Abstract

Background

Triglycerides, cholesterol, and their metabolism are linked due to shared packaging and transport within circulating lipoprotein particles. While a case for a causal role of cholesterol-carrying low-density lipoproteins (LDLs) in atherosclerosis is well made, the body of scientific evidence for a causal role of triglyceride-rich lipoproteins (TRLs) is rapidly growing, with multiple lines of evidence (old and new) providing robust support.

Content

This review will discuss current perspectives and accumulated evidence that an overabundance of remnant lipoproteins stemming from intravascular remodeling of nascent TRLs—chylomicrons and very low-density lipoproteins (VLDL)—results in a proatherogenic milieu that augments cardiovascular risk. Basic mechanisms of TRL metabolism and clearance will be summarized, assay methods reviewed, and pivotal clinical studies highlighted.

Summary

Remnant lipoproteins are rendered highly atherogenic by their high cholesterol content, altered apolipoprotein composition, and physicochemical properties. The aggregate findings from multiple lines of evidence suggest that TRL remnants play a central role in residual cardiovascular risk.

Introduction

Triglycerides, cholesterol, and their metabolism are inextricably linked due to shared packaging and transport within circulating lipoprotein particles. While a causal relationship for cholesterol-carrying low-density lipoproteins (LDLs) in atherosclerosis is indisputable, the body of scientific evidence for a causal role of triglyceride-rich lipoproteins (TRLs) is rapidly growing, with multiple lines of evidence now adding robust support. Overabundance of these TRLs—chylomicrons and very low-density lipoproteins (VLDL)—results from increased production or impaired clearance and is clinically evident as hypertriglyceridemia conveniently detected upon routine lipid testing. The concept that hypertriglyceridemia signals clinically relevant abnormalities in lipid metabolism and a proatherogenic milieu has not been widely embraced by the general medical community. However, building on observations made decades ago, novel kinetic investigations, advances in laboratory methods for lipoprotein quantification, epidemiologic observations, genomic science, and clinical trial data have refocused attention on a potential central role of TRLs in residual cardiovascular risk, i.e., that risk mediated through pathways not addressed by our current LDL management algorithms. To understand these important developments, this narrative review will discuss the relevant biologic and clinical observations pertaining to TRLs and TRL ‘remnants’ and examine their role as therapeutic targets for cardiovascular disease prevention.

Contemporary Estimates of Hypertriglyceridemia Prevalence

The presence of hypertriglyceridemia varies greatly by country and region with limited contemporary comparative data. Nonetheless, a general prevalence estimate of 10% among adult populations has been suggested (1). In the US, over the period 2007–2014, this rate is about 1 in 4 having triglyceride (TG) concentrations >150 mg/dL (1.7 mmol/L) with important ethnic and racial differences, Mexican Americans having the highest prevalence, followed by non-Hispanic Whites and non-Hispanic Blacks (2, 3). Longitudinal data have shown a decline in prevalence that can be linked to increasing use of lipid lowering therapy, in particular, statins that have modest TG-lowering effects (4). Nonetheless, among high risk groups such as persons with diabetes or prior cardiovascular disease, these generally favorable trend data belie an important problem. In US adults with diabetes and LDL-C < 70 mg/dL (<1.8 mmol/L) on statin therapy, about 30% had residual hypertriglyceridemia (TG > 150 mg/dL; >1.7 mmol/L) (5). In a large population-based study (Ontario, Canada) of patients with established cardiovascular disease and controlled LDL-C, 1 in 4 had increased TGs (TG 135–499 mg/dL; 1.52–5.63 mmol/L) (6). While these data reveal opportunities for risk mitigation, the focus on hypertriglyceridemia diverts attention from the underlying physiologic abnormality, a shift in distribution of cholesterol trafficking lipoproteins, that is likely the more credible therapeutic target.

Basic Principles of Triglyceride-Rich Remnant Metabolism

In humans, cholesterol accumulation and only modest triglyceride accumulation characterizes atherosclerotic lesions. Thus, most conventional explanations for atherosclerosis susceptibility do not clearly implicate hypertriglyceridemia. Hypertriglyceridemia fuels TRL production, accelerates remodeling of cholesterol-carrying lipoproteins (7–9), and potentiates inflammatory pathways (10–12) but is unlikely to directly instigate atherosclerosis. TGs are primarily transported by TRLs comprising chylomicrons secreted by intestinal enterocytes, VLDL particles synthesized in the liver and more recently discovered to have intestinal origins (13), and their metabolic derivatives or ‘remnants’. Intravascular lipolysis of newly secreted TRLs gives rise, in the case of chylomicrons, to chylomicron remnants and in the case of VLDL to a spectrum of highly modified particles including remnants within the VLDL density range, intermediate-density lipoproteins (IDLs), and LDLs. These smaller highly modified TRL remnant particles are increasingly believed to be causative agents in atherosclerosis.

The metabolic fate of TRLs (degree of remodeling) and plasma abundance depends on the size of the TG pool, as well as hormonal, nutritional, and genetic traits in a given individual. Their atherogenic burden depends on structural properties that determine plasma residence time, size-related rates of arterial influx, residual cholesterol content, subendothelial retention time, and capacity to elicit foam cell formation and promote plaque formation. Basic knowledge of the origin of these lipoproteins and TRL metabolism pathways (topics discussed below) is essential to understanding their role in atherosclerosis. For a more detailed description, the reader is referred to several recent comprehensive reviews (14–16).

Chylomicrons (diameter >75 nm) are the largest and lowest-density lipoproteins synthesized and secreted from the small intestine after ingestion of dietary fat. Following release from the intestinal mucosa into the lymphatic system, newly secreted (nascent) chylomicrons enter the systemic circulation via the thoracic duct. Chylomicron TGs undergo rapid hydrolysis in a process much faster than VLDL TGs (17–19). Resulting remnants retain almost all the cholesterol contained in their precursors and acquire several molecules of apoE via exchange with high-density lipoprotein (HDL) in plasma or during transit within hepatic tissue (20, 21). Chylomicron remnants are ultimately cleared by binding the LDL receptor or LDL receptor protein-1 (LRP-1) via apoE interactions and via the heparan sulfate proteoglycan pathway in the liver (22–24). Within hepatocytes, cholesterol esters are digested by lysosomal enzymes, which release unesterified cholesterol that becomes available for repackaging in VLDL.

VLDLs are synthesized primarily in the liver and like chylomicrons fulfill a lipid transport function. Two major physically distinct species of VLDL exist: larger VLDL1 (50–80 nm diameter, circa 70% TG mass) and smaller VLDL2 (30–50 nm diameter, circa 30% TG mass). As a function of TG availability, hepatocytes vary the amount of lipid loaded onto lipoprotein particles in the endoplasmic reticulum and can assemble and secrete newly synthesized particles that span the entire size range (VLDL1-VLDL2-IDL-LDL). At normal TG concentrations (<100 mg/dL; <1.2 mmol/L), VLDL1 and VLDL2 circulate in approximately equal proportions. A number of factors influence VLDL1 secretion including hepatic TG stores, insulin resistance, chylomicron remnant clearance pathways, and regulatory pathways for de novo lipogenesis (25, 26). Synthesis of VLDL2 is more influenced by endogenous cholesterol synthesis, is less dependent on increased TGs and is less dominant in hypertriglyeridemia (27–30). Once released, progressive delipidation (removal of TGs) of both VLDLs and intravascular remodeling yields remnant particles in the IDL and LDL density intervals. Compared to chylomicron remnants, which are largely cleared by the liver, 25–75% of VLDL remnants do not undergo hepatic uptake but are rather converted to LDL (14).

Fisher first proposed (31), and Packard and colleagues confirmed (32), that ‘metabolic channeling’ within the VLDL-LDL delipidation cascade occurs such that VLDL1 and VLDL2 have differing rates of conversion to IDL and LDL. LDL derived from VLDL1 was also shown to have a slower clearance rate and longer duration to decay. It has been postulated that these slowly metabolized LDL particles of VLDL1 lineage are a major source of small dense LDL particles (7, 33), thereby providing a critical link between hypertriglyceridemia and other markers of atherogenic dyslipidemia. These observations also suggest that lipoprotein pedigree, whether newly secreted or the result of TRL catabolism, determines physicochemical properties and metabolic fate.

Remnants as Biologic Drivers of Atherosclerosis

Newly secreted chylomicrons, VLDLs, IDLs, and LDLs although structurally distinct from remnants may occupy the same density and size range as TRL remnants that have undergone extensive intravascular remodeling (Figure 1). Thus, size alone cannot be the determinant of remnant atherogenicity. The actions of 2 enzymes, lipoprotein lipase (LPL) and hepatic lipase (HL), and a specific transfer protein, cholesterol ester transfer protein (CETP), induce major structural changes that distinguish remnants from nonremnant lipoproteins and may enhance atherogenicity. Through rapid LPL-mediated (and to a lesser degree HL-mediated) removal of TGs and CETP-mediated exchange of TGs for cholesterol from LDL and HDL, remnant particles contain disproportionately more cholesterol than nascent chylomicrons or VLDL (34). Compared with their parent TRLs, with progressive catabolism, remnants also lose their complement of C apolipoproteins (including apoCIII) and become enriched in apoE (14). These changes appear due to reductions in volume and surface area that preferentially drive the movement of apoCs to HDL and the direct addition of apoE (35–37). However, hypertriglyceridemia alters this redistribution (38). Normal transfer of apoCIII to HDL is disrupted, and remnant apoCIII stores are retained. Hypertriglyceridemia also drives increased hepatic production of VLDL associated apoCIII (39). The net result is that apoCIII accumulates in VLDL, where it plays an inhibitory role on LPL and hepatic VLDL uptake ultimately leading to longer plasma residence time of these particles.

While chylomicrons and large VLDLs are prohibited from transcytosis by virtue of their size, smaller chylomicron and VLDL remnants can and do penetrate the artery wall (40–42). Using fluorescently labeled VLDL, Rutledge and colleagues showed that lipolysis of VLDL enhanced by more than 2-fold its ability to cross the endothelium and accumulate in the subendothelial space (43, 44). Upon entry, these particles become trapped by subendothelial proteoglycans and are susceptible to engulfment by vascular macrophages. As in the liver, apoE is a major determinant of macrophage remnant uptake by receptor-mediated endocytosis via binding to the LDL and LRP-1 receptors (16, 45, 46). In a process akin to LDL, ensuing lipid loading and foam cell formation is thought to elicit a similar cascade of events leading to plaque formation (34). However, the relative contribution of TRLs to this process requires further definition.

Even though remnant particles remain more triglyceride- than cholesterol-rich, their large size means 5–20 times more cholesterol may deposit per particle than LDL (47). Recent data from over 15 000 healthy individuals also indicate that the postprandial state may influence the cholesterol concentrations of TRL particles more so than LDL particles (48). Whether TRL pedigree (remnant vs nonremnant origin) impacts cholesterol delivery has not been well studied. However, TRL modification and longer residence in circulation can result in greater exposure to the vessel wall (44, 49). Unlike native LDL, which may exit the subendothelial space almost as rapidly as it enters, remnants efflux very slowly compared to their rate of entry with increased opportunity for internalization by macrophages and foam cell formation (44, 50). Also distinct from LDL, which is constitutively deficient in apoE and requires oxidation for uptake, remnants do not require oxidation to facilitate accumulation in macrophages due to their apoE enrichment . Another consequence of remnant retention is local TG lipolysis. Like adipocyte and myocytes, atherosclerotic plaque macrophages have the capacity to produces substantial quantities of LPL (51–53). LPL-mediated liberation of free fatty acids from accumulated remnants can induce production of proinflammatory intermediaries (cytokines, interleukins, and adhesion molecules) that accelerate the recruitment of leukocytes to areas of inflammation. Botham and Wheeler-Jones (54) provide an excellent mechanistic review of TRLs in vascular homeostasis. These concepts and experimental findings are consistent with the seminal Zilversmit hypothesis (55, 56), which proposed that TRLs, via remnant infiltration, act additively with LDL to direct atheroma propagation.

Remnant TRL Quantification: To Calculate or to Measure, and How?

As cholesterol trafficking by remnant TRLs is the favored mechanism in atherogenesis, optimal methods to measure remnant TRL cholesterol (TRL-C) are required for clinical studies and, ultimately, clinical practice. Accurate measurement and characterization of remnant cholesterol, however, has proven to be challenging for 2 major reasons: (1) due to their rapid and ongoing catabolism, TRL remnant size, quantity, and composition are highly dynamic; and (2) despite their reduced size and triglyceride content, TRL remnants are difficult to differentiate from their precursors (nonremnant lipoproteins). While no universally accepted definition of TRL remnants currently exists, one that is generally used as a goal for detection is postlipolytic partially TG-depleted particles derived from chylomicrons and VLDL that are relatively enriched in cholesteryl esters and apoE. It is also acknowledged that nomenclature in laboratory methods is highly variable. In this review, we use ‘TRL-C’ to reflect cholesterol content of all TRLs and specify ‘remnant TRL-C’ when referencing methods to isolate remnant cholesterol.

The simplest way to estimate TRL-C is by rearrangement of the Friedewald equation, which was initially developed to estimate LDL-C (i.e., LDL-C = total cholesterol − HDL cholesterol − triglyceride/5) (57). Using this equation, TRL-C (or VLDL-C as initially described) is estimated as 1/5 of triglyceride concentration in mg/dL (or 1/2.2 in mmol/L). However, by using a fixed ratio, the predicted value is equivalent to TG/5 and Friedewald estimated TRL-C does not add clinical information beyond TG concentrations alone. Consensus recommendations from the European Atherosclerosis Society and European Federation of Clinical Chemistry and Laboratory Medicine now recommend that a direct LDL-C measurement be used when the Friedewald formula is used for TRL-C estimation (58). Some have used this equation to estimate remnant TRL-C under the assumption that because of rapid catabolism, all circulating cholesterol in the VLDL component of the formula can be considered remnant cholesterol. This has not been well defined.

Apart from calculation, several direct methods are available by which TRL remnants are identified, isolated, and quantified on the basis of their density, charge, size, specific lipid components, apo composition, or apo specificity (59). Major methods that will be discussed include preparative ultrafiltration, immunoseparation, nuclear magnetic resonance (NMR), and direct homogenous assays, each of which have been shown in clinical studies to identify individuals with increased cardiovascular risk.

Preparative ultracentrifugation is considered the reference standard for lipoprotein separation and relies on relative flotation based on particle density. As remnant TRLs are smaller and denser than their precursors, those derived from VLDL are mainly found in the IDL range (d = 1.006–1.019 g/mL) with some present in the VLDL range (d = 0.930–1.006 g/mL). Chylomicron remnants are found in the chylomicron, VLDL or IDL range. Figure 1 illustrates this overlap and limitation, specifically, ultracentrifugation cannot differentiate TRL remnants from other nonremnant lipoprotein particles of similar density. In addition, most traditional ultracentrifugation methods are time intensive, taking several hours to days for completion. The vertical auto profile (VAP) technique is a modified approach that uses a vertical rotator for lipoprotein separation leveraging the shorter horizontal axis of the centrifuge tube rather than the longer vertical axis used in preparative ultracentrifugation (60). It suffers similar limitations in that remnants span multiple density zones and the method requires a special analyzer that is not available in most clinical laboratories.

Depiction of remnant particles by major apolipoproteins, size, nomenclature, and density. Abbreviations: CM, chylomicron; VLDL, very low-density lipoprotein; IDL, intermediate-density lipoprotein; LDL, low-density lipoprotein; HDL, high-density lipoprotein; LB-LDL, large buoyant LDL; SD-LDL, small dense LDL; Lp(a), lipoprotein(a). Adapted from Chait et al. (14).
Fig. 1.

Depiction of remnant particles by major apolipoproteins, size, nomenclature, and density. Abbreviations: CM, chylomicron; VLDL, very low-density lipoprotein; IDL, intermediate-density lipoprotein; LDL, low-density lipoprotein; HDL, high-density lipoprotein; LB-LDL, large buoyant LDL; SD-LDL, small dense LDL; Lp(a), lipoprotein(a). Adapted from Chait et al. (14).

Immunoseparation is a commercially and widely available method that detects ‘remnant-like particle cholesterol’ (RLP-C), which is the unbound fraction not adsorbed by anti-apoA1 or anti-apoB100 antibodies (61, 62). The anti-apoB100 antibody used does not bind apoE-rich VLDL or TRLs containing apoB48 (intestinally derived chylomicrons and VLDLs) and when combined with an apoA1 antibody, the immunoaffinity gel mixture adsorbs nascent apoB100 lipoproteins (LDL and VLDL) and apoA1-containing lipoproteins (HDL). The unbound fraction is then apoE enriched TRL remnants derived from chylomicrons and VLDL. The method requires pretreatment, specialized equipment, and is time-consuming and, although widely used, the epitope that -allows selective precipitation of apoB in nascent VLDL but not in remnants is not fully characterized and it is unclear whether remnants are under or overestimated by this approach (63, 64).

NMR relies on spectroscopic signals that differentiate lipoproteins based on their bulk lipid mass and requires no physical separation prior to analysis (65). Computational deconvolution of the proton NMR signal of lipid methyl groups is used for lipoprotein classification and subclass delineation. Testing can be quick (<1 minute) and does not require chemical pretreatment. Ongoing refinement of deconvolution algorithms has provided better resolution of TRL subclasses and offers unique information about particle size, particle composition, and particle number. These attributes permit ‘tagging’ of intermediate sized particles that may correspond to remnant lipoproteins. However, like ultracentrifugation, NMR separation of TRL particles does not selectively identify remnant lipoproteins that can span the size and lipid composition range, and this approach generally leads to higher estimates than other methods (14).

Direct homogenous assays offer the significant advantages of convenience and suitability for hospital- and clinic-based testing because these assays do not require pretreatment or skilled operation and can be performed on widely available automated analyzers. The method relies on specific interactions of enzymes and surfactants with lipoproteins (63). First-generation assays detected only a subfraction of TRL-C (<15%) when compared to calculated values (66) with newer assays directly measuring a substantially larger fraction (67). Scatterplots of earlier and newer generation assays from Denka performed in sequential analyses from the Copenhagen General Population Study cohorts illustrate this point (Fig. 2). The second- but not first-generation assay has been shown to associate with cardiovascular events (68, 69).

Scatterplots of measured remnant cholesterol vs calculated remnant cholesterol using older (A) and newer (B) generation direct homogenous TRL-C assays. Reproduced with permission from Varbo et al. (66) and (67).
Fig. 2.

Scatterplots of measured remnant cholesterol vs calculated remnant cholesterol using older (A) and newer (B) generation direct homogenous TRL-C assays. Reproduced with permission from Varbo et al. (66) and (67).

It is clear that no optimal way of accurately quantifying remnant TRLs currently exists. The lack of a universally accepted definition of remnant TRLs is perhaps the greatest barrier as consensus criteria on size, density, lipid, and apolipoprotein content are lacking. Yet, observations from numerous avenues of investigation demonstrate a link between increased remnant TRLs, assessed by a range of methods, and cardiovascular disease. Thus, there is a pressing need for both a consensus definition of TRL remnants, and accurate and reproducible methods for quantitation.

Clinical Studies of TRLs and Future Atherosclerotic Risk

Evidence from Observational Epidemiology

Numerous epidemiologic studies have demonstrated a positive association of remnant lipoprotein concentrations with incident cardiovascular events (Table 1). The largest of these derive from 3 Danish prospective cohorts (the Copenhagen General Population Study, the Copenhagen City Heart Study, and the Copenhagen Ischemic Heart Disease Study) that have provided a plethora of insights into the role of hypertriglyceridemia and remnant cholesterol, nonfasting concentrations in particular, as they relate to cardiovascular risk. Many other analyses have been conducted under fasting conditions that may underestimate risk associations, as fasting allows time for catabolism of dietary TGs and does not capture postprandial surges in remnants. Nonetheless, all but one of 17 major published reports from 13 independent cohorts demonstrated statistically significant associations of increased remnants with future vascular events. Null findings from the Atherosclerosis Risk in Communities Study (ARIC) (69) may relate to several factors, specifically fasting conditions, use of an earlier generation direct homogenous assay, and, importantly, adjustment for HDL in multivariable models. Whether or not to adjust for TG and HDL-C concentrations in risk models is controversial, since adjustment may overcontrol for these correlated risk factors and may be unnecessary as the weight of scientific evidence now suggests that TG and HDL-C concentrations are likely markers of aberrant remnant metabolism rather than independent causal agents.

Table 1

Major prospective studies of remnant cholesterol and incident cardiovascular disease.

AuthorYear publishedSample sizeStudy settingFasting statusMeasurementOutcomeAdjustedarisk association
HR or OR (95% CI)
Phillips NR et al. (70)1993335Montreal Heart StudyFastingUltracentrifugation: IDL-C+VLDL-CComposite: MI, coronary revascularization, cardiac death1.02b (1.01–1.03) per 1 mg/dL increase
Kugiyama K et al. (71)1999147Patients with Angiographic CADFasting

Immunoseparation

remnant TRL-C

Composite: MI, recurrent, or refractory angina requiring coronary revascularization, cardiac death

6.38b (2.3–17.6)

highest vs lowest tertile

McNamara JR et al. (72)20011567Framingham Heart Study, WomenFasting

Immunoseparation

remnant TRL-C

Composite: angina, MI, TS, TIA

2.27b (1.37–3.77)

highest quartile vs all others

Fukushima H et al. (73)2004120Patients with Angiographic CAD and DiabetesFasting

Immunoseparation

remnant TRL-C

Composite: MI, refractory angina requiring coronary revascularization, cardiac death

2.2b (1.2–6.4)

highest quartile vs all others

Imke C et al. (74)20051156Japanese-American MenFasting

Immunoseparation

remnant TRL-C

Composite: MI, revascularization, sudden death

Normal TG: 1.009 (0.939–1.085)

High TG: 1.011 (1.001–1.021)

per mg/dl

Mora S et al. (75)200927 673Women’s Health StudyNonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, IS, coronary revascularization, CVD death

Small VLDL: 1.56 (1.27–1.91)

Medium VLDL: 1.46 (1.17–1.82)

Large VLDL: 1.77 (1.34–2.33)

Total VLDL: 1.71 (1.38–2.12)

highest vs lowest quintile

Varbo A et al. (76)201373 513Copenhagen General Population Study, Copenhagen City Heart Study, and Copenhagen Ischemic Heart Disease StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIschemic heart disease

2.3 (1.7–3.1)

highest vs lowest quintile

Joshi PH et al. (77)20164114

Jackson Heart Study,

Black Participants

FastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.18 (1.00–1.39) per SD increase
Joshi PH et al. (77)2016818Framingham Offspring Cohort StudyFastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.46 (1.05–2.04) per SD increase
Lawler P et al. (78)201711 984Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER)NonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, stroke, hospitalization for unstable angina, arterial revascularization, and CVD death

Small VLDL: 1.16 (0.99–1.36)

Medium VLDL: 1.30 (1.10–1.53)

Large VLDL: 1.17 (1.00–1.37)

Total VLDL: 1.21 (1.04–1.41)

per SD increase

Varbo A et al. (67)2018106 216Copenhagen General Population Study, Stratified by WeightNonfastingCalculated as TC minus HDL-C minus LDL-CMI

Normal Weight: 2.0 (1.3–3.2)

Overweight: 1.9 (1.4–2.6)

Obese: 2.3 (1.4–3.5)

highest vs lowest quartile

Aday A et al. (79)201827 888Women’s Health StudyNonfastingNMR very small, small, medium, large, very large, and total VLDL particle concentrationPAD

Very Small VLDL: 1.01 (0.61–1.67)

Small VLDL: 0.99 (0.62–1.59)

Medium VLDL: 1.98 (1.15–3.41)

Large VLDL: 1.58 (0.94–2.68)

Very Large VLDL: 1.68 (1.05–2.55)

Total VLDL: 1.39 (0.81–2.38)

highest vs lowest tertile

Holmes M et al. (80)2018

4662

(Nested Case-Control)

Chinese Kadoorie Biobank (N = 512 891)NonfastingNMR metabolomics remnant cholesterol (VLDL-C plus IDL-C)MI, IS individually

1.27 (1.15–1.39) for MI

1.20 (1.09–1.32) for IS

per SD increase

Saeed A et al. (69)20189334Atherosclerosis Risk in Communities StudyFasting

Direct homogenous assay for TRL-C,

first generation

CHD, IS, CVD individually

b1.06 (0.88–1.27) for CHD

b1.07 (0.78–1.45) for IS

b1.05 (0.89–1.23) for CVD

highest vs lowest quartile

Vallejo-Vaz et al. (81)20189993Treating to New TargetsFastingCalculated as non-HDL-C minus LDL-CComposite: MI, total stroke, resuscitated cardiac arrest, and CVD death

Atorvastatin 10 mg: 1.48 (1.15–1.92)

Atorvastatin 80 mg: 1.01 (0.76–1.34)

highest vs lowest quintile

Varbo A et al. (82)2019102 964Copenhagen General Population StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIS

1.99 (1.49–2.67)

highest vs lowest quartile

Duran EK et al. (68)2020

976

(Case-Cohort)

Women’s Health Study (N = 27 552)NonfastingDirect homogenous assay for TRL-C, second generationComposite: MI, IS, PAD, and CVD death

1.97 (95% CI: 1.26–3.08)

highest vs lowest quartile

AuthorYear publishedSample sizeStudy settingFasting statusMeasurementOutcomeAdjustedarisk association
HR or OR (95% CI)
Phillips NR et al. (70)1993335Montreal Heart StudyFastingUltracentrifugation: IDL-C+VLDL-CComposite: MI, coronary revascularization, cardiac death1.02b (1.01–1.03) per 1 mg/dL increase
Kugiyama K et al. (71)1999147Patients with Angiographic CADFasting

Immunoseparation

remnant TRL-C

Composite: MI, recurrent, or refractory angina requiring coronary revascularization, cardiac death

6.38b (2.3–17.6)

highest vs lowest tertile

McNamara JR et al. (72)20011567Framingham Heart Study, WomenFasting

Immunoseparation

remnant TRL-C

Composite: angina, MI, TS, TIA

2.27b (1.37–3.77)

highest quartile vs all others

Fukushima H et al. (73)2004120Patients with Angiographic CAD and DiabetesFasting

Immunoseparation

remnant TRL-C

Composite: MI, refractory angina requiring coronary revascularization, cardiac death

2.2b (1.2–6.4)

highest quartile vs all others

Imke C et al. (74)20051156Japanese-American MenFasting

Immunoseparation

remnant TRL-C

Composite: MI, revascularization, sudden death

Normal TG: 1.009 (0.939–1.085)

High TG: 1.011 (1.001–1.021)

per mg/dl

Mora S et al. (75)200927 673Women’s Health StudyNonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, IS, coronary revascularization, CVD death

Small VLDL: 1.56 (1.27–1.91)

Medium VLDL: 1.46 (1.17–1.82)

Large VLDL: 1.77 (1.34–2.33)

Total VLDL: 1.71 (1.38–2.12)

highest vs lowest quintile

Varbo A et al. (76)201373 513Copenhagen General Population Study, Copenhagen City Heart Study, and Copenhagen Ischemic Heart Disease StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIschemic heart disease

2.3 (1.7–3.1)

highest vs lowest quintile

Joshi PH et al. (77)20164114

Jackson Heart Study,

Black Participants

FastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.18 (1.00–1.39) per SD increase
Joshi PH et al. (77)2016818Framingham Offspring Cohort StudyFastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.46 (1.05–2.04) per SD increase
Lawler P et al. (78)201711 984Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER)NonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, stroke, hospitalization for unstable angina, arterial revascularization, and CVD death

Small VLDL: 1.16 (0.99–1.36)

Medium VLDL: 1.30 (1.10–1.53)

Large VLDL: 1.17 (1.00–1.37)

Total VLDL: 1.21 (1.04–1.41)

per SD increase

Varbo A et al. (67)2018106 216Copenhagen General Population Study, Stratified by WeightNonfastingCalculated as TC minus HDL-C minus LDL-CMI

Normal Weight: 2.0 (1.3–3.2)

Overweight: 1.9 (1.4–2.6)

Obese: 2.3 (1.4–3.5)

highest vs lowest quartile

Aday A et al. (79)201827 888Women’s Health StudyNonfastingNMR very small, small, medium, large, very large, and total VLDL particle concentrationPAD

Very Small VLDL: 1.01 (0.61–1.67)

Small VLDL: 0.99 (0.62–1.59)

Medium VLDL: 1.98 (1.15–3.41)

Large VLDL: 1.58 (0.94–2.68)

Very Large VLDL: 1.68 (1.05–2.55)

Total VLDL: 1.39 (0.81–2.38)

highest vs lowest tertile

Holmes M et al. (80)2018

4662

(Nested Case-Control)

Chinese Kadoorie Biobank (N = 512 891)NonfastingNMR metabolomics remnant cholesterol (VLDL-C plus IDL-C)MI, IS individually

1.27 (1.15–1.39) for MI

1.20 (1.09–1.32) for IS

per SD increase

Saeed A et al. (69)20189334Atherosclerosis Risk in Communities StudyFasting

Direct homogenous assay for TRL-C,

first generation

CHD, IS, CVD individually

b1.06 (0.88–1.27) for CHD

b1.07 (0.78–1.45) for IS

b1.05 (0.89–1.23) for CVD

highest vs lowest quartile

Vallejo-Vaz et al. (81)20189993Treating to New TargetsFastingCalculated as non-HDL-C minus LDL-CComposite: MI, total stroke, resuscitated cardiac arrest, and CVD death

Atorvastatin 10 mg: 1.48 (1.15–1.92)

Atorvastatin 80 mg: 1.01 (0.76–1.34)

highest vs lowest quintile

Varbo A et al. (82)2019102 964Copenhagen General Population StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIS

1.99 (1.49–2.67)

highest vs lowest quartile

Duran EK et al. (68)2020

976

(Case-Cohort)

Women’s Health Study (N = 27 552)NonfastingDirect homogenous assay for TRL-C, second generationComposite: MI, IS, PAD, and CVD death

1.97 (95% CI: 1.26–3.08)

highest vs lowest quartile

a

Adjusted for clinical risk factors.

b

Some studies present only models additionally adjusted for TG and/or HDL-C.

Abbreviations: HR, hazards ratio; OR, odds ratio; CAD, coronary artery disease; MI, myocardial infarction; TIA, transient ischemic attack; TS, total stroke; IS, ischemic stroke; ICH, intracranial hemorrhage; PAD, peripheral artery disease; CHD, coronary heart disease; CVD, cardiovascular disease; TC, total cholesterol; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; VLDL, very low-density-lipoprotein cholesterol; IDL-C, intermediate-density lipoprotein cholesterol; TRL, triglyceride-rich lipoproteins; VAP, vertical auto profile; NMR, nuclear magnetic resonance.

Table 1

Major prospective studies of remnant cholesterol and incident cardiovascular disease.

AuthorYear publishedSample sizeStudy settingFasting statusMeasurementOutcomeAdjustedarisk association
HR or OR (95% CI)
Phillips NR et al. (70)1993335Montreal Heart StudyFastingUltracentrifugation: IDL-C+VLDL-CComposite: MI, coronary revascularization, cardiac death1.02b (1.01–1.03) per 1 mg/dL increase
Kugiyama K et al. (71)1999147Patients with Angiographic CADFasting

Immunoseparation

remnant TRL-C

Composite: MI, recurrent, or refractory angina requiring coronary revascularization, cardiac death

6.38b (2.3–17.6)

highest vs lowest tertile

McNamara JR et al. (72)20011567Framingham Heart Study, WomenFasting

Immunoseparation

remnant TRL-C

Composite: angina, MI, TS, TIA

2.27b (1.37–3.77)

highest quartile vs all others

Fukushima H et al. (73)2004120Patients with Angiographic CAD and DiabetesFasting

Immunoseparation

remnant TRL-C

Composite: MI, refractory angina requiring coronary revascularization, cardiac death

2.2b (1.2–6.4)

highest quartile vs all others

Imke C et al. (74)20051156Japanese-American MenFasting

Immunoseparation

remnant TRL-C

Composite: MI, revascularization, sudden death

Normal TG: 1.009 (0.939–1.085)

High TG: 1.011 (1.001–1.021)

per mg/dl

Mora S et al. (75)200927 673Women’s Health StudyNonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, IS, coronary revascularization, CVD death

Small VLDL: 1.56 (1.27–1.91)

Medium VLDL: 1.46 (1.17–1.82)

Large VLDL: 1.77 (1.34–2.33)

Total VLDL: 1.71 (1.38–2.12)

highest vs lowest quintile

Varbo A et al. (76)201373 513Copenhagen General Population Study, Copenhagen City Heart Study, and Copenhagen Ischemic Heart Disease StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIschemic heart disease

2.3 (1.7–3.1)

highest vs lowest quintile

Joshi PH et al. (77)20164114

Jackson Heart Study,

Black Participants

FastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.18 (1.00–1.39) per SD increase
Joshi PH et al. (77)2016818Framingham Offspring Cohort StudyFastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.46 (1.05–2.04) per SD increase
Lawler P et al. (78)201711 984Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER)NonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, stroke, hospitalization for unstable angina, arterial revascularization, and CVD death

Small VLDL: 1.16 (0.99–1.36)

Medium VLDL: 1.30 (1.10–1.53)

Large VLDL: 1.17 (1.00–1.37)

Total VLDL: 1.21 (1.04–1.41)

per SD increase

Varbo A et al. (67)2018106 216Copenhagen General Population Study, Stratified by WeightNonfastingCalculated as TC minus HDL-C minus LDL-CMI

Normal Weight: 2.0 (1.3–3.2)

Overweight: 1.9 (1.4–2.6)

Obese: 2.3 (1.4–3.5)

highest vs lowest quartile

Aday A et al. (79)201827 888Women’s Health StudyNonfastingNMR very small, small, medium, large, very large, and total VLDL particle concentrationPAD

Very Small VLDL: 1.01 (0.61–1.67)

Small VLDL: 0.99 (0.62–1.59)

Medium VLDL: 1.98 (1.15–3.41)

Large VLDL: 1.58 (0.94–2.68)

Very Large VLDL: 1.68 (1.05–2.55)

Total VLDL: 1.39 (0.81–2.38)

highest vs lowest tertile

Holmes M et al. (80)2018

4662

(Nested Case-Control)

Chinese Kadoorie Biobank (N = 512 891)NonfastingNMR metabolomics remnant cholesterol (VLDL-C plus IDL-C)MI, IS individually

1.27 (1.15–1.39) for MI

1.20 (1.09–1.32) for IS

per SD increase

Saeed A et al. (69)20189334Atherosclerosis Risk in Communities StudyFasting

Direct homogenous assay for TRL-C,

first generation

CHD, IS, CVD individually

b1.06 (0.88–1.27) for CHD

b1.07 (0.78–1.45) for IS

b1.05 (0.89–1.23) for CVD

highest vs lowest quartile

Vallejo-Vaz et al. (81)20189993Treating to New TargetsFastingCalculated as non-HDL-C minus LDL-CComposite: MI, total stroke, resuscitated cardiac arrest, and CVD death

Atorvastatin 10 mg: 1.48 (1.15–1.92)

Atorvastatin 80 mg: 1.01 (0.76–1.34)

highest vs lowest quintile

Varbo A et al. (82)2019102 964Copenhagen General Population StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIS

1.99 (1.49–2.67)

highest vs lowest quartile

Duran EK et al. (68)2020

976

(Case-Cohort)

Women’s Health Study (N = 27 552)NonfastingDirect homogenous assay for TRL-C, second generationComposite: MI, IS, PAD, and CVD death

1.97 (95% CI: 1.26–3.08)

highest vs lowest quartile

AuthorYear publishedSample sizeStudy settingFasting statusMeasurementOutcomeAdjustedarisk association
HR or OR (95% CI)
Phillips NR et al. (70)1993335Montreal Heart StudyFastingUltracentrifugation: IDL-C+VLDL-CComposite: MI, coronary revascularization, cardiac death1.02b (1.01–1.03) per 1 mg/dL increase
Kugiyama K et al. (71)1999147Patients with Angiographic CADFasting

Immunoseparation

remnant TRL-C

Composite: MI, recurrent, or refractory angina requiring coronary revascularization, cardiac death

6.38b (2.3–17.6)

highest vs lowest tertile

McNamara JR et al. (72)20011567Framingham Heart Study, WomenFasting

Immunoseparation

remnant TRL-C

Composite: angina, MI, TS, TIA

2.27b (1.37–3.77)

highest quartile vs all others

Fukushima H et al. (73)2004120Patients with Angiographic CAD and DiabetesFasting

Immunoseparation

remnant TRL-C

Composite: MI, refractory angina requiring coronary revascularization, cardiac death

2.2b (1.2–6.4)

highest quartile vs all others

Imke C et al. (74)20051156Japanese-American MenFasting

Immunoseparation

remnant TRL-C

Composite: MI, revascularization, sudden death

Normal TG: 1.009 (0.939–1.085)

High TG: 1.011 (1.001–1.021)

per mg/dl

Mora S et al. (75)200927 673Women’s Health StudyNonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, IS, coronary revascularization, CVD death

Small VLDL: 1.56 (1.27–1.91)

Medium VLDL: 1.46 (1.17–1.82)

Large VLDL: 1.77 (1.34–2.33)

Total VLDL: 1.71 (1.38–2.12)

highest vs lowest quintile

Varbo A et al. (76)201373 513Copenhagen General Population Study, Copenhagen City Heart Study, and Copenhagen Ischemic Heart Disease StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIschemic heart disease

2.3 (1.7–3.1)

highest vs lowest quintile

Joshi PH et al. (77)20164114

Jackson Heart Study,

Black Participants

FastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.18 (1.00–1.39) per SD increase
Joshi PH et al. (77)2016818Framingham Offspring Cohort StudyFastingVAP: VLDL3 plus IDL-CComposite: MI, CHD death, revascularization1.46 (1.05–2.04) per SD increase
Lawler P et al. (78)201711 984Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER)NonfastingNMR small, medium, large, and total VLDL particle concentrationComposite: MI, stroke, hospitalization for unstable angina, arterial revascularization, and CVD death

Small VLDL: 1.16 (0.99–1.36)

Medium VLDL: 1.30 (1.10–1.53)

Large VLDL: 1.17 (1.00–1.37)

Total VLDL: 1.21 (1.04–1.41)

per SD increase

Varbo A et al. (67)2018106 216Copenhagen General Population Study, Stratified by WeightNonfastingCalculated as TC minus HDL-C minus LDL-CMI

Normal Weight: 2.0 (1.3–3.2)

Overweight: 1.9 (1.4–2.6)

Obese: 2.3 (1.4–3.5)

highest vs lowest quartile

Aday A et al. (79)201827 888Women’s Health StudyNonfastingNMR very small, small, medium, large, very large, and total VLDL particle concentrationPAD

Very Small VLDL: 1.01 (0.61–1.67)

Small VLDL: 0.99 (0.62–1.59)

Medium VLDL: 1.98 (1.15–3.41)

Large VLDL: 1.58 (0.94–2.68)

Very Large VLDL: 1.68 (1.05–2.55)

Total VLDL: 1.39 (0.81–2.38)

highest vs lowest tertile

Holmes M et al. (80)2018

4662

(Nested Case-Control)

Chinese Kadoorie Biobank (N = 512 891)NonfastingNMR metabolomics remnant cholesterol (VLDL-C plus IDL-C)MI, IS individually

1.27 (1.15–1.39) for MI

1.20 (1.09–1.32) for IS

per SD increase

Saeed A et al. (69)20189334Atherosclerosis Risk in Communities StudyFasting

Direct homogenous assay for TRL-C,

first generation

CHD, IS, CVD individually

b1.06 (0.88–1.27) for CHD

b1.07 (0.78–1.45) for IS

b1.05 (0.89–1.23) for CVD

highest vs lowest quartile

Vallejo-Vaz et al. (81)20189993Treating to New TargetsFastingCalculated as non-HDL-C minus LDL-CComposite: MI, total stroke, resuscitated cardiac arrest, and CVD death

Atorvastatin 10 mg: 1.48 (1.15–1.92)

Atorvastatin 80 mg: 1.01 (0.76–1.34)

highest vs lowest quintile

Varbo A et al. (82)2019102 964Copenhagen General Population StudyNonfastingCalculated as TC minus HDL-C minus LDL-CIS

1.99 (1.49–2.67)

highest vs lowest quartile

Duran EK et al. (68)2020

976

(Case-Cohort)

Women’s Health Study (N = 27 552)NonfastingDirect homogenous assay for TRL-C, second generationComposite: MI, IS, PAD, and CVD death

1.97 (95% CI: 1.26–3.08)

highest vs lowest quartile

a

Adjusted for clinical risk factors.

b

Some studies present only models additionally adjusted for TG and/or HDL-C.

Abbreviations: HR, hazards ratio; OR, odds ratio; CAD, coronary artery disease; MI, myocardial infarction; TIA, transient ischemic attack; TS, total stroke; IS, ischemic stroke; ICH, intracranial hemorrhage; PAD, peripheral artery disease; CHD, coronary heart disease; CVD, cardiovascular disease; TC, total cholesterol; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; VLDL, very low-density-lipoprotein cholesterol; IDL-C, intermediate-density lipoprotein cholesterol; TRL, triglyceride-rich lipoproteins; VAP, vertical auto profile; NMR, nuclear magnetic resonance.

Evidence from Genetic Studies

Inferring causation from observational data is often problematic because it is not always known which of 2 variables is the cause, which is the effect, and whether both are common effects of a third unobserved variable. Genetic studies, in particular the Mendelian randomization (MR) approach, have been applied to address this uncertainty. Several MR analyses have now been published and support a causal role of TRLs in atherosclerosis. In the MR method, a single (or multiple) genetic variant(s) associated with a particular biomarker is used as a proxy for the biomarker. Outcomes are compared between the group having the affected allele(s) and groups with a reference allele(s). This approach may be considered to approximate a randomized clinical trial (RCT) in that genes are allocated randomly at birth, presumably independent of a range of potential confounding factors such as socioeconomic status and lifestyle choices. In our view, this assumption is flawed as awareness of reporter biomarker levels, especially lipid levels that are commonly measured, is analogous to unblinding in a clinical trial and may influence subsequent ‘postrandomization’ interventions, such as lipid lowering therapy or behavioral modification. These factors are not commonly modeled and may significantly alter genetic risk associations.

Another issue is genetic pleiotropy, whereby a single genetic variant may influence several traits. Almost all genetic variations in TG metabolism identified in human genetic studies also influence at least one other lipid trait, usually HDL-C (83). Three notable MR studies from Varbo et al. (76), Do et al. (84), and Holmes et al. (85) attempted to overcome this limitation by isolating variants that have limited pleiotropy, i.e., genetic effects on a single lipoprotein without effects on other lipoproteins. Varbo (76) et al. grouped genetic variants according to specificity for remnant TRL-C, specificity for both remnant TRL-C and HDL-C combined, specificity for HDL-C alone, or specificity for LDL-C alone. This study demonstrated that genetically increased remnant TRL-C was independently linked to coronary heart disease (CHD) with a roughly 3-fold excess risk per 39 mg/dL (1 mmol/L) increase in nonfasting TRL-C, while low HDL cholesterol was not. Do et al. (84) used multivariable MR to separate TG-associated effects on CHD risk from other lipid determinants. This analysis evaluated 185 common single nucleotide polymorphisms (SNPs) representing independent genetic loci that associate with at least 1 lipid trait. Associations with CHD were adjusted for SNP associations with other lipids. Their findings reaffirmed an isolated LDL-C genetic effect and demonstrated an isolated TG genetic effect, which was similar in magnitude to LDL-C after adjustment for genetic pleiotropy. An isolated genetic effect of HDL-C was not detected. Holmes et al. (85) used weighted risk scores of 135 SNPS associated with TG, HDL-C, and LDL-C. To account for genetic pleiotropy, 3 approaches were used: unrestricted risk scores (including all SNPs associated with the target lipid trait, regardless of association with other lipid traits), restricted risk scores (limited to a smaller number of SNPs associated only with the target lipid target), and statistical adjustment for plasma concentrations of nontarget lipids and statin use. The investigators found that genetically increased TG concentrations using both restricted and unrestricted risk scores were associated with CHD, but this risk was attenuated and the effect was null after adjustment for HDL-C. Given that 2 of 3 approaches provided evidence of causality, the authors concluded that TGs are likely causally related to coronary artery disease.

In another pivotal study, Ference and colleagues (86) utilized the large UK Biobank database (>650 000 participants; 91 129 with CHD). Using MR, they compared the impact of 2 genetic risk scores, 1 based on all known variants in the LPL gene associated with TG concentrations and 1 based on all known variants in the LDL-R gene associated with LDL-C concentrations. The exposure alleles were classified as those variants associated with lower TG or lower LDL, respectively, such that a higher score was expected to correlate with a lower risk of CHD. The scores were then standardized to each other by unit effects on a 10 mg/dL reduction in apoB-containing lipoproteins. For each 10-mg/dL lower plasma apoB concentration associated with variants in the LPL score, there was a corresponding 69.9-mg/dL lower TG concentration, with no change in LDL-C and a lower risk of CHD [odds ratio (OR): 0.771, 95% confidence interval (CI): 0.741–0.802]. Remarkably, for the same 10-mg/dL lower plasma apoB concentration associated with variants in the LDL-R score, there is a corresponding 14.1-mg/dL lower LDL-C concentration with no change in triglycerides, and a similar lower risk of CHD (OR: 0.773, 95% CI: 0.747–0.801). These findings indicate that the clinical benefit of lowering TGs is similar to the clinical benefit of lowering LDL-C per unit change in apoB and, further, that all apoB-containing lipoproteins, including TRLs and their remnants as well as LDL particles, have approximately the same per-particle effect on coronary risk.

Evidence from Fibrate and Omega-3 Fatty Acid Clinical Trials

Fibrates lower triglyceride concentrations by increasing VLDL clearance, principally in the VLDL1 density range, and by reducing postprandial lipemia (15, 87–89). Effects are likely mediated via their action on the peroxisome proliferator-activated receptor-α (PPAR-α) agonist response element within the apoCIII gene promoter region resulting in repressed transcription and, as a consequence, enhanced LPL activity (15). However, currently available fibrates are weak PPAR-α agonists with clinical utility limited by dose-dependent side effects and a lackluster performance in cardiovascular outcomes trials.

Five large RCTs have evaluated the effects of fibrates on cardiovascular risk (Table 2). Although early trials suggested benefit of fibrate monotherapy, the more recent FIELD (96) and ACCORD (98) studies showed no benefit of fenofibrate on cardiovascular outcomes in the setting of background statin therapy (unplanned drop-in in FIELD and by design in ACCORD). Importantly, none of these trials enrolled participants on the basis of hypertriglyceridemia and in each trial, post hoc subgroup analyses have suggested marked clinical benefit in this patient population (Table 2). For example, in meta-analyses evaluating subgroup effects (92, 99), consistently greater benefit was found in patients with high TG concentrations or mixed dyslipidemia (increased TG and low HDL-C). In these subgroups, fibrates appear to reduce cardiovascular risk by 28% (95% CI, 15–39%; P <0.001) or 30% (95% CI, 19–40%, P <0.0001), respectively, but only by 6% (95% CI, −2–13%, P =0.13) in subjects without these lipid abnormalities. The ongoing PROMINENT study (100) is a placebo-controlled cardiovascular outcomes trial of pemafibrate, a potent and selective PPAR-α (SPPARM-α) agonist, in patients with diabetes and mixed dyslipidemia [TG: 200–499 mg/dL (2.26–5.64 mmol/L) and HDL-C: ≤40 mg/dL (1.03 mmol/L)]. The trial has fully recruited (n = 10 514) and is expected to report by early 2023. Importantly, Phase II trial data in European populations on background statin therapy, have shown that pemafibrate reduces TGs, calculated TRL-C, and measured TRL-C each by roughly 40% at 12 weeks using doses currently being studied in PROMINENT (unpublished data: Duran et al., presented at the European Atherosclerosis Society Congress 2020).

Table 2

Effects of fibrates on cardiovascular events in large randomized controlled trials.

TrialDrugPatient characteristicsCV outcomeaTrial duration (years)RR reduction entire cohortAtherogenic dyslipidemia subgroupRR reduction subgroupb
HHS (90, 91)Gemfibrozil

Non-HDL-C  > 5.2 mmol/L

No CHD

Men

Nonfatal MI

and CHD death

5.0

−34%

(P < 0.02)

TG >204 mg/dL

LDL-C/HDL-C ratio >5.0

−71%

(P = 0.005)

VA-HIT (92–94)Gemfibrozil

HDL-C < 1.0 mmol/l

CHD

Men

Nonfatal MI

and CHD death

5.1−22% (P = 0.006)

TG >180 mg/dL

<40 mg/dL

−30%

(P < 0.05)

BIP (95)Bezafibrate

Previous MI or angina

Men and women

Fatal/nonfatal MI

and sudden death

6.2

−7%

(P = 0.26)

TG ≥200 mg/dL

−40%

(P = 0.02)

FIELD (96, 97)Fenofibrate

Type 2 diabetes

Some patients receiving statins

Men and women

MI, stroke, CVD death, coronary, or carotid revascularizationa5.0

−11

(P = 0.035)

TG ≥204 mg/dL

HDL-C < 40 mg/dL (men) or <50 mg/dL (women)

−27%

(P = 0.005)

ACCORD (92, 98)Fenofibrate

Type 2 diabetes

CVD or >2 CVD risk factors

Patients receiving simvastatin

Men and women

Nonfatal MI, nonfatal stroke, and CVD death4.7

−8%

(P = 0.32)

TG ≥204 mg/dL

HDL-C ≤ 34 mg/dL

−29%

(P < 0.05)

TrialDrugPatient characteristicsCV outcomeaTrial duration (years)RR reduction entire cohortAtherogenic dyslipidemia subgroupRR reduction subgroupb
HHS (90, 91)Gemfibrozil

Non-HDL-C  > 5.2 mmol/L

No CHD

Men

Nonfatal MI

and CHD death

5.0

−34%

(P < 0.02)

TG >204 mg/dL

LDL-C/HDL-C ratio >5.0

−71%

(P = 0.005)

VA-HIT (92–94)Gemfibrozil

HDL-C < 1.0 mmol/l

CHD

Men

Nonfatal MI

and CHD death

5.1−22% (P = 0.006)

TG >180 mg/dL

<40 mg/dL

−30%

(P < 0.05)

BIP (95)Bezafibrate

Previous MI or angina

Men and women

Fatal/nonfatal MI

and sudden death

6.2

−7%

(P = 0.26)

TG ≥200 mg/dL

−40%

(P = 0.02)

FIELD (96, 97)Fenofibrate

Type 2 diabetes

Some patients receiving statins

Men and women

MI, stroke, CVD death, coronary, or carotid revascularizationa5.0

−11

(P = 0.035)

TG ≥204 mg/dL

HDL-C < 40 mg/dL (men) or <50 mg/dL (women)

−27%

(P = 0.005)

ACCORD (92, 98)Fenofibrate

Type 2 diabetes

CVD or >2 CVD risk factors

Patients receiving simvastatin

Men and women

Nonfatal MI, nonfatal stroke, and CVD death4.7

−8%

(P = 0.32)

TG ≥204 mg/dL

HDL-C ≤ 34 mg/dL

−29%

(P < 0.05)

HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; TG, triglycerides; T2D, Type 2 diabetes.

To convert TG from mg/dL to mmol/L, multiply by 0.0113; to convert HDL-C from mg/dL to mmol/L, multiply by 0.0259.

a

The CV outcome presented is the prespecified primary endpoint in all trials except FIELD. In this trial, the primary endpoint (CHD) was not reported in subgroups and the data are shown for the secondary endpoint of total CVD.

b

Risk reductions and P values for subgroups when not presented in publications by trial investigators were taken from the meta-analysis of Bruckert et al. (92).

Table 2

Effects of fibrates on cardiovascular events in large randomized controlled trials.

TrialDrugPatient characteristicsCV outcomeaTrial duration (years)RR reduction entire cohortAtherogenic dyslipidemia subgroupRR reduction subgroupb
HHS (90, 91)Gemfibrozil

Non-HDL-C  > 5.2 mmol/L

No CHD

Men

Nonfatal MI

and CHD death

5.0

−34%

(P < 0.02)

TG >204 mg/dL

LDL-C/HDL-C ratio >5.0

−71%

(P = 0.005)

VA-HIT (92–94)Gemfibrozil

HDL-C < 1.0 mmol/l

CHD

Men

Nonfatal MI

and CHD death

5.1−22% (P = 0.006)

TG >180 mg/dL

<40 mg/dL

−30%

(P < 0.05)

BIP (95)Bezafibrate

Previous MI or angina

Men and women

Fatal/nonfatal MI

and sudden death

6.2

−7%

(P = 0.26)

TG ≥200 mg/dL

−40%

(P = 0.02)

FIELD (96, 97)Fenofibrate

Type 2 diabetes

Some patients receiving statins

Men and women

MI, stroke, CVD death, coronary, or carotid revascularizationa5.0

−11

(P = 0.035)

TG ≥204 mg/dL

HDL-C < 40 mg/dL (men) or <50 mg/dL (women)

−27%

(P = 0.005)

ACCORD (92, 98)Fenofibrate

Type 2 diabetes

CVD or >2 CVD risk factors

Patients receiving simvastatin

Men and women

Nonfatal MI, nonfatal stroke, and CVD death4.7

−8%

(P = 0.32)

TG ≥204 mg/dL

HDL-C ≤ 34 mg/dL

−29%

(P < 0.05)

TrialDrugPatient characteristicsCV outcomeaTrial duration (years)RR reduction entire cohortAtherogenic dyslipidemia subgroupRR reduction subgroupb
HHS (90, 91)Gemfibrozil

Non-HDL-C  > 5.2 mmol/L

No CHD

Men

Nonfatal MI

and CHD death

5.0

−34%

(P < 0.02)

TG >204 mg/dL

LDL-C/HDL-C ratio >5.0

−71%

(P = 0.005)

VA-HIT (92–94)Gemfibrozil

HDL-C < 1.0 mmol/l

CHD

Men

Nonfatal MI

and CHD death

5.1−22% (P = 0.006)

TG >180 mg/dL

<40 mg/dL

−30%

(P < 0.05)

BIP (95)Bezafibrate

Previous MI or angina

Men and women

Fatal/nonfatal MI

and sudden death

6.2

−7%

(P = 0.26)

TG ≥200 mg/dL

−40%

(P = 0.02)

FIELD (96, 97)Fenofibrate

Type 2 diabetes

Some patients receiving statins

Men and women

MI, stroke, CVD death, coronary, or carotid revascularizationa5.0

−11

(P = 0.035)

TG ≥204 mg/dL

HDL-C < 40 mg/dL (men) or <50 mg/dL (women)

−27%

(P = 0.005)

ACCORD (92, 98)Fenofibrate

Type 2 diabetes

CVD or >2 CVD risk factors

Patients receiving simvastatin

Men and women

Nonfatal MI, nonfatal stroke, and CVD death4.7

−8%

(P = 0.32)

TG ≥204 mg/dL

HDL-C ≤ 34 mg/dL

−29%

(P < 0.05)

HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; TG, triglycerides; T2D, Type 2 diabetes.

To convert TG from mg/dL to mmol/L, multiply by 0.0113; to convert HDL-C from mg/dL to mmol/L, multiply by 0.0259.

a

The CV outcome presented is the prespecified primary endpoint in all trials except FIELD. In this trial, the primary endpoint (CHD) was not reported in subgroups and the data are shown for the secondary endpoint of total CVD.

b

Risk reductions and P values for subgroups when not presented in publications by trial investigators were taken from the meta-analysis of Bruckert et al. (92).

Omega-3 fatty acids [eicosapentaenoic (EPA) and docosahexanoic (DHA) acids and their derivatives] given in high doses (>2 g/d) lower VLDL proportionate to dose and pretreatment TG concentration. In moderate hypertriglyceridemia, these agents reduce TGs by ∼20%. The landmark REDUCE-IT trial (101) showed a 25% risk reduction in major adverse cardiovascular events among patients with moderate hypertriglyceridemia assigned to icosapent ethyl at 4 g/d. Some suggest that this benefit is overestimated due to an increase in atherogenic lipoproteins and inflammatory markers in the comparison group that received a mineral oil placebo. Nonetheless, the large risk reductions cannot be easily or solely explained by placebo adjusted changes in these biomarkers and the clinical safety of employing mineral oil placebos has been recently reviewed (102). The notable finding that achieved TG concentrations in REDUCE-IT did not match achieved risk reduction was surprising but not unique for TG-lowering trials. Risk reductions in neither the HHS (103) nor VA-HIT (104) were correlated with achieved TG concentrations despite significant TG reduction in both of these fibrate trials. In HHS, the change in fasting TG with gemfibrozil therapy was not associated with the trial endpoint in either the total cohort or the subgroup with increased baseline concentrations [TG ≥ 177 mg/dL (2 mmol/L)] (103). Neither was on treatment fasting TG a significant predictor of the primary endpoint in VA-HIT [RR per 50 mg/dL 1.05 (0.98–0.97); P = 0.16] (104). As discussed in this review, risk reduction may better correlate with TRL-C concentrations; however, these data have not been available from these trials.

Conclusions and Future Directions

As discussed in this review, the scientific landscape with regard to the involvement of triglyceride-rich remnants in development of cardiovascular disease is rapidly changing. The lipoprotein system is comprised of a continuum of particles of differing size and composition that begin as chylomicrons and VLDL with catabolism to LDL. Hypertriglyceridemia is associated with aberrant TRL modification yielding remnants that are increasingly recognized as proatherogenic. The data now lead many to believe that TG-rich remnant particles by virtue of their excess cholesterol content, apolipoprotein composition, and structural properties are causal factors and along with their metabolic pathways are now potential targets for therapeutic modulation. Several novel agents (Table 3) are being tested in this regard and hold substantial promise to address residual cardiovascular risk that remains an important clinical problem. Continued scientific work must better inform our knowledge gaps, reliable biomarkers must be developed for use in clinical practice, and clinical trials must demonstrate treatment benefits to ultimately determine whether this line of investigation translates into new and effective paradigms for lipid management.

Table 3

Emerging therapies for treatment of hypertriglyceridemia.

Primary mechanism of TRL loweringTherapeutic classExamplesa
Suppression of TRL productionSelective PPAR modulators

Pemafibrate (K-877)

Elafibranor (GFT505)

MTTP inhibitors

JuxtapidTM (lomitapide)

Slx-4090

DGAT1 inhibitorPradigastat
LPL activationLPL activatorIbrolipim
ApoC3 antisense oligonucleotideVolanesorsen (ISIS 304801)
ANGPTL3 inhibitor

Evinacumab (REGN1500)

IONIS-ANGPTL3-LRx

Complex effectsPrescription omega-3 fatty acid preparations

VascepaTM (EPA)

EpanovaTM (EPA plus DHA)

LovazaTM (EPA plus DHA)

OmtrgTM (EPA plus DHA)

Icosabutate (EPA)

Primary mechanism of TRL loweringTherapeutic classExamplesa
Suppression of TRL productionSelective PPAR modulators

Pemafibrate (K-877)

Elafibranor (GFT505)

MTTP inhibitors

JuxtapidTM (lomitapide)

Slx-4090

DGAT1 inhibitorPradigastat
LPL activationLPL activatorIbrolipim
ApoC3 antisense oligonucleotideVolanesorsen (ISIS 304801)
ANGPTL3 inhibitor

Evinacumab (REGN1500)

IONIS-ANGPTL3-LRx

Complex effectsPrescription omega-3 fatty acid preparations

VascepaTM (EPA)

EpanovaTM (EPA plus DHA)

LovazaTM (EPA plus DHA)

OmtrgTM (EPA plus DHA)

Icosabutate (EPA)

PPAR, peroxisome proliferator activator receptor; MTTP, microsomal triglyceride transfer protein; DGAT1, diacylglycerol acyltransferase 1; LPL, lipoprotein lipase; ApoC3, apolipoprotein C3; ANGPTL3, angiopoietin-like protein-3; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid.

a

Some already approved for clinical use in familial hypercholesterolemia or severe hypertriglyceridemia.

Table 3

Emerging therapies for treatment of hypertriglyceridemia.

Primary mechanism of TRL loweringTherapeutic classExamplesa
Suppression of TRL productionSelective PPAR modulators

Pemafibrate (K-877)

Elafibranor (GFT505)

MTTP inhibitors

JuxtapidTM (lomitapide)

Slx-4090

DGAT1 inhibitorPradigastat
LPL activationLPL activatorIbrolipim
ApoC3 antisense oligonucleotideVolanesorsen (ISIS 304801)
ANGPTL3 inhibitor

Evinacumab (REGN1500)

IONIS-ANGPTL3-LRx

Complex effectsPrescription omega-3 fatty acid preparations

VascepaTM (EPA)

EpanovaTM (EPA plus DHA)

LovazaTM (EPA plus DHA)

OmtrgTM (EPA plus DHA)

Icosabutate (EPA)

Primary mechanism of TRL loweringTherapeutic classExamplesa
Suppression of TRL productionSelective PPAR modulators

Pemafibrate (K-877)

Elafibranor (GFT505)

MTTP inhibitors

JuxtapidTM (lomitapide)

Slx-4090

DGAT1 inhibitorPradigastat
LPL activationLPL activatorIbrolipim
ApoC3 antisense oligonucleotideVolanesorsen (ISIS 304801)
ANGPTL3 inhibitor

Evinacumab (REGN1500)

IONIS-ANGPTL3-LRx

Complex effectsPrescription omega-3 fatty acid preparations

VascepaTM (EPA)

EpanovaTM (EPA plus DHA)

LovazaTM (EPA plus DHA)

OmtrgTM (EPA plus DHA)

Icosabutate (EPA)

PPAR, peroxisome proliferator activator receptor; MTTP, microsomal triglyceride transfer protein; DGAT1, diacylglycerol acyltransferase 1; LPL, lipoprotein lipase; ApoC3, apolipoprotein C3; ANGPTL3, angiopoietin-like protein-3; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid.

a

Some already approved for clinical use in familial hypercholesterolemia or severe hypertriglyceridemia.

Author Contributions

All authors confirmed they have contributed to the intellectual content of this paper and have met the following 4 requirements: (a) significant contributions to the conception and design, acquisition of data, or analysis and interpretation of data; (b) drafting or revising the article for intellectual content; (c) final approval of the published article; and (d) agreement to be accountable for all aspects of the article thus ensuring that questions related to the accuracy or integrity of any part of the article are appropriately investigated and resolved.

Authors’ Disclosures or Potential Conflicts of Interest

Upon manuscript submission, all authors completed the author disclosure form. Disclosures and/or potential conflicts of interest:

Employment or Leadership: None declared.

Consultant or Advisory Role: None declared.

Stock Ownership: None declared.

Honoraria: A.D. Pradhan, Medintelligence, Metabolic Endocrine Education Foundation, North American Center for Continuing Medical Education, Physician's Education Resource LLC.

Research Funding: E.K. Duran, the National Institutes of Health under Award Number 5T32HL007575-33; A.D. Pradhan, investigator-initiated research support from Kowa Research Institute, Kowa Pharmaceuticals Europe Co. Ltd, Denka Seiken, co-Principal Investigator of the PROMINENT trial (NCT03071692).

Expert Testimony: None declared.

Patents:None declared.

References

1

Laufs
U
,
Parhofer
KG
,
Ginsberg
HN
,
Hegele
RA.
Clinical review on triglycerides
.
Eur Heart J
2020
;
41
:
99
109c
.

2

Sumner
AE
,
Cowie
CC.
Ethnic differences in the ability of triglyceride levels to identify insulin resistance
.
Atherosclerosis
2008
;
196
:
696
703
.

3

Carroll
M
,
Kit
B
,
Lacher
D.
Trends in elevated triglyceride in adults: United States, 2001
2012
.
NCHS Data Brief
2015
;
198
:
1
–8.

4

Gu
Q
,
Paulose-Ram
R
,
Vl
B
,
Bk
K.
Prescription cholesterol-lowering medication use in adults aged 40 and over: United States, 2003
2012
.
NCHS Data Brief
2014
;
1
8
.

5

Fan
W
,
Philip
S
,
Granowitz
C
,
Toth
PP
,
Wong
ND.
Residual hypertriglyceridemia and estimated atherosclerotic cardiovascular disease risk by statin use in U.S. adults with diabetes: National Health and Nutrition Examination Survey 2007
2014
.
Diabetes Care
2019
;
42
:
2307
14
.

6

Lawler
PR
,
Kotrri
G
,
Koh
M
,
Goodman
SG
,
Farkouh
ME
,
Lee
DS
, et al.
Real-world risk of cardiovascular outcomes associated with hypertriglyceridaemia among individuals with atherosclerotic cardiovascular disease and potential eligibility for emerging therapies
.
Eur Heart J
2020
;
41
:
86
94
.

7

Packard
CJ
,
Shepherd
J.
Lipoprotein heterogeneity and apolipoprotein B metabolism
.
Arterioscler Thromb Vasc Biol
1997
;
17
:
3542
56
.

8

Ginsberg
HN.
New perspectives on atherogenesis: role of abnormal triglyceride-rich lipoprotein metabolism
.
Circulation
2002
;
106
:
2137
42
.

9

Dallinga-Thie
GM
,
Kroon
J
,
Boren
J
,
Chapman
MJ.
Triglyceride-rich lipoproteins and remnants: targets for therapy?
Curr Cardiol Rep
2016
;
18
:
67
.

10

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

11

Bernelot Moens
SJ
,
Verweij
SL
,
Schnitzler
JG
,
Stiekema
LCA
,
Bos
M
,
Langsted
A
, et al.
Remnant cholesterol elicits arterial wall inflammation and a multilevel cellular immune response in humans
.
Arterioscler Thromb Vasc Biol
2017
;
37
:
969
75
.

12

Hansen
SEJ
,
Madsen
CM
,
Varbo
A
,
Nordestgaard
BG.
Low-grade inflammation in the association between mild-to-moderate hypertriglyceridemia and risk of acute pancreatitis: a study of more than 115000 individuals from the general population
.
Clin Chem
2019
;
65
:
321
32
.

13

Bjornson
E
,
Packard
CJ
,
Adiels
M
,
Andersson
L
,
Matikainen
N
,
Soderlund
S
, et al.
Apolipoprotein B48 metabolism in chylomicrons and very low-density lipoproteins and its role in triglyceride transport in normo- and hypertriglyceridemic human subjects
.
J Intern Med
2020
;
288
:
422
38
.

14

Chait
A
,
Ginsberg
HN
,
Vaisar
T
,
Heinecke
JW
,
Goldberg
IJ
,
Bornfeldt
KE.
Remnants of the triglyceride-rich lipoproteins, diabetes, and cardiovascular disease
.
Diabetes
2020
;
69
:
508
16
.

15

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

16

Salinas
CAA
,
Chapman
MJ.
Remnant lipoproteins: are they equal to or more atherogenic than LDL?
Curr Opin Lipidol
2020
;
31
:
132
9
.

17

Potts
JL
,
Fisher
RM
,
Humphreys
SM
,
Coppack
SW
,
Gibbons
GF
,
Frayn
KN.
Peripheral triacylglycerol extraction in the fasting and post-prandial states
.
Clin Sci (Lond)
1991
;
81
:
621
6
.

18

Goldberg
IJ.
Lipoprotein lipase and lipolysis: central roles in lipoprotein metabolism and atherogenesis
.
J Lipid Res
1996
;
37
:
693
707
.

19

Bickerton
AS
,
Roberts
R
,
Fielding
BA
,
Hodson
L
,
Blaak
EE
,
Wagenmakers
AJ
, et al.
Preferential uptake of dietary fatty acids in adipose tissue and muscle in the postprandial period
.
Diabetes
2007
;
56
:
168
76
.

20

Illingworth
DR.
Lipoprotein metabolism
.
Am J Kidney Dis
1993
;
22
:
90
7
.

21

Ji
ZS
,
Fazio
S
,
Lee
YL
,
Mahley
RW.
Secretion-capture role for apolipoprotein E in remnant lipoprotein metabolism involving cell surface heparan sulfate proteoglycans
.
J Biol Chem
1994
;
269
:
2764
72
.

22

Curtiss
LK
,
Boisvert
WA.
Apolipoprotein E and atherosclerosis
.
Curr Opin Lipidol
2000
;
11
:
243
51
.

23

Redgrave
TG.
Chylomicron metabolism
.
Biochem Soc Trans
2004
;
32
:
79
82
.

24

Mahley
RW
,
Huang
Y.
Atherogenic remnant lipoproteins: role for proteoglycans in trapping, transferring, and internalizing
.
J Clin Invest
2007
;
117
:
1
8
.

25

Barrows
BR
,
Parks
EJ.
Contributions of different fatty acid sources to very low-density lipoprotein-triacylglycerol in the fasted and fed states
.
J Clin Endocrinol Metab
2006
;
91
:
1446
52
.

26

Taskinen
MR
,
Adiels
M
,
Westerbacka
J
,
Soderlund
S
,
Kahri
J
,
Lundbom
N
, et al.
Dual metabolic defects are required to produce hypertriglyceridemia in obese subjects
.
Arterioscler Thromb Vasc Biol
2011
;
31
:
2144
50
.

27

Gaw
A
,
Packard
CJ
,
Lindsay
GM
,
Griffin
BA
,
Caslake
MJ
,
Lorimer
AR
,
Shepherd
J.
Overproduction of small very low density lipoproteins (Sf 20-60) in moderate hypercholesterolemia: relationships between apolipoprotein B kinetics and plasma lipoproteins
.
J Lipid Res
1995
;
36
:
158
71
.

28

James
RW
,
Martin
B
,
Pometta
D
,
Fruchart
JC
,
Duriez
P
,
Puchois
P
, et al.
Apolipoprotein B metabolism in homozygous familial hypercholesterolemia
.
J Lipid Res
1989
;
30
:
159
69
.

29

Millar
JS
,
Maugeais
C
,
Ikewaki
K
,
Kolansky
DM
,
Barrett
PH
,
Budreck
EC
, et al.
Complete deficiency of the low-density lipoprotein receptor is associated with increased apolipoprotein B-100 production
.
Arterioscler Thromb Vasc Biol
2005
;
25
:
560
5
.

30

Prinsen
BH
,
Romijn
JA
,
Bisschop
PH
,
de Barse
MM
,
Barrett
PH
,
Ackermans
M
, et al.
Endogenous cholesterol synthesis is associated with VLDL-2 apoB-100 production in healthy humans
.
J Lipid Res
2003
;
44
:
1341
8
.

31

Fisher
WR.
Apoprotein B kinetics in man: concepts and questions. In:
Berman
M
,
Grundy
SM
,
Howard
BV
, editors.
Lipoprotein kinetics and modeling.
New York
:
Academic Press
;
1982
. p.
43
68
.

32

Packard
CJ
,
Gaw
A
,
Demant
T
,
Shepherd
J.
Development and application of a multicompartmental model to study very low density lipoprotein subfraction metabolism
.
J Lipid Res
1995
;
36
:
172
87
.

33

Packard
CJ
,
Demant
T
,
Stewart
JP
,
Bedford
D
,
Caslake
MJ
,
Schwertfeger
G
, et al.
Apolipoprotein B metabolism and the distribution of VLDL and LDL subfractions
.
J Lipid Res
2000
;
41
:
305
18
.

34

Schwartz
EA
,
Reaven
PD.
Lipolysis of triglyceride-rich lipoproteins, vascular inflammation, and atherosclerosis
.
Biochim Biophys Acta
2012
;
1821
:
858
66
.

35

Havel
RJ
,
Kane
JP
,
Kashyap
ML.
Interchange of apolipoproteins between chylomicrons and high density lipoproteins during alimentary lipemia in man
.
J Clin Invest
1973
;
52
:
32
8
.

36

Mjos
OD
,
Faergeman
O
,
Hamilton
RL
,
Havel
RJ.
Characterization of remnants produced during the metabolism of triglyceride-rich lipoproteins of blood plasma and intestinal lymph in the rat
.
J Clin Invest
1975
;
56
:
603
15
.

37

Redgrave
TG
,
Small
DM.
Quantitation of the transfer of surface phospholipid of chylomicrons to the high density lipoprotein fraction during the catabolism of chylomicrons in the rat
.
J Clin Invest
1979
;
64
:
162
71
.

38

Marcoux
C
,
Tremblay
M
,
Fredenrich
A
,
Davignon
J
,
Cohn
JS.
Lipoprotein distribution of apolipoprotein C-III and its relationship to the presence in plasma of triglyceride-rich remnant lipoproteins
.
Metabolism
2001
;
50
:
112
9
.

39

Batal
R
,
Tremblay
M
,
Barrett
PH
,
Jacques
H
,
Fredenrich
A
,
Mamer
O
, et al.
Plasma kinetics of apoC-III and apoE in normolipidemic and hypertriglyceridemic subjects
.
J Lipid Res
2000
;
41
:
706
18
.

40

Stender
S
,
Zilversmit
DB.
Transfer of plasma lipoprotein components and of plasma proteins into aortas of cholesterol-fed rabbits. Molecular size as a determinant of plasma lipoprotein influx
.
Arteriosclerosis
1981
;
1
:
38
49
.

41

Nordestgaard
BG
,
Wootton
R
,
Lewis
B.
Selective retention of VLDL, IDL, and LDL in the arterial intima of genetically hyperlipidemic rabbits in vivo. Molecular size as a determinant of fractional loss from the intima-inner media
.
Arterioscler Thromb Vasc Biol
1995
;
15
:
534
42
.

42

Hodis
HN.
Triglyceride-rich lipoprotein remnant particles and risk of atherosclerosis
.
Circulation
1999
;
99
:
2852
4
.

43

Rutledge
JC
,
Mullick
AE
,
Gardner
G
,
Goldberg
IJ.
Direct visualization of lipid deposition and reverse lipid transport in a perfused artery: roles of VLDL and HDL
.
Circ Res
2000
;
86
:
768
73
.

44

Proctor
SD
,
Vine
DF
,
Mamo
JC.
Arterial permeability and efflux of apolipoprotein B-containing lipoproteins assessed by in situ perfusion and three-dimensional quantitative confocal microscopy
.
Arterioscler Thromb Vasc Biol
2004
;
24
:
2162
7
.

45

Van Lenten
BJ
,
Fogelman
AM
,
Jackson
RL
,
Shapiro
S
,
Haberland
ME
,
Edwards
PA.
Receptor-mediated uptake of remnant lipoproteins by cholesterol-loaded human monocyte-macrophages
.
J Biol Chem
1985
;
260
:
8783
8
.

46

Marais
AD.
Apolipoprotein E in lipoprotein metabolism, health and cardiovascular disease
.
Pathology
2019
;
51
:
165
76
.

47

Wilhelm
MG
,
Cooper
AD.
Induction of atherosclerosis by human chylomicron remnants: a hypothesis
.
J Atheroscler Thromb
2003
;
10
:
132
9
.

48

Farukhi
ZM
,
Demler
OV
,
Caulfield
MP
,
Kulkarni
K
,
Wohlgemuth
J
,
Cobble
M
, et al.
Comparison of nonfasting and fasting lipoprotein subfractions and size in 15,397 apparently healthy individuals: an analysis from the Vitamin D and OmegA-3 Trial
.
J Clin Lipidol
2020
;
14
:
241
51
.

49

Proctor
SD
,
Mamo
JC.
Retention of fluorescent-labelled chylomicron remnants within the intima of the arterial wall—evidence that plaque cholesterol may be derived from post-prandial lipoproteins
.
Eur J Clin Invest
1998
;
28
:
497
503
.

50

Elsegood
CL
,
Pal
S
,
Roach
PD
,
Mamo
JC.
Binding and uptake of chylomicron remnants by primary and THP-1 human monocyte-derived macrophages: determination of binding proteins
.
Clin Sci (Lond)
2001
;
101
:
111
9
.

51

Yla-Herttuala
S
,
Lipton
BA
,
Rosenfeld
ME
,
Goldberg
IJ
,
Steinberg
D
,
Witztum
JL.
Macrophages and smooth muscle cells express lipoprotein lipase in human and rabbit atherosclerotic lesions
.
Proc Natl Acad Sci USA
1991
;
88
:
10143
7
.

52

O'Brien
KD
,
Gordon
D
,
Deeb
S
,
Ferguson
M
,
Chait
A.
Lipoprotein lipase is synthesized by macrophage-derived foam cells in human coronary atherosclerotic plaques
.
J Clin Invest
1992
;
89
:
1544
50
.

53

Hasham
SN
,
Pillarisetti
S.
Vascular lipases, inflammation and atherosclerosis
.
Clin Chim Acta
2006
;
372
:
179
83
.

54

Botham
KM
,
Wheeler-Jones
CP.
Postprandial lipoproteins and the molecular regulation of vascular homeostasis
.
Prog Lipid Res
2013
;
52
:
446
64
.

55

Zilversmit
DB.
A proposal linking atherogenesis to the interaction of endothelial lipoprotein lipase with triglyceride-rich lipoproteins
.
Circ Res
1973
;
33
:
633
8
.

56

Zilversmit
DB.
Atherogenesis: a postprandial phenomenon
.
Circulation
1979
;
60
:
473
85
.

57

Friedewald
WT
,
Levy
RI
,
Fredrickson
DS.
Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge
.
Clin Chem
1972
;
18
:
499
502
.

58

Langlois
MR
,
Nordestgaard
BG
,
Langsted
A
,
Chapman
MJ
,
Aakre
KM
,
Baum
H
, et al.
Quantifying atherogenic lipoproteins for lipid-lowering strategies: consensus-based recommendations from EAS and EFLM
.
Clin Chem Lab Med
2020
;
58
:
496
517
.

59

Cohn
JS
,
Marcoux
C
,
Davignon
J.
Detection, quantification, and characterization of potentially atherogenic triglyceride-rich remnant lipoproteins
.
Arterioscler Thromb Vasc Biol
1999
;
19
:
2474
86
.

60

Kulkarni
KR.
Cholesterol profile measurement by vertical auto profile method
.
Clin Lab Med
2006
;
26
:
787
802
.

61

Nakajima
K
,
Saito
T
,
Tamura
A
,
Suzuki
M
,
Nakano
T
,
Adachi
M
, et al.
Cholesterol in remnant-like lipoproteins in human serum using monoclonal anti apo B-100 and anti apo A-I immunoaffinity mixed gels
.
Clin Chim Acta
1993
;
223
:
53
71
.

62

Nakajima
K
,
Okazaki
M
,
Tanaka
A
,
Pullinger
C
,
Wang
T
,
Nakano
T.
Separation and determination of remnant-like particles in human serum using monoclonal antibodies to apoB-100 and apoA-1
.
J Clin Ligand Assay
1996
;
19
:
177
83
.

63

Miyauchi
K
,
Kayahara
N
,
Ishigami
M
,
Kuwata
H
,
Mori
H
,
Sugiuchi
H
, et al.
Development of a homogeneous assay to measure remnant lipoprotein cholesterol
.
Clin Chem
2007
;
53
:
2128
35
.

64

Schaefer
EJ.
Limitations of automated remnant lipoprotein cholesterol assay for diagnostic use
.
Clin Chem
2009
;
55
:
2061
2; author reply 2062. 3
.

65

Otvos
J.
Measurement of triglyceride-rich lipoproteins by nuclear magnetic resonance spectroscopy
.
Clin Cardiol
1999
;
22
:
II-21
7
.

66

Varbo
A
,
Freiberg
JJ
,
Nordestgaard
BG.
Extreme nonfasting remnant cholesterol vs extreme LDL cholesterol as contributors to cardiovascular disease and all-cause mortality in 90000 individuals from the general population
.
Clin Chem
2015
;
61
:
533
43
.

67

Varbo
A
,
Freiberg
JJ
,
Nordestgaard
BG.
Remnant cholesterol and myocardial infarction in normal weight, overweight, and obese individuals from the Copenhagen General Population Study
.
Clin Chem
2018
;
64
:
219
30
.

68

Duran
EK
,
Aday
AW
,
Cook
NR
,
Buring
JE
,
Ridker
PM
,
Pradhan
AD.
Triglyceride-rich lipoprotein cholesterol, small dense LDL cholesterol, and incident cardiovascular disease
.
J Am Coll Cardiol
2020
;
75
:
2122
35
.

69

Saeed
A
,
Feofanova
EV
,
Yu
B
,
Sun
W
,
Virani
SS
,
Nambi
V
, et al.
Remnant-like particle cholesterol, low-density lipoprotein triglycerides, and incident cardiovascular disease
.
J Am Coll Cardiol
2018
;
72
:
156
69
.

70

Phillips
NR
,
Waters
D
,
Havel
RJ.
Plasma lipoproteins and progression of coronary artery disease evaluated by angiography and clinical events
.
Circulation
1993
;
88
:
2762
70
.

71

Kugiyama
K
,
Doi
H
,
Takazoe
K
,
Kawano
H
,
Soejima
H
,
Mizuno
Y
, et al.
Remnant lipoprotein levels in fasting serum predict coronary events in patients with coronary artery disease
.
Circulation
1999
;
99
:
2858
60
.

72

McNamara
JR
,
Shah
PK
,
Nakajima
K
,
Cupples
LA
,
Wilson
PW
,
Ordovas
JM
,
Schaefer
EJ.
Remnant-like particle (RLP) cholesterol is an independent cardiovascular disease risk factor in women: results from the Framingham Heart Study
.
Atherosclerosis
2001
;
154
:
229
36
.

73

Fukushima
H
,
Sugiyama
S
,
Honda
O
,
Koide
S
,
Nakamura
S
,
Sakamoto
T
, et al.
Prognostic value of remnant-like lipoprotein particle levels in patients with coronary artery disease and type II diabetes mellitus
.
J Am Coll Cardiol
2004
;
43
:
2219
24
.

74

Imke
C
,
Rodriguez
BL
,
Grove
JS
,
McNamara
JR
,
Waslien
C
,
Katz
AR
, et al.
Are remnant-like particles independent predictors of coronary heart disease incidence? The Honolulu Heart Study
.
Arterioscler Thromb Vasc Biol
2005
;
25
:
1718
22
.

75

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
9
.

76

Varbo
A
,
Benn
M
,
Tybjærg-Hansen
A
,
Jørgensen
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
.

77

Joshi
PH
,
Khokhar
AA
,
Massaro
JM
,
Lirette
ST
,
Griswold
ME
,
Martin
SS
, et al.
Remnant lipoprotein cholesterol and incident coronary heart disease: the Jackson Heart and Framingham Offspring Cohort Studies
.
J Am Heart Assoc
2016
;
5
:e002765.

78

Lawler
PR
,
Akinkuolie
AO
,
Chu
AY
,
Shah
SH
,
Kraus
WE
,
Craig
D
, et al.
Atherogenic lipoprotein determinants of cardiovascular disease and residual risk among individuals with low low-density lipoprotein cholesterol
.
J Am Heart Assoc
2017
;
6
:e005549.

79

Aday
AW
,
Lawler
PR
,
Cook
NR
,
Ridker
PM
,
Mora
S
,
Pradhan
AD.
Lipoprotein particle profiles, standard lipids, and peripheral artery disease incidence
.
Circulation
2018
;
138
:
2330
41
.

80

Holmes
MV
,
Millwood
IY
,
Kartsonaki
C
,
Hill
MR
,
Bennett
DA
,
Boxall
R
, et al.
Lipids, lipoproteins, and metabolites and risk of myocardial infarction and stroke
.
J Am Coll Cardiol
2018
;
71
:
620
32
.

81

Vallejo-Vaz
AJ
,
Fayyad
R
,
Boekholdt
SM
,
Hovingh
GK
,
Kastelein
JJ
,
Melamed
S
,
Barter
P
, 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
.

82

Varbo
A
,
Nordestgaard
BG.
Remnant cholesterol and risk of ischemic stroke in 112,512 individuals from the general population
.
Ann Neurol
2019
;
85
:
550
9
.

83

Dron
JS
,
Hegele
RA.
Genetics of triglycerides and the risk of atherosclerosis
.
Curr Atheroscler Rep
2017
;
19
:
31
.

84

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

85

Holmes
MV
,
Asselbergs
FW
,
Palmer
TM
,
Drenos
F
,
Lanktree
MB
,
Nelson
CP
, et al.
Mendelian randomization of blood lipids for coronary heart disease
.
Eur Heart J
2015
;
36
:
539
50
.

86

Ference
BA
,
Kastelein
JJP
,
Ray
KK
,
Ginsberg
HN
,
Chapman
MJ
,
Packard
CJ
, et al.
Association of triglyceride-lowering LPL variants and LDL-C-lowering LDLR variants with risk of coronary heart disease
.
JAMA
2019
;
321
:
364
73
.

87

Shepherd
J
,
Packard
CJ
,
Stewart
JM
,
Atmeh
RF
,
Clark
RS
,
Boag
DE
, et al.
Apolipoprotein A and B (Sf 100-400) metabolism during bezafibrate therapy in hypertriglyceridemic subjects
.
J Clin Invest
1984
;
74
:
2164
77
.

88

Ginsberg
HN.
Changes in lipoprotein kinetics during therapy with fenofibrate and other fibric acid derivatives
.
Am J Med
1987
;
83
:
66
70
.

89

Packard
CJ.
Overview of fenofibrate.
Eur Heart J
1998
;
19(Suppl A
):
A62
5
.

90

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
.

91

Manninen
V
,
Tenkanen
L
,
Koskinen
P
,
Huttunen
JK
,
Manttari
M
,
Heinonen
OP
,
Frick
MH.
Joint effects of serum triglyceride and LDL cholesterol and HDL cholesterol concentrations on coronary heart disease risk in the Helsinki Heart Study. Implications for treatment
.
Circulation
1992
;
85
:
37
45
.

92

Bruckert
E
,
Labreuche
J
,
Deplanque
D
,
Touboul
PJ
,
Amarenco
P.
Fibrates effect on cardiovascular risk is greater in patients with high triglyceride levels or atherogenic dyslipidemia profile: a systematic review and meta-analysis
.
J Cardiovasc Pharmacol
2011
;
57
:
267
72
.

93

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
.

94

Robins
SJ
,
Collins
D
,
Wittes
JT
,
Papademetriou
V
,
Deedwania
PC
,
Schaefer
EJ
,
McNamara
JR
, et al.
Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial
.
JAMA
2001
;
285
:
1585
91
.

95

Bezafibrate Infarction Prevention Study. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease
.
Circulation
2000
;
102
:
21
7
.

96

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
.

97

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
.,.

98

Group
AS
,
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
.

99

Lee
M
,
Saver
JL
,
Towfighi
A
,
Chow
J
,
Ovbiagele
B.
Efficacy of fibrates for cardiovascular risk reduction in persons with atherogenic dyslipidemia: a meta-analysis
.
Atherosclerosis
2011
;
217
:
492
8
.

100

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
.

101

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
.

102

Olshansky
B
,
Chung
MK
,
Budoff
MJ
,
Philip
S
,
Jiao
L
,
Doyle
RT
Jr
, et al.
Mineral oil: safety and use as placebo in REDUCE-IT and other clinical studies
.
Eur Heart J Suppl
2020
;
22
:
J34
J48
.

103

Manninen
V
,
Elo
MO
,
Frick
MH
,
Haapa
K
,
Heinonen
OP
,
Heinsalmi
P
, et al.
Lipid alterations and decline in the incidence of coronary heart disease in the Helsinki Heart Study
.
JAMA
1988
;
260
:
641
51
.

104

Williams
KJ
,
Chen
K.
Recent insights into factors affecting remnant lipoprotein uptake
.
Curr Opin Lipidol
2010
;
21
:
218
28
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)