaction between the haptoglobin genotype and vitamin E on cardiovascular disease in diabetes. Curr Diab Rep. 2017;17:42. [DOI] [PubMed] [Google Scholar]
  • 80. Brown JD, Plutzky J. Peroxisome proliferator‐activated receptors as transcriptional nodal points and therapeutic targets. Circulation. 2007;115:518‐533. [DOI] [PubMed] [Google Scholar]
  • 81. Derosa G, Sahebkar A, Maffioli P. The role of various peroxisome proliferator‐activated receptors and their ligands in clinical practice. J Cell Physiol. 2018;233:153‐161. [DOI] [PubMed] [Google Scholar]
  • 82. Musso G, Gambino R, Durazzo M, Cassader M. Noninvasive assessment of liver disease severity with liver fat score and CK‐18 in NAFLD: prognostic value of liver fat equation goes beyond hepatic fat estimation. Hepatology. 2010;51:715‐717. [DOI] [PubMed] [Google Scholar]
  • 83. 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] [PMC free article] [PubMed] [Google Scholar]
  • 84. Cusi K, Orsak B, Bril F, et al. 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] [PubMed] [Google Scholar]
  • 85. Corey KE, Wilson LA, Altinbas A, et al. Relationship between resolution of non‐alcoholic steatohepatitis and changes in lipoprotein sub‐fractions: a post‐hoc analysis of the PIVENS trial. Aliment Pharmacol Ther. 2019;49:1205‐1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Goldberg RB, Kendall DM, Deeg MA, et al. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diab Care. 2005;28:1547‐1554. [DOI] [PubMed] [Google Scholar]
  • 87. Hanefeld M. The role of pioglitazone in modifying the atherogenic lipoprotein profile. Diab Obes Metab. 2009;11:742‐756. [DOI] [PubMed] [Google Scholar]
  • 88. Dormandy JA, Charbonnel B, Eckland DJA, et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet. 2005;366:1279‐1289. [DOI] [PubMed] [Google Scholar]
  • 89. Erdmann E, Harding S, Lam H, Perez A. Ten‐year observational follow‐up of PROactive: a randomized cardiovascular outcomes trial evaluating pioglitazone in type 2 diabetes. Diab Obes Metab. 2016;18:266‐273. [DOI] [PubMed] [Google Scholar]
  • 90. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med. 2007;356:2457‐2471. [DOI] [PubMed] [Google Scholar]
  • 91. DREAM (Diabetes REduction Assessment with ramipril and rosiglitazone Medication) Trial Investigators , Gerstein HC, Yusuf S, et al. Effect of rosiglitazone on the frequency of diabetes in patients with impaired glucose tolerance or impaired fasting glucose: a randomised controlled trial. Lancet. 2006;368:1096‐1105. [DOI] [PubMed] [Google Scholar]
  • 92. Home PD, Pocock SJ, Beck‐Nielsen H, et al. Rosiglitazone evaluated for cardiovascular outcomes–an interim analysis. N Engl J Med. 2007;357:28‐38. [DOI] [PubMed] [Google Scholar]
  • 93. Park JY, Kim KA, Kang MH, Kim SL, Shin JG. Effect of rifampin on the pharmacokinetics of rosiglitazone in healthy subjects. Clin Pharmacol Ther. 2004;75:157‐162. [DOI] [PubMed] [Google Scholar]
  • 94. Ratziu V, Harrison SA, Francque S, et al. Elafibranor, an agonist of the peroxisome proliferator‐activated receptor‐alpha and ‐delta, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening. Gastroenterology. 2016;150:1147‐1159.45. [DOI] [PubMed] [Google Scholar]
  • 95. Bays HE, Schwartz S, Littlejohn T 3rd, et al. 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] [PubMed] [Google Scholar]
  • 96. Choi YJ, Roberts BK, Wang X, et al. Effects of the PPAR‐delta agonist MBX‐8025 on atherogenic dyslipidemia. Atherosclerosis. 2012;220:470‐476. [DOI] [PubMed] [Google Scholar]
  • 97. Gawrieh S, Mazen N, Loo NM, et al. A phase 2, prospective, multicenter, double‐blind, randomized study of saroglitazar magnesium 1 mg, 2 mg or 4 mg versus placebo in patients with nonalcoholic fatty liver disease and/or nonalcoholic steatohepatitis (EVIDENCES IV). Hepatology. 2019;70:1484‐1485.31127640 [Google Scholar]
  • 98. Kaul U, Parmar D, Manjunath K, et al. New dual peroxisome proliferator activated receptor agonist‐saroglitazar in diabetic dyslipidemia and non‐alcoholic fatty liver disease: integrated analysis of the real world evidence. Cardiovasc Diabetol. 2019;18:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Nauck MA, Meier JJ. The incretin effect in healthy individuals and those with type 2 diabetes: physiology, pathophysiology, and response to therapeutic interventions. Lancet Diab Endocrinol. 2016;4:525‐536. [DOI] [PubMed] [Google Scholar]
  • 100. Dey AK, Groenendyk J, Mehta NN, Gourgari E. The effect of sodium‐glucose cotransporter 2 inhibitors and glucagon‐like peptide 1 agonists on cardiovascular disease in patients with type 2 diabetes. Clin Cardiol. 2019;42:406‐412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Bethel MA, Patel RA, Merrill P, et al. Cardiovascular outcomes with glucagon‐like peptide‐1 receptor agonists in patients with type 2 diabetes: a meta‐analysis. Lancet Diab Endocrinol. 2018;6:105‐113. [DOI] [PubMed] [Google Scholar]
  • 102. Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double‐blind, randomised placebo‐controlled trial. Lancet. 2019;394:121‐130. [DOI] [PubMed] [Google Scholar]
  • 103. Prasad‐Reddy L, Isaacs D. A clinical review of GLP‐1 receptor agonists: efficacy and safety in diabetes and beyond. Drugs Context. 2015;4:212283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Ariel D, Kim SH, Abbasi F, Lamendola CA, Liu A, Reaven GM. Effect of liraglutide administration and a calorie‐restricted diet on lipoprotein profile in overweight/obese persons with prediabetes. Nutr Metab Cardiovasc Dis. 2014;24:1317‐1322. [DOI] [PubMed] [Google Scholar]
  • 105. Bays H, Pi‐Sunyer X, Hemmingsson JU, Claudius B, Jensen CB, Van Gaal L. Liraglutide 3.0 mg for weight management: weight‐loss dependent and independent effects. Curr Med Res Opin. 2017;33:225‐229. [DOI] [PubMed] [Google Scholar]
  • 106. Armstrong MJ, Gaunt P, Aithal GP, et al. Liraglutide safety and efficacy in patients with non‐alcoholic steatohepatitis (LEAN): a multicentre, double‐blind, randomised, placebo‐controlled phase 2 study. Lancet. 2016;387:679‐690. [DOI] [PubMed] [Google Scholar]
  • 107. Stenlöf K, Cefalu WT, Kim K‐A, et al. Long‐term efficacy and safety of canagliflozin monotherapy in patients with type 2 diabetes inadequately controlled with diet and exercise: findings from the 52‐week CANTATA‐M study. Curr Med Res Opin. 2014;30:163‐175. [DOI] [PubMed] [Google Scholar]
  • 108. McGill JB. The SGLT2 inhibitor empagliflozin for the treatment of type 2 diabetes mellitus: a bench to bedside review. Diab Ther. 2014;5:43‐63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Hayashi T, Fukui T, Nakanishi N, et al. Dapagliflozin decreases small dense low‐density lipoprotein‐cholesterol and increases high‐density lipoprotein 2‐cholesterol in patients with type 2 diabetes: comparison with sitagliptin. Cardiovasc Diabetol. 2017;16:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Bays HE, Sartipy P, Xu J, Sjöström CD, Underberg JA. Dapagliflozin in patients with type II diabetes mellitus, with and without elevated triglyceride and reduced high‐density lipoprotein cholesterol levels. J Clin Lipidol. 2017;11(2):450‐458.e1. [DOI] [PubMed] [Google Scholar]
  • 111. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117‐2128. [DOI] [PubMed] [Google Scholar]
  • 112. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377:644‐657. [DOI] [PubMed] [Google Scholar]
  • 113. Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380:347‐357. [DOI] [PubMed] [Google Scholar]
  • 114. Bonaventura A, Carbone S, Dixon DL, Abbate A, Montecucco F. Pharmacologic strategies to reduce cardiovascular disease in type 2 diabetes mellitus: focus on SGLT‐2 inhibitors and GLP‐1 receptor agonists. J Intern Med. 2019;286:16‐31. [DOI] [PubMed] [Google Scholar]
  • 115. Sattar N, McLaren J, Kristensen SL, Preiss D, McMurray JJ. SGLT2 Inhibition and cardiovascular events: why did EMPA‐REG Outcomes surprise and what were the likely mechanisms? Diabetologia. 2016;59:1333‐1339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Scheen AJ. Beneficial effects of SGLT2 inhibitors on fatty liver in type 2 diabetes: a common comorbidity associated with severe complications. Diab Metab. 2019;45:213‐223. [DOI] [PubMed] [Google Scholar]
  • 117. Akuta N, Watanabe C, Kawamura Y, et al. Effects of a sodium‐glucose cotransporter 2 inhibitor in nonalcoholic fatty liver disease complicated by diabetes mellitus: preliminary prospective study based on serial liver biopsies. Hepatol Commun. 2017;1:46‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Cusi K, Bril F, Barb D, et al. Effect of canagliflozin treatment on hepatic triglyceride content and glucose metabolism in patients with type 2 diabetes. Diab Obes Metab. 2019;21:812‐821. [DOI] [PubMed] [Google Scholar]
  • 119. Inoue M, Hayashi A, Taguchi T, et al. Effects of canagliflozin on body composition and hepatic fat content in type 2 diabetes patients with non‐alcoholic fatty liver disease. J Diab Investig. 2019;10:1004‐1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Sattar N, Fitchett D, Hantel S, George JT, Zinman B. Empagliflozin is associated with improvements in liver enzymes potentially consistent with reductions in liver fat: results from randomised trials including the EMPA‐REG OUTCOME(R) trial. Diabetologia. 2018;61:2155‐2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Kuchay MS, Krishan S, Mishra SK, et al. Effect of empagliflozin on liver fat in patients with type 2 diabetes and nonalcoholic fatty liver disease: a randomized controlled trial (E‐LIFT Trial). Diab Care. 2018;41:1801‐1808. [DOI] [PubMed] [Google Scholar]
  • 122. Lai LL, Vethakkan SR, Nik Mustapha NR, Mahadeva S, Chan WK. Empagliflozin for the treatment of nonalcoholic steatohepatitis in patients with type 2 diabetes mellitus. Dig Dis Sci. 2020;65:623‐631. [DOI] [PubMed] [Google Scholar]
  • 123. Schaap FG, Trauner M, Jansen PL. Bile acid receptors as targets for drug development. Nat Rev Gastroenterol Hepatol. 2014;11:55‐67. [DOI] [PubMed] [Google Scholar]
  • 124. de Aguiar Vallim TQ, Tarling EJ, Edwards PA. Pleiotropic roles of bile acids in metabolism. Cell Metab. 2013;17:657‐669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Kast HR, Nguyen CM, Sinal CJ, et al. Farnesoid X‐activated receptor induces apolipoprotein C‐II transcription: a molecular mechanism linking plasma triglyceride levels to bile acids. Mol Endocrinol. 2001;15:1720‐1728. [DOI] [PubMed] [Google Scholar]
  • 126. Claudel T, Inoue Y, Barbier O, et al. Farnesoid X receptor agonists suppress hepatic apolipoprotein CIII expression. Gastroenterology. 2003;125:544‐555. [DOI] [PubMed] [Google Scholar]
  • 127. Fuchs CD, Schwabl P, Reiberger T, Trauner M. Liver capsule: FXR agonists against liver disease. Hepatology. 2016;64:1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Claudel T, Sturm E, Duez H, et al. Bile acid‐activated nuclear receptor FXR suppresses apolipoprotein A‐I transcription via a negative FXR response element. J Clin Invest. 2002;109:961‐971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Zhang Y, Yin L, Anderson J, et al. Identification of novel pathways that control farnesoid X receptor‐mediated hypocholesterolemia. J Biol Chem. 2010;285:3035‐3043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Ghosh Laskar M, Eriksson M, Rudling M, Angelin B. Treatment with the natural FXR agonist chenodeoxycholic acid reduces clearance of plasma LDL whilst decreasing circulating PCSK9, lipoprotein(a) and apolipoprotein C‐III. J Intern Med. 2017;281:575‐585. [DOI] [PubMed] [Google Scholar]
  • 131. Dong B, Singh AB, Guo GL, Young M, Liu J. Activation of FXR by obeticholic acid induces hepatic gene expression of SR‐BI through a novel mechanism of transcriptional synergy with the nuclear receptor LXR. Int J Mol Med. 2019;43:1927‐1938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Mudaliar S, Henry RR, Sanyal AJ, et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology. 2013;145:574‐582.e1. [DOI] [PubMed] [Google Scholar]
  • 133. Neuschwander‐Tetri BA, Loomba R, Sanyal AJ, et al. Farnesoid X nuclear receptor ligand obeticholic acid for non‐cirrhotic, non‐alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo‐controlled trial. Lancet. 2015;385:956‐965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Siddiqui MS, Van Natta ML, Connelly MA, et al. Impact of obeticholic acid on the lipoprotein profile in patients with non‐alcoholic steatohepatitis. J Hepatol. 2020;72:25‐33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Pencek R, Marmon T, Roth JD, Liberman A, Hooshmand‐Rad R, Young MA. Effects of obeticholic acid on lipoprotein metabolism in healthy volunteers. Diabetes Obes Metab. 2016;18:936‐940. [DOI] [PubMed] [Google Scholar]
  • 136. Pockros PJ, Fuchs M, Freilich B, et al. CONTROL: a randomized phase 2 study of obeticholic acid and atorvastatin on lipoproteins in nonalcoholic steatohepatitis patients. Liver Int. 2019;39:2082‐2093. [DOI] [PubMed] [Google Scholar]
  • 137. Younossi ZM, Ratziu V, Loomba R, et al. Obeticholic acid for the treatment of non‐alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo‐controlled phase 3 trial. Lancet. 2019;394:2184‐2196. [DOI] [PubMed] [Google Scholar]
  • 138. Zhou M, Wang X, Phung V, et al. Separating tumorigenicity from bile acid regulatory activity for endocrine hormone FGF19. Cancer Res. 2014;74:3306‐3316. [DOI] [PubMed] [Google Scholar]
  • 139. Harrison SA, Rinella ME, Abdelmalek MF, et al. NGM282 for treatment of non‐alcoholic steatohepatitis: a multicentre, randomised, double‐blind, placebo‐controlled, phase 2 trial. Lancet. 2018;391:1174‐1185. [DOI] [PubMed] [Google Scholar]
  • 140. Harrison SA, Rossi SJ, Paredes AH, et al. NGM282 improves liver fibrosis and histology in 12 weeks in patients with nonalcoholic steatohepatitis. Hepatology. 2020;71:1198‐1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Rinella ME, Trotter JF, Abdelmalek MF, et al. Rosuvastatin improves the FGF19 analogue NGM282‐associated lipid changes in patients with nonalcoholic steatohepatitis. J Hepatol. 2019;70:735‐744. [DOI] [PubMed] [Google Scholar]
  • 142. Charles ED, Neuschwander‐Tetri BA, Pablo Frias J, et al. Pegbelfermin (BMS‐986036), PEGylated FGF21, in patients with obesity and type 2 diabetes: results from a randomized phase 2 study. Obesity (Silver Spring). 2019;27:41‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Sanyal A, Charles ED, Neuschwander‐Tetri BA, et al. Pegbelfermin (BMS‐986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non‐alcoholic steatohepatitis: a randomised, double‐blind, placebo‐controlled, phase 2a trial. Lancet. 2019;392:2705‐2717. [DOI] [PubMed] [Google Scholar]
  • 144. Lawitz EJ, Coste A, Poordad F, et al. Acetyl‐CoA Carboxylase inhibitor GS‐0976 for 12 weeks reduces hepatic de novo lipogenesis and steatosis in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2018;16:1983‐1991.e3. [DOI] [PubMed] [Google Scholar]
  • 145. Loomba R, Kayali Z, Noureddin M, et al. GS‐0976 reduces hepatic steatosis and fibrosis markers in patients with nonalcoholic fatty liver disease. Gastroenterology. 2018;155:1463‐1473.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Mehal WZ. Acetyl‐Coenzyme A carboxylase inhibition delivers, as anticipated, for patients with nonalcoholic steatohepatitis. Gastroenterology. 2018;155:1304‐1306. [DOI] [PubMed] [Google Scholar]
  • 147. Kim C‐W, Addy C, Kusunoki J, et al. Acetyl CoA Carboxylase inhibition reduces hepatic steatosis but elevates plasma triglycerides in mice and humans: a bedside to bench investigation. Cell Metab. 2017;26:576. [DOI] [PubMed] [Google Scholar]
  • 148. Bergman A, Carvajal‐Gonzalez S, Tarabar S, Saxena AR, Esler WP, Amin N. Safety, tolerability, pharmacokinetics and pharmacodynamics of liver‐targeting ACC inhibitor (PF‐05521304) following single and multiple oral doses. J Hepatol. 2018;68:S582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Friedman SL, Ratziu V, Harrison SA, et al. A randomized, placebo‐controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology. 2018;67:1754‐1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Ratziu V, Sanyal A, Harrison SA, et al. Cenicriviroc treatment for adults with nonalcoholic steatohepatitis and fibrosis: final analysis of the phase 2b CENTAUR Study. Hepatology. 2020. 10.1002/hep.31108. PubMed ahead of print [DOI] [PubMed] [Google Scholar]
  • 151. Loomba R, Lawitz E, Mantry PS, et al. The ASK1 inhibitor selonsertib in patients with nonalcoholic steatohepatitis: a randomized, phase 2 trial. Hepatology. 2018;67:549‐559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Harrison SA, Abdelmalek MF, Caldwell S, et al. Simtuzumab is ineffective for patients with bridging fibrosis or compensated cirrhosis caused by nonalcoholic steatohepatitis. Gastroenterology. 2018;155:1140‐1153. [DOI] [PubMed] [Google Scholar]
  • 153. Shiffman M, Freilich B, Vuppalanchi R, et al. Randomised clinical trial: emricasan versus placebo significantly decreases ALT and caspase 3/7 activation in subjects with non‐alcoholic fatty liver disease. Aliment Pharmacol Ther. 2019;49:64‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Garcia‐Tsao G, Fuchs M, Shiffman M, et al. Emricasan (IDN‐6556) lowers portal pressure in patients with compensated cirrhosis and severe portal hypertension. Hepatology. 2019;69:717‐728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Frenette CT, Morelli G, Shiffman ML, et al. Emricasan improves liver function in patients with cirrhosis and high model for end‐stage liver disease scores compared with placebo. Clin Gastroenterol Hepatol. 2019;17:774‐783.e4. [DOI] [PubMed] [Google Scholar]
  • 156. Harrison SA, Goodman Z, Jabbar A, et al. A randomized, placebo‐controlled trial of emricasan in patients with NASH and F1–F3 fibrosis. J Hepatol. 2020;72:816‐827. [DOI] [PubMed] [Google Scholar]
  • 157. Kelly MJ, Pietranico‐Cole S, Larigan JD, et al. Discovery of 2‐[3,5‐dichloro‐4‐(5‐isopropyl‐6‐oxo‐1,6‐dihydropyridazin‐3‐yloxy)phenyl]‐3,5‐dio xo‐2,3,4,5‐tetrahydro[1,2,4]triazine‐6‐carbonitrile (MGL‐3196), a highly selective thyroid hormone receptor beta agonist in clinical trials for the treatment of dyslipidemia. J Med Chem. 2014;57:3912‐3923. [DOI] [PubMed] [Google Scholar]
  • 158. Taub R, Chiang E, Chabot‐Blanchet M, et al. Lipid lowering in healthy volunteers treated with multiple doses of MGL‐3196, a liver‐targeted thyroid hormone receptor‐beta agonist. Atherosclerosis. 2013;230:373‐380. [DOI] [PubMed] [Google Scholar]
  • 159. Harrison SA, Bashir MR, Guy CD, et al. Resmetirom (MGL‐3196) for the treatment of non‐alcoholic steatohepatitis: a multicentre, randomised, double‐blind, placebo‐controlled, phase 2 trial. Lancet. 2019;394:2012‐2024. [DOI] [PubMed] [Google Scholar]
  • 160. Safadi R, Konikoff FM, Mahamid M, et al. The fatty acid‐bile acid conjugate Aramchol reduces liver fat content in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2014;12:2085‐2091.e1. [DOI] [PubMed] [Google Scholar]
  • 161. Colca J. NASH (nonalcoholic steatohepatitis), diabetes, and macrovascular disease: multiple chronic conditions and a potential treatment at the metabolic root. Expert Opin Investig Drugs. 2020;29:191‐196. [DOI] [PubMed] [Google Scholar]
  • 162. McCommis KS, Hodges WT, Brunt EM, et al. Targeting the mitochondrial pyruvate carrier attenuates fibrosis in a mouse model of nonalcoholic steatohepatitis. Hepatology. 2017;65:1543‐1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Harrison SA, Alkhouri N, Davison BA, et al. Insulin sensitizer MSDC‐0602K in non‐alcoholic steatohepatitis: a randomized, double‐blind, placebo‐controlled phase IIb study. J Hepatol. 2019;72:613‐626. [DOI] [PubMed] [Google Scholar]

Articles from Alimentary Pharmacology & Therapeutics are provided here courtesy of Wiley

RESOURCES