Skip to main content

Lessons from PROMINENT and prospects for pemafibrate

Abstract

The neutral result of the PROMINENT trial has led to questions about the future for pemafibrate. This commentary discusses possible reasons for the lack of benefit observed in the trial. There were, however, indicators suggesting therapeutic potential in microvascular ischaemic complications associated with peripheral artery disease, with subsequent analysis showing reduction in the incidence of lower extremity ischaemic ulceration or gangrene. Reassurance about the safety of pemafibrate, together with emerging data from PROMINENT and experimental studies, also suggest benefit with pemafibrate in non-alcoholic fatty liver disease (alternatively referred to as metabolic dysfunction-associated steatotic liver disease) and microangiopathy associated with diabetes, which merit further study.

For decades, there has been controversy about the role of triglyceride (TG)-rich lipoproteins (TRLs) and their remnants-for which plasma TGs are a surrogate biomarker-in atherosclerotic cardiovascular disease (ASCVD). Observational data from prospective cohort studies showed an association between elevated TGs and ASCVD [1,2,3,4]. Additionally, in high-risk patients (including those with type 2 diabetes mellitus [T2DM]) and well controlled levels of low-density lipoprotein cholesterol (LDL-C), residual cardiovascular risk was higher in those with high versus lower TG levels [5,6,7]. Genetic studies have helped to disentangle vascular risk attributable to elevated TG levels from that attributable to low levels of high-density lipoprotein cholesterol (HDL-C) showing that elevated levels of TRLs were independently linked with increased coronary heart disease risk whereas low HDL-C was not [8, 9]. The advent of Mendelian randomization design further helped to establish elevated TRLs as a likely causal factor in ASCVD [3].

Demonstrating that lowering TGs with a conventional fibrate (a peroxisome proliferator-activated receptor alpha [PPARα] agonist) reduced cardiovascular events in high-risk patients has been challenging. While early trials suggested benefit from fibrate monotherapy [10, 11], later trials in patients receiving contemporary evidence-based therapy including a statin (unplanned drop-in of about 20% in the Fenofibrate Intervention and Event Lowering in Diabetes [FIELD] study and as planned in the Action to Control Cardiovascular Risk in Diabetes [ACCORD] study) were not positive [12, 13], in part confounded by issues with trial design and patient selection [13]. Post hoc analyses did, however, suggest that patients with elevated TG levels or mixed dyslipidaemia (high TG levels and low HDL-C]) derived benefit from fibrate therapy [14,15,16], although the limitations inherent in such post hoc analyses should be borne in mind.

Given that conventional fibrates have relatively weak PPARα agonistic potency, and the potential for reversible elevation in serum creatinine (with fenofibrate) [17, 18], as well as liver enzyme elevation especially in combination with a statin [19], alternative PPARα agonists were sought that may offer improved selectivity and potency for PPARα and better tolerability. The result was the selective peroxisome proliferator-activated modulator-α (SPPARMα) pemafibrate, which has shown greater potency for PPARα activation (> 2,500 times versus fenofibric acid, the active form of fenofibrate) and improved specificity for PPARα over conventional fibrates [20,21,22]. In phase II/III trials in Japanese and European patients with dyslipidaemia pemafibrate in combination with a statin was effective in lowering TG levels (by up to 50%) irrespective of pre-existing renal dysfunction [23, 24], with only minor changes in serum creatinine (less than with fenofibrate) [13, 24, 25], as well as reduction in liver enzymes, compared with increases in studies with fenofibrate [24, 25]. Based on this favourable risk/benefit profile, pemafibrate was selected to test the rationale that lowering TG levels reduces cardiovascular risk in the PROMINENT (Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes) trial [26].

The PROMINENT study

PROMINENT was a well-designed, large, randomized, placebo-controlled multinational study. The investigators were careful to ensure inclusion of an appropriate patient population with elevated TGs and low HDL-C to avoid criticism as in past fibrate trials [27]. Thus, PROMINENT planned to include ~ 10,000 high-risk patients with the mixed dyslipidaemia of T2DM, defined by TG levels of 200–499 mg/dL (2.26–5.64 mmol/L) and low HDL-C (≤ 40 mg/dL or 1.03 mmol/L), who were randomized to treatment with pemafibrate (0.2 mg twice daily) or matching placebo. The average expected follow-up period was 3.75 years. Patients were required to receive either moderate-to-high intensity statin therapy or meet specified LDL-C criteria. At baseline, median TGs were 271 mg/dL (3.1 mmol/L), median LDL-C was 78 mg/dL (~ 2.0 mmol/L) with almost all patients on a statin (69% on high-intensity statin treatment), and about one-third of the cohort were high-risk primary cardiovascular prevention patients.

Despite a well-argued rationale, supported by strong observational, genetic, and mechanistic evidence [3, 27], PROMINENT was terminated early after full recruitment (n = 10,497) on the advice of an independent data and safety monitoring board for reasons of futility. Despite lowering TGs by 26% (placebo-corrected), treatment with pemafibrate did not reduce major adverse cardiovascular events, a composite of nonfatal myocardial infarction, ischaemic stroke, coronary revascularization, or death from cardiovascular causes (hazard ratio 1.03, 95% confidence interval [CI], 0.91 to 1.15), with no apparent effect modification in any prespecified subgroup [26].

PROMINENT did provide further insights into the safety of pemafibrate. Overall, pemafibrate did not differ significantly from placebo with respect to the incidence of adverse events, including infections and musculoskeletal complications. There was a slight excess in the incidence of investigator-reported adverse renal events with pemafibrate versus placebo (1463 vs. 1347 patients, p = 0.004), with increases in both chronic kidney disease (CKD, 180 vs. 117 patients, p < 0.001) and acute kidney injury (160 vs. 106 patients, p = 0.001). Pemafibrate treatment was also associated with a small increase in median serum creatinine and decrease in estimated glomerular filtration rate, although both returned to baseline levels similar to that of the placebo group after discontinuation of treatment [26]. Similar findings have been reported with fenofibrate in the ACCORD and FIELD trials [12, 18], although another study suggested that long-term fenofibrate may delay impairment in renal function in T2DM patients [17]. The underlying mechanisms to explain the differences in impact on kidney function between pemafibrate and fenofibrate, as well as the drug-induced elevation of serum creatinine, are not fully understood.

Compared with placebo, the pemafibrate treatment group had an increase in the number of patients with investigator-reported venous thromboembolism events (71 vs. 35, p < 0.001), pulmonary embolism (40 vs. 19, p = 0.008), and deep-vein thrombosis 45 vs. 19, p = 0.001) [26]. Similarly, the FIELD trial reported a significant increase in pulmonary embolism with fenofibrate versus placebo (0.7%vs. 1.1%, p = 0.022) [12], in line with some previous reports [28, 29] suggesting possible caution in patients with a history of thromboembolic events. On the other hand, PROMINENT reported a decrease in investigator-reported non-alcoholic fatty liver disease (NAFLD) events [26], consistent with previous reports [30]. To overcome issues associated with the terms ‘non-alcoholic’ and ‘fatty’, the nomenclature metabolic dysfunction-associated steatotic liver disease (MASLD) as advocated by expert consensus [31] has been adopted in this review.

The reasons why pemafibrate failed to show significant benefit on cardiovascular events are uncertain. One factor may relate to the intensity of background statin therapy in PROMINENT. TG lowering with pemafibrate was less than that anticipated based on phase II/III clinical trials in patients treated with less intense statin regimens [21, 24, 26] (~ 45% to > 50%%) [23, 32]. Subgroup analyses of PROMINENT showed that the TG-lowering response with pemafibrate was attenuated with high-intensity statin treatment compared with less intense statin regimens (TG reduction by 24.6% versus 28.5% for patients on a moderate-intensity statin and 34.3% on low intensity or no statin). As statins decrease both LDL-C and TG in patients with hypertriglyceridaemia, with efficacy dependent on baseline levels of each lipid parameter [33], the TG-lowering effects of high-intensity statin treatment are likely to have impacted TG-lowering with pemafibrate in this subgroup. Indeed, while there was no evidence of heterogeneity in treatment effects when analysed according to statin intensity groups, among 2,636 patients on a moderate-intensity statin, incidence rates for the primary endpoint appeared to favour pemafibrate treatment (incidence rates 3.63 vs. 3.90 on placebo), although this was not statistically significant [26].

The influence of low baseline LDL-C levels in PROMINENT (median 78 mg/dL or ~ 2.0 mmol/L) also merits discussion [34]. Pemafibrate treatment reduced remnant cholesterol by 43.6% (25.6% after correction for placebo) [26], but in contrast to phase II/III trials [24], did not lower levels of small dense LDL (sdLDL), as estimated by Sampson’s Equation [35, 36]. A recent study in 1,508 T2DM patients (over 50% on a statin) and 670 controls showed that the slope of the regression curve between sdLDL and TG flattened at lower LDL-C levels, implying that rigorous control of LDL-C might minimize the inhibitory effect of pemafibrate on sdLDL production [37]. The underlying mechanisms are not defined, although it has been suggested that lower LDL-C concentrations may attenuate the involvement of TG in sdLDL generation, or alternatively, activation of hepatic TG lipase and promotion of sdLDL generation by pemafibrate treatment may counteract the decrease in sdLDL-C due to lower TG [36].

Some have suggested that the neutral results of PROMINENT may relate to the small increases in LDL-C and apolipoprotein B100 (apoB) with pemafibrate, although differences between the groups subsequently declined, and were similar after 30 days washout at study close [26]. While evidence supports apoB as a predictor of coronary heart disease (CHD) risk, this may be an oversimplification as it is an indirect measure of the total concentration of all apoB-containing particles which differ substantially by class. Although apoB is strongly correlated with LDL, it may fail to adequately capture variations in other apoB containing lipoproteins present at much lower concentration. The composition of LDL particles also merits consideration, with a recent prospective study showing a positive association between mean TG molecules per LDL particle and CHD risk but no associations with other lipid fractions or lipid particles [38]. Thus, LDL-TG rather than TG plasma concentration may represent a better biomarker of cardiovascular risk in the setting of hypertriglyceridaemia with well controlled LDL-C levels. Alternatively, absolute mass changes in remnant cholesterol, LDL-C, and apoB may be more relevant, with both higher total atherogenic cholesterol and higher apoB explaining the results of PROMINENT [39].

Increases in LDL-C levels are not unique to pemafibrate. REDUCE-IT (Reduction of Cardiovascular Events With Icosapent Ethyl-Intervention Trial) also showed a median 3.1% increase in LDL-C levels with high-dose icosapent ethyl (supplement to publication), but treatment was associated with 25% reduction in the primary composite endpoint (cardiovascular death, myocardial infarction, stroke, coronary revascularization, or unstable angina requiring hospitalization) [40]. Similarly, increases in LDL-C have been reported with the sodium-glucose cotransporter 2 (SGLT2) inhibitors, of the order of 4–5 mg/dL or ~ 10% increase from baseline with empagliflozin or canagliflozin [41,42,43,44]. In a meta-analysis of 60 randomized controlled trials with SGLT2 inhibitors, increases in total and LDL cholesterolwere similar (0.09 and 0.08 mmol/L), varying slightly by drug dose and ethnicity [45]. However, given the beneficial effects of this drug class in reducing cardiovascular death in T2DM patients [41,42,43], these modest lipid changes are unlikely to be clinically relevant. The mechanism(s) of this increase in LDL-C is uncertain, possibly involving both delayed LDL clearance from the circulation and increased plasma lipoprotein lipase activity [46]. While this finding aligns with the proposed explanation of enhanced lipolysis of TRLs increasing LDL-C levels with pemafibrate, it does not explain the neutral effect on cardiovascular outcomes observed in PROMINENT.

The above discussion focuses on potential factors influencing the efficacy of pemafibrate on cholesterol-related residual risk. However, evidence from an analysis of three major multinational trials that investigated the effect of TG-lowering on cardiovascular outcomes in high-risk patients receiving contemporary statin treatment, i.e., PROMINENT, REDUCE-IT, and STRENGTH (Long-Term Outcomes Study to Assess Statin Residual Risk with Epanova in High Cardiovascular Risk Patients with Hypertriglyceridemia) has underlined the importance of residual inflammatory risk. In this analysis, residual inflammatory risk (as measured by high-sensitivity C-reactive protein [hsCRP]) was a stronger predictor of cardiovascular outcomes than residual cholesterol risk [47]. This is relevant to the PROMINENT patient population, which was characterized by high inflammatory risk (more than half of patients had hsCRP levels ≥ 2 mg/L) and implies that intervention targeting both cholesterol and inflammatory components is needed to sufficiently impact residual cardiovascular risk inT2DM patients.

In summary, the take-home message from PROMINENT is that pemafibrate does not reduce cardiovascular events in T2DM patients with elevated TG and low HDL-C and well controlled LDL-C levels on intense statin therapy. Although there may be benefit when LDL-C is less tightly controlled, evidence that residual inflammatory risk is a stronger predictor of cardiovascular outcomes than cholesterol residual risk, especially in T2DM patients [47], argues for combination therapy targeting both components of risk.

Does PROMINENT suggest new prospects for pemafibrate?

The lack of a positive outcomes trial does not signal the end of the road for pemafibrate. Emerging data from PROMINENT suggest potential benefit with pemafibrate on complications of peripheral artery disease (PAD), as well as MASLD events (referred to as NAFLD in the trial), both of which have unmet clinical needs. In patients with T2DM, PAD tends to occur earlier and is often more severe and diffuse in than in nondiabetic patients, with the underlying pathophysiology driven by both progression of atherosclerotic disease and microvascular damage (mostly peripheral neuropathy) from chronic hyperglycaemia. In PROMINENT, pemafibrate treatment was associated with a clinically-relevant 13% relative reduction in new or worsening PAD events (hazard ratio 0.87, 95% CI 0.69–1.09) [26]. A secondary analysis [48] evaluated treatment effects on complications of PAD, i.e., incident lower ischaemic extremity ulceration or gangrene, defined as the new occurrence of lower extremity ulceration (leg or foot) or gangrene with diagnostic testing indicative of new or worsening obstructive PAD. Incidence rates (per 1000 person-years) for this composite outcome were 2.1 in the pemafibrate group vs. 3.4 in the placebo group, resulting in a 37% relative reduction in risk (hazard ratio, 0.63; 95% CI 0.41–0.95; p = 0.03), as well as 53% reduction in the risk of gangrene (hazard ratio 0.47, 95% CI 0.25–0.87; p = 0.01), and a lower incidence of ulcer (not statistically significant) [48]. These findings suggest novel therapeutic potential for pemafibrate in a setting with an unmet need for preventive therapies for distal small vessel ischaemic complications associated with PAD. Thus, while the combination of high-intensity statin therapy with pemafibrate may obscure the added advantage of pemafibrate in preventing macrovascular events, such as cardiovascular events, it does not seem to affect its efficacy in addressing microvascular complications as shown by this exploratory analysis of PROMINENT. This distinction in outcomes could be attributed to the lipid-independent mechanisms of action associated with pemafibrate.

For severe limb ischaemia caused by occlusive PAD, however, surgical treatment involving autologous vein grafts is the main therapeutic option, but graft failure is common within the first postoperative year [49, 50]. Insights into the cellular and molecular mechanisms that underlie vein graft failure could offer new therapeutic targets. In a recent study using a combination of proteomics, network analysis, and high-resolution ultrasonography in an experimental vein graft disease model, PPARα activation mediated by pemafibrate suppressed the development of vein graft failure and arteriovenous fistula lesions [51]. Although preliminary, these promising data merit further study and extrapolation to a clinical setting.

PROMINENT also hinted at possible benefit with pemafibrate in MASLD. Pemafibrate significantly reduced any hepatic adverse event (incidence per 100 person-years, 1.35 vs. 1.64, hazard ratio 0.83, 95% CI 0.69–0.99, p = 0.04), as well as investigator-reported MASLD events (referred to as NAFLD in the trial) (incidence per 100 person-years 0.95 vs. 1.22, hazard ratio 0.78, 95% CI 0.63–0.96, p = 0.02) [26]. Although absolute event numbers were small, these findings warrant further analysis. Other studies have shown that pemafibrate treatment decreased markers of liver dysfunction and non-invasive surrogates for liver fibrosis [52,53,54,55]. In a randomized placebo-controlled trial, treatment with pemafibrate reduced liver stiffness assessed by magnetic resonance elastography, although there was no significant reduction in liver fat [56]. With MASLD recognized as among the most prevalent chronic diseases globally, especially in low-to-middle income countries [57], these findings with pemafibrate are encouraging. An ongoing trial is investigating combination treatment with pemafibrate and an SGLT2 inhibitor in patients with non-alcohol related steatohepatitis and liver fibrosis, the more severe presentation of MASLD, with results anticipated in 2025 (ClinicalTrials.gov Identifier NCT05327127).

Novel insights from preclinical studies

There are also intriguing insights suggesting a possible beneficial role for pemafibrate in several settings with unmet clinical needs, notably diabetic eye disease [58]. One area of interest is in diabetic retinopathy, especially given evidence of benefit with fenofibrate in the FIELD and ACCORD trials, specifically in reduction in retinopathy progression, as shown by 31% reduction in first laser treatment in FIELD [59] and 40% reduction in progression, as assessed by the Early Treatment Diabetic Retinopathy Study Severity Scale in ACCORD [60]. Moreover, a meta-analysis of large cardiovascular trials showed that fenofibrate treatment reduced the need for retinal laser treatment by over 20% versus placebo [61]. Given a role of PPARα activation in this indication, and the higher specificity of pemafibrate for PPARα [20], suggests potential benefit with pemafibrate, although there are so far no data from PROMINENT. However, there are encouraging findings from preclinical models. Oral administration of pemafibrate inhibited retinal inflammation and retinal vascular leukostasis and leakage in a mouse model of carotid artery occlusion-induced ocular ischaemia [62], and in an experimental rat model of diabetic retinopathy improved systemic metabolism, protected retinal function [63], and improved inner retinal dysfunction [64]. Pemafibrate also suppressed retinal pathological neovascularization in a mouse model of oxygen-induced retinopathy [65], and suppressed choroidal neovascularization, an important cause of age-related macular degeneration [66]. These findings suggest a rationale for further study.

Beyond these effects, there are experimental data that suggest therapeutic potential for renal protective effects of pemafibrate in CKD patients. In an adenine-induced mouse model of CKD, administration of pemafibrate suppressed increases in plasma creatinine and blood urea nitrogen levels, decreased renal fibrosis and inhibited upregulation of inflammatory mediators such as interleukin-6 [67]. Clinical studies have demonstrated the efficacy and safety of pemafibrate in CKD patients with a wide range of renal insufficiency [68], and in a case study in patients with IgA nephropathy [69].An ongoing trial, PROFIT-CKD (Pemafibrate, open-label, Randomized cOntrolled study to evaluate the renal protective eFfect In hyperTriglyceridemia patients with Chronic Kidney Disease) aims to assess renal protective effects of pemafibrate in CKD patients, with change in urine protein/creatinine ratio over 12 months as study outcome [70]. Prevention of abdominal aortic aneurysm (AAA) rupture, which so far lacks effective preventive treatments beyond surgical approaches, is another area of interest with pemafibrate, given beneficial effects on inflammation and oxidative stress (Table 1).

Table 1 Potential of pemafibrate for therapeutic areas of unmet need in diabetes patients

Conclusion

The fibrates have had a chequered history in clinical outcomes studies, and PROMINENT has provided another twist to this story. While some have claimed that PROMINENT represents the ‘swan song of the fibrates’ [71], further data emerging from this trial, together with experimental studies, suggest otherwise. In particular, the possibility of favourable effects on diabetic-related microangiopathy suggests that the SPPARMα agonist pemafibrate may offer new opportunities to addressing the largely unmet clinical need of residual microvascular risk, specifically in PAD complications, as well as in MASLD (NAFLD) [72]. The answers await further research driven by the fine details emerging from PROMINENT.

Data availability

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AAA:

Abdominal aortic aneurysm

ACCORD:

Action to Control Cardiovascular Risk in Diabetes trial

ApoB:

Apolipoprotein B100

ASCVD:

Atherosclerotic cardiovascular disease

CHD:

Coronary heart disease

CI:

Confidence interval

CKD:

Chronic kidney disease

FIELD:

Fenofibrate Intervention in Event Lowering in Diabetes trial

HDL-C:

High-density lipoprotein cholesterol

hsCRP:

High-sensitivity C-reactive protein

LDL-C:

Low-density lipoprotein cholesterol

MASLD:

Metabolic dysfunction-associated steatotic liver disease

NAFLD:

Non-alcoholic fatty liver disease

NASH:

Non-alcoholic steatohepatitis

PAD:

Peripheral arterial disease

PROFIT-CKD:

Pemafibrate, open-label, Randomized cOntrolled study to evaluate the renal protective eFfect In hyperTriglyceridemia patients with Chronic Kidney Disease

PROMINENT:

Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes) trial

sdLDL-C:

Small dense LDL-C

SGLT2:

Sodium-glucose cotransporter 2

SPPARMα:

Selective peroxisome proliferator-activated modulator-α

T2DM:

Type 2 diabetes mellitus

TG:

Triglyceride

TRL:

Triglyceride-rich lipoprotein

References

  1. Austin MA. Plasma triglyceride and coronary heart disease. Arterioscler Thromb. 1991;11:2–14.

    Article  CAS  PubMed  Google Scholar 

  2. Nordestgaard BG, Benn M, Schnohr P, Tybjaerg-Hansen A. Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. JAMA. 2007;298:299–308.

    Article  CAS  PubMed  Google Scholar 

  3. Nordestgaard BG. Triglyceride-rich lipoproteins and atherosclerotic cardiovascular disease: new insights from epidemiology, genetics, and biology. Circ Res. 2016;118:547–63.

    Article  CAS  PubMed  Google Scholar 

  4. Sarwar N, Danesh J, Eiriksdottir G, Sigurdsson G, Wareham N, Bingham S, et al. Triglycerides and the risk of coronary heart disease: 10,158 incident cases among 262,525 participants in 29 western prospective studies. Circulation. 2007;115:450–8.

    Article  CAS  PubMed  Google Scholar 

  5. Raposeiras-Roubin S, Rossello X, Oliva B, Fernandez-Friera L, Mendiguren JM, Andres V, et al. Triglycerides and residual atherosclerotic risk. J Am Coll Cardiol. 2021;77:3031–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vallejo-Vaz AJ, Fayyad R, Boekholdt SM, Hovingh GK, Kastelein JJ, Melamed S, et al. Triglyceride-rich lipoprotein cholesterol and risk of cardiovascular events among patients receiving statin therapy in the TNT Trial. Circulation. 2018;138:770–81.

    Article  CAS  PubMed  Google Scholar 

  7. Nichols GA, Philip S, Reynolds K, Granowitz CB, Fazio S. Increased residual cardiovascular risk in patients with diabetes and high versus normal triglycerides despite statin-controlled LDL cholesterol. Diabetes Obes Metab. 2019;21:366–71.

    Article  CAS  PubMed  Google Scholar 

  8. Varbo A, Benn M, Tybjaerg-Hansen A, Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG. Remnant cholesterol as a causal risk factor for ischemic heart disease. J Am Coll Cardiol. 2013;61:427–36.

    Article  CAS  PubMed  Google Scholar 

  9. Do R, Willer CJ, Schmidt EM, Sengupta S, Gao C, Peloso GM, et al. Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat Genet. 2013;45:1345–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Frick MH, Elo O, Haapa K, Heinonen OP, Heinsalmi P, Helo P, et al. Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle-aged men with dyslipidemia. Safety of treatment, changes in risk factors, and incidence of coronary heart disease. N Engl J Med. 1987;317:1237–45.

    Article  CAS  PubMed  Google Scholar 

  11. Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Elam MB, et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med. 1999;341:410–8.

    Article  CAS  PubMed  Google Scholar 

  12. Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, et al. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. 2005;366:1849–61.

    Article  CAS  PubMed  Google Scholar 

  13. ACCORD Study Group; Ginsberg HN, Elam MB, Lovato LC, Crouse JR 3rd, Leiter LA, et al. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med. 2010;362:1563–74.

    Article  PubMed  Google Scholar 

  14. Scott R, O’Brien R, Fulcher G, Pardy C, D’Emden M, Tse D, et al. Effects of fenofibrate treatment on cardiovascular disease risk in 9,795 individuals with type 2 diabetes and various components of the metabolic syndrome: the Fenofibrate intervention and event lowering in diabetes (FIELD) study. Diabetes Care. 2009;32:493–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Elam MB, Ginsberg HN, Lovato LC, Corson M, Largay J, Leiter LA, et al. Association of fenofibrate therapy with long-term cardiovascular risk in statin-treated patients with type 2 diabetes. JAMA Cardiol. 2017;2:370–80.

    Article  PubMed  Google Scholar 

  16. Sacks FM, Carey VJ, Fruchart JC. Combination lipid therapy in type 2 diabetes. N Engl J Med. 2010;363:692–4. author reply 4–5.

    Article  CAS  PubMed  Google Scholar 

  17. Davis TM, Ting R, Best JD, Donoghoe MW, Drury PL, Sullivan DR, et al. Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate intervention and event lowering in diabetes (FIELD) study. Diabetologia. 2011;54:280–90.

    Article  CAS  PubMed  Google Scholar 

  18. Mychaleckyj JC, Craven T, Nayak U, Buse J, Crouse JR, Elam M, et al. Reversibility of fenofibrate therapy-induced renal function impairment in ACCORD type 2 diabetic participants. Diabetes Care. 2012;35:1008–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sobukawa Y, Hatta T, Funaki D, Nakatani E. Safety of combined statin and fibrate therapy: risks of liver injury and acute kidney injury in a cohort study from the Shizuoka Kokuho Database. Drugs Real World Outcomes. 2024. https://doi.org/10.1007/s40801-024-00426-1.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Fruchart JC, Santos RD, Aguilar-Salinas C, Aikawa M, Al Rasadi K, Amarenco P, et al. The selective peroxisome proliferator-activated receptor alpha modulator (SPPARMalpha) paradigm: conceptual framework and therapeutic potential: a consensus statement from the International Atherosclerosis Society (IAS) and the residual risk reduction Initiative (R3i) Foundation. Cardiovasc Diabetol. 2019;18:71.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Yamashita S, Masuda D, Matsuzawa Y. Pemafibrate, a new selective PPARalpha Modulator: drug concept and its clinical applications for dyslipidemia and metabolic diseases. Curr Atheroscler Rep. 2020;22:5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kim K, Ginsberg HN, Choi SH. New, novel lipid-lowering agents for reducing cardiovascular risk: beyond statins. Diabetes Metab J. 2022;46:517–32.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ginsberg HN, Hounslow NJ, Senko Y, Suganami H, Bogdanski P, Ceska R, et al. Efficacy and safety of K-877 (Pemafibrate), a selective PPARalpha Modulator, in European patients on statin therapy. Diabetes Care. 2022;45:898–908.

    Article  CAS  PubMed  Google Scholar 

  24. Yamashita S, Arai H, Yokote K, Araki E, Matsushita M, Nojima T, et al. Efficacy and safety of pemafibrate, a novel selective peroxisome proliferator-activated receptor alpha modulator (SPPARMalpha): pooled analysis of phase 2 and 3 studies in dyslipidemic patients with or without statin combination. Int J Mol Sci. 2019;20:5537.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Arai H, Yamashita S, Yokote K, Araki E, Suganami H, Ishibashi S, et al. Efficacy and safety of pemafibrate versus fenofibrate in patients with high triglyceride and low HDL cholesterol levels: a multicenter, placebo-controlled, double-blind, randomized trial. J Atheroscler Thromb. 2018;25:521–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Das Pradhan A, Glynn RJ, Fruchart JC, MacFadyen JG, Zaharris ES, Everett BM, et al. Triglyceride lowering with pemafibrate to reduce cardiovascular rRisk. N Engl J Med. 2022;387:1923–34.

    Article  PubMed  Google Scholar 

  27. Pradhan AD, Paynter NP, Everett BM, Glynn RJ, Amarenco P, Elam M, et al. Rationale and design of the Pemafibrate to Reduce Cardiovascular outcomes by reducing triglycerides in patients with diabetes (PROMINENT) study. Am Heart J. 2018;206:80–93.

    Article  CAS  PubMed  Google Scholar 

  28. Delluc A, Tromeur C, Le Moigne E, Nowak E, Mottier D, Le Gal G, et al. Lipid lowering drugs and the risk of recurrent venous thromboembolism. Thromb Res. 2012;130:859–63.

    Article  CAS  PubMed  Google Scholar 

  29. Humbert X, Dolladille C, Sassier M, Valnet-Rabier MB, Vial T, Guitton E, et al. [Fibrates and risk of venous thromboembolism: Case/no-case study in French pharmacovigilance database]. Therapie. 2017;72:677–82.

    Article  PubMed  Google Scholar 

  30. Yokote K, Yamashita S, Arai H, Araki E, Matsushita M, Nojima T, et al. Effects of pemafibrate on glucose metabolism markers and liver function tests in patients with hypertriglyceridemia: a pooled analysis of six phase 2 and phase 3 randomized double-blind placebo-controlled clinical trials. Cardiovasc Diabetol. 2021;20:96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Rinella ME, Lazarus JV, Ratziu V, Francque SM, Sanyal AJ, Kanwal F, et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. J Hepatol. 2023;79:1542–56.

    Article  CAS  PubMed  Google Scholar 

  32. Araki E, Yamashita S, Arai H, Yokote K, Satoh J, Inoguchi T, et al. Effects of pemafibrate, a novel selective PPARalpha modulator, on lipid and glucose metabolism in patients with type 2 diabetes and hypertriglyceridemia: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care. 2018;41:538–46.

    Article  CAS  PubMed  Google Scholar 

  33. Stein EA, Lane M, Laskarzewski P. Comparison of statins in hypertriglyceridemia. Am J Cardiol. 1998;81:B66–9.

    Article  Google Scholar 

  34. Packard CJ, Boren J, Taskinen MR. Causes and consequences of hypertriglyceridemia. Front Endocrinol (Lausanne). 2020;11:252.

    Article  PubMed  Google Scholar 

  35. Sampson M, Wolska A, Warnick R, Lucero D, Remaley AT. A new equation based on the standard lipid panel for calculating small dense low-density lipoprotein-cholesterol and its use as a risk-enhancer test. Clin Chem. 2021;67:987–97.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Hirano T. No change in small low-density lipoprotein cholesterol levels with pemafibrate might explain the negative results of the PROMINENT trial. J Diabetes Investig. 2023;14:630–1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hirano T, Ito Y. The influence of triglycerides on small dense low-density lipoprotein cholesterol levels is attenuated in low low-density lipoprotein-cholesterol range: implications for the negative results of the PROMINENT trial. J Diabetes Investig. 2023. https://doi.org/10.1111/jdi.14013.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Jin D, Trichia E, Islam N, Besevic J, Lewington S, Lacey B. Lipoprotein characteristics and incident coronary heart disease: prospective cohort of nearly 90 000 individuals in UK Biobank. J Am Heart Assoc. 2023;12:e029552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Doi T, Langsted A, Nordestgaard BG. Remnant cholesterol, LDL cholesterol, and apoB absolute mass changes explain results of the PROMINENT trial. Atherosclerosis. 2024;393:117556.

    Article  CAS  PubMed  Google Scholar 

  40. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl J Med. 2019;380:11–22.

    Article  CAS  PubMed  Google Scholar 

  41. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–28.

    Article  CAS  PubMed  Google Scholar 

  42. Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377:644–57.

    Article  CAS  PubMed  Google Scholar 

  43. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380:347–57.

    Article  CAS  PubMed  Google Scholar 

  44. Schernthaner G, Gross JL, Rosenstock J, Guarisco M, Fu M, Yee J, et al. Canagliflozin compared with sitagliptin for patients with type 2 diabetes who do not have adequate glycemic control with metformin plus sulfonylurea: a 52-week randomized trial. Diabetes Care. 2013;36:2508–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bechmann LE, Emanuelsson F, Nordestgaard BG, Benn M. SGLT2-inhibition increases total, LDL, and HDL cholesterol and lowers triglycerides: Meta-analyses of 60 randomized trials, overall and by dose, ethnicity, and drug type. Atherosclerosis. 2023:117236.

  46. Basu D, Huggins LA, Scerbo D, Obunike J, Mullick AE, Rothenberg PL, et al. Mechanism of increased LDL (low-Density lipoprotein) and decreased triglycerides with SGLT2 (sodium-Glucose cotransporter 2) inhibition. Arterioscler Thromb Vasc Biol. 2018;38:2207–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ridker PM, Bhatt DL, Pradhan AD, Glynn RJ, MacFadyen JG, Nissen SE, et al. Inflammation and cholesterol as predictors of cardiovascular events among patients receiving statin therapy: a collaborative analysis of three randomised trials. Lancet. 2023;401:1293–301.

    Article  CAS  PubMed  Google Scholar 

  48. Marino L, Everett BM, Aday AY, Visseren FL, MacFadyen J, Zaharri E, et al. Pemafibrate reduces incidence of lower extremity ischemia ulcer and gangrene: evidence from PROMINENT. Abstract 23102. Circulation. 2023;148:e282–317.

    Google Scholar 

  49. Reifsnyder T, Arhuidese IJ, Hicks CW, Obeid T, Massada KE, Khaled A, et al. Contemporary outcomes for open infrainguinal bypass in the endovascular era. Ann Vasc Surg. 2016;30:52–8.

    Article  PubMed  Google Scholar 

  50. Conte MS, Bandyk DF, Clowes AW, Moneta GL, Seely L, Lorenz TJ, et al. Results of PREVENT III: a multicenter, randomized trial of edifoligide for the prevention of vein graft failure in lower extremity bypass surgery. J Vasc Surg. 2006;43:742–51. discussion 51.

    Article  PubMed  Google Scholar 

  51. Decano JL, Singh SA, Gasparotto Bueno C, Ho Lee L, Halu A, Chelvanambi S, et al. Systems approach to discovery of therapeutic targets for vein graft disease: PPARalpha pivotally regulates metabolism, activation, and heterogeneity of macrophages and lesion development. Circulation. 2021;143:2454–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Morishita A, Oura K, Takuma K, Nakahara M, Tadokoro T, Fujita K, et al. Pemafibrate improves liver dysfunction and non-invasive surrogates for liver fibrosis in patients with non-alcoholic fatty liver disease with hypertriglyceridemia: a multicenter study. Hepatol Int. 2023;17:606–14.

    Article  PubMed  Google Scholar 

  53. Ikeda S, Sugihara T, Kihara T, Matsuki Y, Nagahara T, Takata T, et al. Pemafibrate ameliorates liver dysfunction and fatty liver in patients with non-alcoholic fatty liver disease with hypertriglyceridemia: a retrospective study with the outcome after a mid-term follow-up. Diagnostics (Basel). 2021;11. https://doi.org/10.3390/diagnostics11122316.

  54. Hatanaka T, Kakizaki S, Saito N, Nakano Y, Nakano S, Hazama Y, et al. Impact of pemafibrate in patients with hypertriglyceridemia and metabolic dysfunction-associated fatty liver disease pathologically diagnosed with non-alcoholic steatohepatitis: a retrospective, single-arm study. Intern Med. 2021;60:2167–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hatanaka T, Kosone T, Saito N, Takakusagi S, Tojima H, Naganuma A, et al. Effect of 48-week pemafibrate on non-alcoholic fatty liver disease with hypertriglyceridemia, as evaluated by the FibroScan-aspartate aminotransferase score. JGH Open. 2021;5:1183–9.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Nakajima A, Eguchi Y, Yoneda M, Imajo K, Tamaki N, Suganami H, et al. Randomised clinical trial: Pemafibrate, a novel selective peroxisome proliferator-activated receptor alpha modulator (SPPARMalpha), versus placebo in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2021;54:1263–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Jiang W, Mao X, Liu Z, Zhang T, Jin L, Chen X. Global burden of nonalcoholic fatty liver disease, 1990 to 2019: findings from the global burden of Disease Study 2019. J Clin Gastroenterol. 2023;57:631–9.

    Article  CAS  PubMed  Google Scholar 

  58. Lee D, Tomita Y, Negishi K, Kurihara T. Therapeutic roles of PPARalpha activation in ocular ischemic diseases. Histol Histopathol. 2023;38:391–401.

    CAS  PubMed  Google Scholar 

  59. Keech AC, Mitchell P, Summanen PA, O’Day J, Davis TM, Moffitt MS, et al. Effect of fenofibrate on the need for laser treatment for diabetic retinopathy (FIELD study): a randomised controlled trial. Lancet. 2007;370:1687–97.

    Article  CAS  PubMed  Google Scholar 

  60. ACCORD Study Group; ACCORD Eye Study Group; Chew EY, Ambrosius WT, Davis MD, Danis RP, et al. Effects of medical therapies on retinopathy progression in type 2 diabetes. N Engl J Med. 2010;363:233–44.

    Article  Google Scholar 

  61. Preiss D, Spata E, Holman RR, Coleman RL, Lovato L, Ginsberg HN, et al. Effect of fenofibrate therapy on laser treatment for diabetic retinopathy: a meta-analysis of randomized controlled trials. Diabetes Care. 2022;45:e1–2.

    Article  PubMed  Google Scholar 

  62. Shiono A, Sasaki H, Sekine R, Abe Y, Matsumura Y, Inagaki T, et al. PPARalpha activation directly upregulates thrombomodulin in the diabetic retina. Sci Rep. 2020;10:10837.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Tomita Y, Lee D, Miwa Y, Jiang X, Ohta M, Tsubota K, et al. Pemafibrate protects against retinal dysfunction in a murine model of diabetic retinopathy. Int J Mol Sci. 2020;21. https://doi.org/10.3390/ijms21176243.

  64. Lee D, Tomita Y, Jeong H, Miwa Y, Tsubota K, Negishi K, et al. Pemafibrate prevents retinal dysfunction in a mouse model of unilateral common carotid artery occlusion. Int J Mol Sci. 2021;22. https://doi.org/10.3390/ijms22179408.

  65. Tomita Y, Ozawa N, Miwa Y, Ishida A, Ohta M, Tsubota K, et al. Pemafibrate prevents retinal pathological neovascularization by increasing FGF21 level in a murine oxygen-induced retinopathy model. Int J Mol Sci. 2019;20. https://doi.org/10.3390/ijms20235878.

  66. Lee D, Nakai A, Miwa Y, Negishi K, Tomita Y, Kurihara T. Pemafibrate prevents choroidal neovascularization in a mouse model of neovascular age-related macular degeneration. PeerJ. 2023;11:e14611.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Horinouchi Y, Murashima Y, Yamada Y, Yoshioka S, Fukushima K, Kure T, et al. Pemafibrate inhibited renal dysfunction and fibrosis in a mouse model of adenine-induced chronic kidney disease. Life Sci. 2023;321:121590.

    Article  CAS  PubMed  Google Scholar 

  68. Yokote K, Yamashita S, Arai H, Araki E, Suganami H, Ishibashi S, et al. Long-term efficacy and safety of pemafibrate, a novel selective peroxisome proliferator-activated receptor-alpha modulator (SPPARMalpha), in dyslipidemic patients with renal impairment. Int J Mol Sci. 2019;20. https://doi.org/10.3390/ijms20030706.

  69. Tanaka A, Nakamura T, Sato E, Chihara A, Node K. Effect of pemafibrate, a novel selective peroxisome proliferator-activated receptor-alpha modulator (SPPARMalpha), on urinary protein excretion in IgA nephropathy with hypertriglyceridemia. CEN Case Rep. 2020;9:141–6.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Seki M, Nakano T, Tanaka S, Matsukuma Y, Funakoshi K, Ohkuma T, et al. Design and methods of an open-label, randomized controlled trial to evaluate the effect of pemafibrate on proteinuria in CKD patients (PROFIT-CKD). Clin Exp Nephrol. 2023;27:358–64.

    Article  CAS  PubMed  Google Scholar 

  71. van Sloten TT, Martens F, Visseren FLJ. [The PROMINENT trial: swan song of the fibrates]. Ned Tijdschr Geneeskd. 2023;167:D7528.

    PubMed  Google Scholar 

  72. Avogaro A, Fadini GP. Microvascular complications in diabetes: a growing concern for cardiologists. Int J Cardiol. 2019;291:29–35.

    Article  PubMed  Google Scholar 

  73. Amioka N, Miyoshi T, Yonezawa T, Kondo M, Akagi S, Yoshida M, et al. Pemafibrate prevents rupture of angiotensin II-induced abdominal aortic aneurysms. Front Cardiovasc Med. 2022;9:904215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The PROMINENT study was funded by Kowa. This manuscript was prepared independent of funding.

Author information

Authors and Affiliations

Authors

Contributions

J-C.F, J.F-N, and M.P.H. drafted the main manuscript text. All authors reviewed and approved the final manuscript.

Corresponding authors

Correspondence to Jean-Charles Fruchart or Jamila Fruchart-Najib.

Ethics declarations

Ethics approval and consent to participate

Not applicable for this comment. All patients in the PROMINENT study gave written informed consent. The study was approved at participating centres by the responsible institutional review board or ethics committee, as applicable, and by regulatory authorities in the 24 countries where the trial was conducted.

Consent for publication

Not applicable.

Competing interests

J-CF and JF-N report personal fees for consultancy from Kowa Company, Ltd. SY reports grants and personal fees from Kowa Company, Ltd., Otsuka Pharmaceutical Co., Ltd., Shionogi & Co., Ltd., Bayer Yakuhin, Ltd., MSD K.K., Takeda Pharmaceutical Company, Ltd., Sanwa Kagaku Kenkyusho Co., Ltd., Astellas Pharma Inc., Daiichi-Sankyo Company, Ltd., Astra Zeneka K.K., Kaken Pharmaceutical Co., Ltd., grants from Nippon Boehringer Ingelheim Co., Ltd., National Institute of Biomedical Innovation, Kyowa Medex Co., Ltd., Mochida Pharmaceutical Company, Ltd., Hayashibara Co., Ltd., Teijin Pharma Limited and Kissei; and personal fees from Ono Pharmaceutical Company, Ltd., Skylight Biotec, Inc., Pfizer, Astellas Amgen, Sanofi, and Aegerion In addition, M Yamashita has a patent PCT/JP2016/074402 (Assisting Method for the Diagnosis of Type III Hyperlipidemia) pending to Fujirebio & Osaka University, a patent PCT/JP2017/038766 (Method for Selecting Subject Needing Treatment for Dyslipidemia and Reagent for Such Selection) pending to Osaka University & Kyowa Medex Co., Ltd., and a patent PCT/JP2017/038715 (Method for Measuring Oxidized High-Density Lipoprotein) pending to Osaka University & Kyowa Medex Co.PL reports a research grant from Novartis and honoraria as a scientific advisory board member for Dalcor Pharmaceuticals, and provides unpaid consultancy for Amgen, AstraZeneca, Ionis Pharmaceuticals, Kowa Pharmaceuticals, Pfizer, Sanofi-Regeneron, XBiotech Inc., Corvidia Therapeutics, IFM Therapeutics, Olatec Therapeutics, Medimmune and Esperion Therapeutics.KY reports grants and personal fees from Kowa Pharmaceutical Co, Astellas, AstraZeneca, MSD, Sanofi, Takeda, Pfizer, Mochida, and personal fees from Kowa Company, Astellas-Amgen Biopharm, and Bayer.TK is the recipient of a research grant from Kowa Company. YT reports research grants from the Alcon Research Institute, Manpei Suzuki Diabetic Foundation, Oguchi Foundation, and Kowa Life Science Foundation. PMR reports grants from Kowa, Inc, Novartis, and Pfizer. AZ reports honoraria for lectures from Abbott, Amgen, Sanofi and Mylan. MPH reports no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it.The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fruchart, JC., Fruchart-Najib, J., Yamashita, S. et al. Lessons from PROMINENT and prospects for pemafibrate. Cardiovasc Diabetol 23, 279 (2024). https://doi.org/10.1186/s12933-024-02305-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12933-024-02305-z

Keywords