Abstract

Context:

While substantial benefit has accrued with respect to prevention and treatment of atherosclerotic cardiovascular disease (ASCVD) since the advent of statin therapy, much remains unknown and there is considerable need to address residual risk beyond statins. Moreover, many individuals are unable to tolerate statins.

Evidence Acquisition:

As a result of several recent clinical trials and publications describing early Phase 1–3 clinical trials, the authors briefly discuss the current situation regarding pharmacological management for the prevention and treatment of individuals with disorders of lipid and lipoprotein metabolism, outline some of the unanswered questions, and speculate on where we might expect to be in 5–10 years.

Evidence Synthesis:

Fortunately, recent developments in drug therapy hold considerable promise of additional benefits. In addition, new drugs in the pipeline, ongoing clinical trials, and new approaches to treatment hold promise for further improvements in therapy in the foreseeable future. During the next 5–10 years, we expect to know whether the PCSK9 inhibitors indeed live up to their promise and result in the hoped-for reduction in ASCVD events, whether triglyceride lowering indeed adds additional benefit, how best to approach HDL, and the importance of lipoprotein (a). Advances in the use of molecular biological approaches such as anti-sense oligonucleotides and RNA silencing, and the use of biological agents such as PSCK9 antibodies, is likely to play an important role in these advances.

Conclusions:

The advent of PCSK9 inhibitors is likely to provide a major breakthrough in the management of individuals with heterozygous familial hypercholesterolemia, patients with established ASCVD who are unable to reach targets with other therapies, and high-risk individuals with statin intolerance. The next 5–10 years should also clarify uncertainties concerning the pharmacological management of individuals with low levels of HDL-cholesterol, hypertriglyceridemia, and elevated lipoprotein (a).

The relationship between lipids, lipoproteins, and atherosclerotic cardiovascular disease (ASCVD) has been known for more than a half a century. Tremendous progress in understanding the relationship between lipoproteins and cardiovascular disease (CVD) has occurred during the past 3 decades, including major advances in the prevention and treatment of CVD. By far the most successful of the therapeutic strategies has been the use of statins. However, despite their widespread use and current cost-effectiveness, CVD remains the leading cause of death in the United States, and considerable residual risk remains, much of which is attributable to lipids and lipoproteins. This article will focus on emerging therapeutics, and on unresolved issues that need to be addressed during the next 5–10 years, all of which are aimed at reducing the poststatin residual risk of CVD. To date, the greatest success has been achieved by a strategy that has focused on reduction of low-density lipoprotein (LDL) levels.

LDL

The robust relationship between plasma cholesterol and CVD has been appreciated for a very long time (1, 2). As research began to focus on plasma lipoproteins rather than lipids, it became clear that the relationship between cholesterol and CVD was primarily due to LDL cholesterol. Statins reduce LDL levels primarily by inhibiting 3-hydroxy-3-methylglutaryl-coenzyme A-reductase, the rate-limiting enzyme in cholesterol synthesis. This leads to an upregulation of LDL receptors (3), but also is associated with a modest reduction in LDL production (4). Since the introduction of statins in the mid-1980s, their use has resulted in a remarkable reduction of coronary heart disease (CHD) events. Statins have proved to be of value in both primary and secondary prevention, and also have reduced cardiovascular mortality (5, 6). Moreover, they have also reduced the incidence of stroke (5, 7), despite stroke having a less robust relationship with LDL levels than CHD. Statins have proved to be beneficial in reducing CVD risk, whatever the nature of the risk. Thus, CVD risk is reduced by statins in women, men, (5, 6), Caucasians (6), blacks (8, 9), people with diabetes (8, 10, 11), smokers, hypertensives, and in individuals with other lipoprotein-related risk factors such as hypertriglyceridemia, low high-density lipoprotein (HDL)-cholesterol (HDL-C) levels (12), and high levels of lipoprotein (a) (13). Nonetheless, as noted earlier, despite the reduction in CVD events and mortality that started in the late 1960s (14, 15), considerable residual risk remains and CVD remains the major cause of death in the developed world.

What can be done about this residual risk? One possibility is that LDL-cholesterol (LDL-C) levels achievable by the use of statins alone may not be low enough. Strategies to achieve even lower LDL levels are now available, including the addition of cholesterol absorption inhibitors such as ezetimibe, and the recently available PCSK9 antibodies, the introduction of which will allow testing of whether achieving extremely low LDL levels will further reduce CVD events, and also safety of achieving very low levels of LDL-C. In addition, new LDL drugs in the pipeline offer promise for additional LDL lowering, particularly in those individuals who are unable to tolerate statins.

Combination therapy of statins and cholesterol absorption inhibitors

Considerable evidence suggests that “the lower the better” is a reasonable approach for reducing CVD by lowering LDL levels (16, 17). One way of achieving very low levels of LDL cholesterol is by the combined use of a statin and a cholesterol absorption inhibitor. Currently, the only approved drug in this class is ezetimibe, the target of which is the enzyme NPC1L1, the rate-limiting enzyme in the absorption of both exogenous and endogenous cholesterol in the intestine (18). The recently completed Examining Outcomes in Subjects With Acute Coronary Syndrome: Vytorin (Ezetimibe/Simvastatin) vs Simvastatin (IMPROVE-IT) trial showed that reducing LDL to very low levels by the addition of ezetimibe to a statin resulted in the expected additional lowering of CVD events predicted by the known regression relationship between on-trial LDL-C levels and clinical endpoints (19). Of interest, this additional lowering resulted in levels of LDL-C of 53 mg/dl in combined ezetimibe/simvastatin arm vs 69 mg/dl in the simvastatin/placebo arm with an overall risk reduction of 8%. These findings add credence to the notion that lowering LDL by any means would likely have a beneficial effect on clinical outcomes (20), and that lowering LDL by statins alone is not a magic bullet. Thus, the addition of a cholesterol absorption inhibitor to a statin may prove to be a good way of getting LDL to lower levels, especially when this agent becomes generic in the near future.

Cholesterol absorption inhibitors in 5–10 years

It would be informative to know the relative benefit of the addition of a cholesterol absorption inhibitor such as ezetimibe to a statin in subjects whose LDL levels were not as low as was observed in the IMPROVE-IT trial. However, to the best of our knowledge, there are no such clinical trials ongoing or planned at present, so no new data from randomized clinical trials are likely to be available in the next 5–10 years. Thus the physician will have to rely on clinical judgement in his or her quest to achieve LDL-C levels as low as possible in suitable very high risk patients.

PCSK9 inhibitors

Another way of getting LDL to very low levels is by using the new PCSK9 antibodies. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a proprotein convertase originally identified at INSERM in Paris wherein mutations were associated with hypercholesterolemia (21, 22). The role of PCSK9 appears to direct LDL receptor toward lysosomal degradation rather than recycling to the plasma membrane, thereupon reducing the number and function of LDL receptors in mediating LDL clearance (23). Subsequently, both gain-of-function and loss-of-function mutations were identified that rapidly generated interest in pharmacotherapeutics, specifically human monoclonal antibodies directed against PCSK9 (24). In the summer of 2015, phase 3 clinical trials were sufficient in demonstrating LDL-C lowering in addition to safety for approval of alirocumab and evolocumab for LDL-C lowering. In fact, the highest doses of these drugs exceeded the reduction of LDL-C of maximum dose statins (25, 26). Currently, PCSK9 inhibitors are indicated as adjuncts to diet and maximally tolerated statin therapy for the treatment of adults with heterozygous familial hypercholesterolemia or clinical atherosclerotic cardiovascular disease, who require additional lowering of LDL-C. Although firm evidence from ongoing clinical trials is not yet available, a reduction in all-cause mortality, CVD mortality, and myocardial infarction has been reported from all the recent phase 2 and 3 randomized controlled trials (27).

PCSK9s and ASCVD in 5–10 years

For years now, LDL-C has been viewed by regulatory agencies as a surrogate for ASCVD. An unanswered question is whether this view holds at very low levels of LDL-C. The relationship between total cholesterol and CHD death is curvilinear, with less benefit with levels approaching 175 mg/dL (28). However, data from 27 randomized clinical trials suggest a linear relationship between the effect of LDL-C lowering with statins and reduced ASCVD events and related deaths from ASCVD (5). Data from IMPROVE-IT with ezetimibe + simvastatin (19) and post hoc data from Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER; rosuvastatin in patients with elevated highly sensitive C-reactive protein) (29) indicate that additional benefit ensues when LDL-C is reduced to less than 55 or 40 mg/dL, respectively. Presently, four large ASCVD outcome trials including an expected enrollment of more than 70,000 patients are ongoing, one each with alirocumab (ODYSSEY) and evolocumab (Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk) and two with bococizumab (SPIRE-1, SPIRE-2). Considering the doses of the PCSK9 inhibitors used in these trials, many high-risk patients will have recurrent LDL-C levels below 40 mg/dL and a significant minority below 25 mg/dL (30, 31). Although the early data suggest an ASCVD benefit (30, 31), only after completion of the trials will we learn whether a threshold level of LDL-C achieves maximum benefit.

One clue to the potential safety of very low LDL-C levels comes from individuals with hypobetalipoproteinemia. These individuals have a mutation in the apoB gene, which results in very low lifelong LDL-C levels (32, 33). Despite these low levels, there is no evidence of adverse health effects, including cancer, Alzheimer's disease, or any other chronic disease, other than for an increase in fatty liver (34), likely because of impaired ability to secrete hepatic triglyceride [TG]) in the form of very low-density lipoprotein (VLDL). This side effect is unlikely to occur with the use of PCSK9 inhibitors because of their different mechanism of action. Nonetheless, individuals with hypobetalipoproteinemia are characterized by longevity and protection against CVD (33), suggesting that lowering LDL to very low levels by pharmacological means is not likely to have adverse effects. However, the extremely low LDL and TG levels associated with mutations in the microsomal TG transport gene, abetalipoproteinemia (35), is associated with severe neurological abnormalities because of the inability to transport fat soluble vitamins (32, 36). Therefore, fatty liver and fat-soluble vitamin deficiencies will need to be carefully looked for in ongoing PCSK9 clinical trials and in posttrial surveillance. Other approaches to inhibit PCSK9, such as the use of antisense oligonucleotides and RNA interference approaches might well be used in the future.

ATP citrate lyase inhibition

Inhibition of PCSK9 requires the use of injectable compounds. However, another orally administered LDL-lowering compound, ETC-1002, a once-daily small molecule designed to lower levels of LDL-C and to avoid side effects associated with existing LDL-C lowering therapies (37), is entering phase 3 clinical trials. ETC-1002 inhibits ATP citrate lyase, a key enzyme that supplies substrate for cholesterol and fatty acid synthesis in the liver (38). In phase 2 clinical trials, it has been reported to lower LDL-C levels by approximately 20–40% (37, 39, 40). Because its use has not been associated with some of the side effects that have been attributed to statins, it may be of particular use in individuals who are intolerant of statins (40). The drug also inhibits AMP kinase (41). This inhibitory property was associated with a lowering of blood glucose levels in an early clinical trial (39).

ApoB antisense oligonucleotides and microsomal TG transport inhibitors

Two other approaches have been used to lower LDL levels in individuals with very high levels. The first uses an antisense approach to inhibit the production of apoB-100, the major apolipoprotein of VLDL and LDL. Mipomersen, an injectable apoB antisense oligonucleotide (42), is approved in the United States for the treatment of homozygous familial hypercholesterolemia. However, the extremely high cost and the fatty liver that result from its use is likely to preclude its more widespread use in the foreseeable future.

Lomitapide is an inhibitor of microsomal TG transport protein, necessary for chylomicron absorption in the intestine and VLDL assembly and secretion in the liver (43, 44). It too has been approved in the United States as an orphan drug for the treatment of homozygous familial hypercholesterolemia. Like mipomersen, it is extremely expensive and fatty liver results from it use. Also, steatorrhea can occur unless dietary fat is reduced to less than 20% of total caloric intake. These adverse effects also make it highly unlikely that this drug will enjoy more widespread use for the management of more common forms of hypercholesterolemia.

Role of HDL

Plasma levels of HDL-C are strongly and inversely related to the risk of CVD (45, 46). However, recent genetic studies that failed to show the expected benefit of polymorphisms that increase HDL-C levels (47) and some clinical trials that failed to show a benefit on CVD of raising HDL-C have cast doubt on the role of HDL as a cardioprotective lipoprotein. For example, elevation of HDL-C levels by cholesteryl ester transfer protein (CETP) inhibitors in the ILLUMINATE (Investigation of Lipid Level Management to Understand Its Impact in Atherosclerotic Events) trial failed to show the expected benefit on CVD end points, which in fact got worse with the use of torcetrapib. Torcetrapib, when added to atorvastatin, raised HDL-C levels by 72% and resulted in an additional 25% reduction in LDL-C levels compared to the group receiving atorvastatin alone, yet resulted in a significant excess in cardiovascular and noncardiovascular mortality (48). Although this worsening of CVD end points has been suggested to be due to an off target effect of the drug on increasing aldosterone levels and blood pressure (49), a clinical trial with dalcetrapib was discontinued because of futility (50), the development of evacetrapib has been halted, and only evacetrapib remains in an ongoing clinical trial. Moreover, two recent studies of the effect of the addition to statin therapy of niacin, which effectively raises HDL-C levels, have also been negative. In the Atherothrombosis Intervention in Metabolic Syndrome with low HDL/High Triglycerides: Impact on Global Health trial, the addition of extended release niacin to statin-treated subjects with very low baseline LDL-C levels on statins alone failed to show any additional benefit of niacin treatment (51). In the Treatment of HDL to Reduce the Incidence of Vascular Events trial, the addition to statin therapy of extended-release niacin in combination with laropiprant to inhibit flushing, also failed to confer clinical benefit beyond that observed in the statin-alone group (52). Moreover, the adverse effects of extended-release niacin were unacceptably high. These negative findings occurred despite niacin having been effective when used as monotherapy in the Coronary Drug Project in the prestatin era (53, 54). This raises the question of whether the benefit of niacin seen in that study was the result of niacin-induced LDL lowering.

Despite these negative genetic studies and clinical trials, studies in mice show a clear benefit on atherogenesis of increasing the level of HDL's major apolipoprotein, apoA-I (55, 56). Conversely, apoA-I deficiency confers increased risk of developing atherosclerosis in mice (57). Therefore, the relationship between HDL and ASCVD is more complex than hitherto appreciated. HDL is believed to protect against the development of atherosclerosis via it ability to stimulate reverse cholesterol transport from cells of the artery wall (5860). Moreover, HDL has several other potentially antiatherogenic properties, including its ability to inhibit inflammation, oxidation, endothelial activation, and thrombogenesis.

This raises the question of whether HDL-C, rather than some other measurement of HDL function, is the appropriate metric for assessing the effect of interventions on HDL. It is therefore of considerable interest that measurement of cholesterol efflux capacity from macrophages, a metric of HDL function, showed a strong inverse association with both carotid intima-media thickness and the likelihood of angiographic coronary artery disease, independently of HDL-C levels (61). Similarly, in the Dallas Heart Study, cholesterol efflux capacity was inversely associated with the incidence of cardiovascular events in a population-based cohort (62). In the prospective nested case-control European Prospective Investigation of Cancer-Norfolk study of more than 25,000 individuals, cholesterol efflux capacity was significantly and inversely associated with incident CHD events, independent of age, sex, diabetes, hypertension, smoking and alcohol use, waist:hip ratio, body mass index, LDL-C concentration, log-triglycerides, HDL-C, and apoA-I concentration (63). Measurement of cholesterol efflux capacity depends on an ex vivo cellular assay which will be difficult to standardize, is time-consuming and expensive, and is unlikely to be available for widespread clinical use in the near future. Research techniques that have been used to measure reverse cholesterol transport in vivo in mice (64) are beginning to be applied to humans, but also are unlikely to be clinically applicable in the foreseeable future.

HDL and apoA-I infusions

Epidemiological studies that showed that a genetic variant of apoA-I, apoA-I Milano, was associated with a reduced incidence of CHD (65, 66), were supported by preclinical studies that also showed the ability of apoA-I infusion to halt the progression and cause regression of atherosclerosis (67). This resulted in the development of a recombinant apoA-I Milano phospholipid complex for testing in clinical trials. Although there was rapid regression by IV ultrasound in a small number of subjects with unstable angina, no wild-type apoA-I control was included (67). However, progress in developing a product for clinical use appears to have stalled.

Several companies have been working on making apoA-I/phospholipid complexes for infusion in patients with acute coronary syndromes. The most promising is a product named CSL112 (68), which is being tested in ongoing clinical trials.

ApoA-I mimetics

Another approach to increase HDL levels and facilitate reverse cholesterol transport has been the use of apoA-I mimetics. Several apoA-I mimetic peptides, based on the helical structure of apoA-I, have been developed with the hope that they would mimic some of the antiatherogenic properties of HDL. To overcome the problem of intestinal digestion, some were synthesized with D amino acids, and recently they have been made by transgenic tomatoes (69). Several peptides have been shown to reduce atherosclerosis in animal models (7073), although the mechanisms by which they do so is unclear and may relate more to their antioxidative properties than to their role in reverse cholesterol transport (74). To date, these apoA-1 mimetics have not been used in clinical trials.

HDL and ASCVD in 5–10 years

The role of altering HDL by lifestyle and pharmacological means has proved more difficult to sort out than originally imagined. Yet on the basis of emerging data, especially in animal models, there is hope that by focusing on measures of HDL functionality, rather than HDL-C levels, a better understanding of potentially beneficial modalities of therapy can be achieved. Therefore, in the next 5–10 years we are likely to see additional validation of measures of HDL functionality that predict CVD outcomes, and their incorporation into clinical trials and ultimately into clinical practice. Application of such techniques may confirm that HDL-C raising strategies such as CETP inhibitors and niacin may be of little value in CVD prevention. The jury is still out regarding fibrates (see the following section), which in addition to lowering TG levels, also modestly increase HDL-C.

As noted earlier, there has been considerable recent interest in HDL and apoA-I infusions, and the use of apoA-I mimetics, although mainly for use in the setting of acute coronary syndrome. A more rational approach to long term elevation of HDL that can facilitate reverse cholesterol transport has been strategies to increase apoA-I production. The most promising approach to date has been the use of apabetalone or RVX-208, a small-molecule drug that increases apoA-I production by an epigenetic mechanism as a result of it being a bromodomain extraterminal protein inhibitor (75). It increases the production of prebeta HDL, a particle that is believed to initiate reverse cholesterol transport. However, in small phase II intravascular ultrasound clinical trials in CVD patients (ApoA-1 Synthesis Stimulation and Intravascular Ultrasound for Coronary Atheroma Regression Evaluation), RVX-208 failed to reduce atheroma volume significantly; however, it also failed to raise HDL-C to a great extent (76, 77). Nonetheless, this compound will be entering phase 3 clinical trials in the near future. It is likely that we will see additional approaches along these lines during the next 5–10 years, and will hopefully, for once and for all, be able to answer the question of whether therapeutic approaches that target HDL and reverse cholesterol transport have merit.

Role of TG

TGs are mostly carried in chylomicrons, VLDL, and chylomicron and VLDL remnants. It has been well known for decades that hypertriglyceridemia is associated with increased CVD risk, specifically CHD in the early years (78, 79), but this risk is influenced in part by plasma levels of HDL that confer an independent and inverse relationship to CVD (80, 81). Although smaller TG-rich particles can cross the subintimal space and be found in atherosclerotic plaque (82), most plaque lipid is cholesteryl ester not TG. Moreover, still uncertain is whether the association of hypertriglyceridemia with CVD is causative or mediated indirectly by lipoprotein lipase (LPL)-mediated release of fatty acids or other toxins from TG-rich lipoproteins. Another mechanism is the adverse metabolic company that hypertriglyceridemia keeps with abdominal obesity, glucose intolerance, hypertension, and other components of the insulin resistant or metabolic syndrome (83, 84). In addition, increasing evidence suggests that nonfasting TG may be as if not more informative about CVD risk than fasting values (85, 86) and that higher levels of apoC-III, an apolipoprotein bound to TG-rich lipoproteins in addition to other particles, relates to atherogenesis (87). Proof of concept that hypertriglyceridemia is an independent predictor of CVD should be based on clinical trials. Pharmacological agents that work mostly on lowering fasting and nonfasting TG are the fibrates and omega-3 fatty acids.

Fibrates.

Fibrates lower plasma TG by peroxisome proliferator-activated receptor-α–mediated effects on fatty acid oxidation diverting fatty acid incorporation away from VLDL-TG synthesis and secretion (88). Although the earliest fibrate trials were done with clofibrate, it was gemfibrozil that proved to reduce CVD events in the Helsinki Heart Study (89); however, this effect was mostly seen in patients who had very high levels of LDL and higher TG levels, a setting in which both TG and LDL-C were lowered (90). In the Veterans Administration HDL Intervention Trial (VA-HIT) gemfibrozil vs placebo was given to men with low levels of HDL-C and existing CVD, and in this study events were reduced by 25% in the absence of changes in LDL-C (91). Of note, however, the amount of lowering of plasma TG with gemfibrozil did not correlate with reduction of risk (92). The Bezafibrate Infarction Prevention Study (93) and Fenofibrate Intervention on Endpoint Lowering in Diabetes Study (94) were both negative studies but in the latter, there was a substantial statin drop-in in the placebo group. When the data were examined post hoc, subgroups of patients with plasma TG higher than 200 mg/dL and TG at least 204 mg/dL plus HDL-C lower than 42 mg/dL, demonstrated benefit (P < .02) or borderline benefit (P = .07) respectively. In the Action to Control Cardiovascular Risk in Diabetes Lipid Study in which patients with type 2 diabetes were randomized to fenofibrate vs placebo on top of statin therapy, there was no benefit on the primary outcome but again, a subgroup analysis in patients with a fasting TG of at least 204 mg/dL and HDL-C lower than 34 mg/dL hinted at benefit, P = .057 (95).

Omega-3 fatty acids.

Compared to fibrates, omega-3 fatty acids have been infrequently employed in ASCVD outcome trials, in part related to the current indications reserved for patients with TG at least 500 mg/dL. Although Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardico-Prevenzione Trial was a low-dose eicosapentaenoic acid (EPA) (1.0 g) monotherapy trial in patients with a myocardial infarction that demonstrated a positive outcome on the primary CVD end point including all-cause mortality (96), the most frequently cited study is a trial done in Japan, the Jelis Study (97). This study enrolled more than 18 000 subjects (2/3 women) with a balance between those with and without existing ASCVD and baseline total cholesterol higher than 6.5 mmol/liter (575 mg/dl). Patients were all treated with low-dose statin ± 1.8 g of EPA, with a 4.6-year follow-up demonstrating a significant benefit in the total population, but only for secondary prevention. When a subgroup of patients with TG higher than 150 and HDL-C lower than 40 mg/dL was examined post hoc, a 53% reduction was seen (98). And from meta-analyses, other than a borderline benefit for cardiac death, there appears to be no benefit for omega-3 supplementation in patients with existing ASCVD (99).

Triglycerides and ASCVD in 5–10 years

At present, the optimal trial for TG lowering with a favorable CVD outcome awaits us. Such a trial would enroll patients with a previous CVD event or those at very high risk for such event, already treated with a statin (100) and with fasting TG between 200 and 500 mg/dl. For fibrates, the Veterans Administration Fenofibrate Intervention Trial has been planned and approved but not yet funded. Two such trials using omega-3 fatty acids are ongoing, the Reduction of Cardiovascular Events with EPA-Intervention Trial using ethyl-EPA (101) and STRENGTH (Statin Residual Risk Reduction with EpaNova in High Risk Patients with Hypertriglyceridemia) using omega-3 carboxylic acids. Because patients with increases in apoC-III are at increased risk of CVD and loss-of-function mutations of apoC-III are associated with a reduced risk of CVD (102, 103), new therapies directed at apoC-III may be another pathway to lower CVD risk related to hypertriglyceridemia. The relationship between apoC-III and hypertriglyceridemia has been attributed to inhibition of LPL-mediated lipolysis of circulating TG-rich lipoproteins (104); however, alternative mechanisms related to hepatic processing of chylomicron and VLDL remnants has also been raised (105). Antisense inhibition of apoC-III in hypertriglyceridemic patients with or without ongoing statin or fibrate therapy has been recently shown to reduce fasting TG by 40–80% in a dose-dependent manner (106). Another promising area for TG lowering is the relationship of angiopoietins to TG-rich lipoprotein metabolism. Angiopoietin-3 and angiopoietin-4 have been shown to inhibit LPL (107, 108). Yet to be replicated in humans are results from hypertriglyceridemic mice indicating that an antibody directed against angiopoietin-4 can lower fasting TG (109). Another consideration is either dual peroxisome proliferator-activated receptor-α/λ or peroxisome proliferator-activated receptor-α/δ agonists which result in moderate TG lowering (110); however, this class of agents has been delayed for approval based on adverse effects. Whichever novel therapy proves safe and effective in TG lowering and CVD outcomes to follow, there are a substantial number of patients with hypertriglyceridemia with or without statin therapy and with or without the metabolic syndrome who would benefit. The most recent National Health and Nutrition Examination Study data indicate that 31% of Americans have fasting TG higher than 150 mg/dL and 16% are above 200 mg/dL (111).

Lipoprotein (a)

Lipoprotein (a) is an apoB-containing lipoprotein that includes apolipoprotein (a) covalently bound to apoB, wherein plasma concentrations are determined mostly by genetics. The size of the apo (a) isoform is dependent on a variable number of kringle IV repeats with fewer repeats predicting a higher level of plasma lipoprotein (a) (112). Lipoprotein (a) concentrations vary substantially, with a several-fold higher level in populations of African and Asian descent (113). Increases in lipoprotein (a) and CVD have been repeatedly documented (114) and a threshold of plasma levels higher than 30–50 mg/dL indicates an increased risk of CVD (115). Although this view of risk has been held for years, data from the Copenhagen Heart Study has suggested a gradient relationship between levels of lipoprotein (a) and CVD (116, 117). Moreover, in JUPITER, levels of lipoprotein (a) at baseline were not only associated with added CVD risk, in Caucasian statin-treated patients, higher levels predicted more events (13).

Lipoprotein (a) synthesis occurs mostly in the liver but some binding of the apo(a) protein to LDL apoB may occur in plasma (118). Elevation in lipoprotein (a) particles relate to CVD by their inability to be cleared by the LDL receptor, more atherogenic proinflammatory properties mediated by oxidized phospholipids, and antifibrinolytic activity secondary to structural homology to plasminogen (119).

Several approved drugs and one procedure lower lipoprotein (a): niacin, mipomersen, estrogens, and LDL apheresis (120); however, currently there are no data demonstrating independent effects of these interventions that reduce lipoprotein (a) results on CVD events. Of interest, in the Women's Health Initiative, estrogens may have conferred benefit on CVD events in postmenopausal women with the highest quintile of lipoprotein (a); and yet this outcome has been mostly ignored (121).

Lipoprotein (a) and CVD in 5–10 years

A promise for the future is antisense technology. phase 1 data have revealed a dose-dependent 40–80% reduction in lipoprotein (a) using ISIS-APO(a)RX (122).

When phase 2 antisense or alternative strategies for lipoprotein (a) lowering are completed, the definitive truth of the antiatherogenic effects of lipoprotein (a) lowering will require a long-term outcome study in patients with elevated lipoprotein (a) levels who are at high CVD risk and on statins and other evidence-based risk reduction therapies, a result that may be forthcoming by 10 years.

Table 1 provides some predictions of where the science of each of these four lipoprotein classes will be in 5–10 years.

Table 1.

Predictions for 5–10 Years Hence

LipoproteinPrediction
LDL1. PCSK9 trials will lower ASCVD events but no added effect will exist at LDL-C ≤40 mg/dl; the FDA may no longer consider LDL as a surrogate for ASCVD.
2. ETC-1002 will be approved but no outcome trials will be done.
3. ACC/AHA and other cholesterol guidelines will be updated.
4. Lomitapide will no longer be marketed.
HDL1. CETP inhibitor outcome trials will be negative.
2. Measures of HDL function will be optimized and made clinically available.
3. ApoA-1 mimetics and drugs to increase apoA-1 synthesis will be further developed but not FDA approved.
TG-rich lipoproteins (VLDL, chylomicrons)1. The optimal fibrate trial in patients with fasting TG ≥200 but ≤500 mg/dl will demonstrate a reduction in CVD events in high-risk patients on maximum tolerated statin therapy and a new FDA indication will follow.
2. The omega-3 fatty acids trial in patients with fasting TG ≥200 but ≤500 mg/dL in high-risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
3. Antisense ApoC-III in high risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
4. Antisense ApoC-III will also be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
5. LPL gene therapy will be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
6. Antibodies and antisense molecules that inhibit angiopoietin-4 will be further tested.
Lipoprotein (a)1. Antisense lipoprotein (a) strategies to reduce ASCVD risk will be extended to phase 3 outcome trials and be FDA approved for this indication.
LipoproteinPrediction
LDL1. PCSK9 trials will lower ASCVD events but no added effect will exist at LDL-C ≤40 mg/dl; the FDA may no longer consider LDL as a surrogate for ASCVD.
2. ETC-1002 will be approved but no outcome trials will be done.
3. ACC/AHA and other cholesterol guidelines will be updated.
4. Lomitapide will no longer be marketed.
HDL1. CETP inhibitor outcome trials will be negative.
2. Measures of HDL function will be optimized and made clinically available.
3. ApoA-1 mimetics and drugs to increase apoA-1 synthesis will be further developed but not FDA approved.
TG-rich lipoproteins (VLDL, chylomicrons)1. The optimal fibrate trial in patients with fasting TG ≥200 but ≤500 mg/dl will demonstrate a reduction in CVD events in high-risk patients on maximum tolerated statin therapy and a new FDA indication will follow.
2. The omega-3 fatty acids trial in patients with fasting TG ≥200 but ≤500 mg/dL in high-risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
3. Antisense ApoC-III in high risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
4. Antisense ApoC-III will also be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
5. LPL gene therapy will be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
6. Antibodies and antisense molecules that inhibit angiopoietin-4 will be further tested.
Lipoprotein (a)1. Antisense lipoprotein (a) strategies to reduce ASCVD risk will be extended to phase 3 outcome trials and be FDA approved for this indication.

Abbreviations: ACC, American College of Cardiology; AHA, American Heart Association; FDA, Food and Drug Administration.

Table 1.

Predictions for 5–10 Years Hence

LipoproteinPrediction
LDL1. PCSK9 trials will lower ASCVD events but no added effect will exist at LDL-C ≤40 mg/dl; the FDA may no longer consider LDL as a surrogate for ASCVD.
2. ETC-1002 will be approved but no outcome trials will be done.
3. ACC/AHA and other cholesterol guidelines will be updated.
4. Lomitapide will no longer be marketed.
HDL1. CETP inhibitor outcome trials will be negative.
2. Measures of HDL function will be optimized and made clinically available.
3. ApoA-1 mimetics and drugs to increase apoA-1 synthesis will be further developed but not FDA approved.
TG-rich lipoproteins (VLDL, chylomicrons)1. The optimal fibrate trial in patients with fasting TG ≥200 but ≤500 mg/dl will demonstrate a reduction in CVD events in high-risk patients on maximum tolerated statin therapy and a new FDA indication will follow.
2. The omega-3 fatty acids trial in patients with fasting TG ≥200 but ≤500 mg/dL in high-risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
3. Antisense ApoC-III in high risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
4. Antisense ApoC-III will also be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
5. LPL gene therapy will be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
6. Antibodies and antisense molecules that inhibit angiopoietin-4 will be further tested.
Lipoprotein (a)1. Antisense lipoprotein (a) strategies to reduce ASCVD risk will be extended to phase 3 outcome trials and be FDA approved for this indication.
LipoproteinPrediction
LDL1. PCSK9 trials will lower ASCVD events but no added effect will exist at LDL-C ≤40 mg/dl; the FDA may no longer consider LDL as a surrogate for ASCVD.
2. ETC-1002 will be approved but no outcome trials will be done.
3. ACC/AHA and other cholesterol guidelines will be updated.
4. Lomitapide will no longer be marketed.
HDL1. CETP inhibitor outcome trials will be negative.
2. Measures of HDL function will be optimized and made clinically available.
3. ApoA-1 mimetics and drugs to increase apoA-1 synthesis will be further developed but not FDA approved.
TG-rich lipoproteins (VLDL, chylomicrons)1. The optimal fibrate trial in patients with fasting TG ≥200 but ≤500 mg/dl will demonstrate a reduction in CVD events in high-risk patients on maximum tolerated statin therapy and a new FDA indication will follow.
2. The omega-3 fatty acids trial in patients with fasting TG ≥200 but ≤500 mg/dL in high-risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
3. Antisense ApoC-III in high risk patients on maximum tolerated statin therapy will also demonstrate ASCVD risk reduction and a new FDA indication will follow.
4. Antisense ApoC-III will also be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
5. LPL gene therapy will be approved for the reduction in pancreatitis incidence in patients with severe hypertriglyceridemia.
6. Antibodies and antisense molecules that inhibit angiopoietin-4 will be further tested.
Lipoprotein (a)1. Antisense lipoprotein (a) strategies to reduce ASCVD risk will be extended to phase 3 outcome trials and be FDA approved for this indication.

Abbreviations: ACC, American College of Cardiology; AHA, American Heart Association; FDA, Food and Drug Administration.

Gene therapy

This use of this approach is likely to be limited to genetic conditions in which a mutation in a single gene results in a difficult to treat form of hyperlipidemia. Gene therapy for LPL deficiency, using an adeno-associated viral vector (123), is currently approved for this rare condition in Europe, but not yet in the United States. Replacement of the LDL receptor gene for homozygous familial hypercholesterolemia was attempted several years ago, but failed as a result of problems with the viral vector used. Conceivably, gene therapy for this condition might be tried again in the next several years, now that better viral vectors are available (124). Gene therapy with apoA-I, especially with apoA-I Milano, has also been suggested as a possible way of increasing functional HDL and facilitating reverse cholesterol transport. However, this approach awaits much more information regarding the role of HDL in atherosclerosis prevention.

Acknowledgments

Disclosure Summary: A.C. has consulted for Merck, ISIS, Takeda, and Tekmira. R.H.E. is a consultant for ISIS Pharmaceuticals, Regeneron/Sanofi, and Janssen. He is on the advisory board for ISIS Pharmaceuticals and has received grants from ISIS Pharmaceuticals and Janssen.

Abbreviations

     
  • ASCVD

    atherosclerotic cardiovascular disease

  •  
  • CETP

    cholesteryl ester transfer protein

  •  
  • CHD

    coronary heart disease

  •  
  • CVD

    cardiovascular disease

  •  
  • EPA

    eicosapentaenoic acid

  •  
  • HDL

    high-density lipoprotein

  •  
  • HDL-C

    high-density lipoprotein-cholesterol

  •  
  • LDL

    low-density lipoprotein

  •  
  • LDL-C

    low-density lipoprotein-cholesterol

  •  
  • LPL

    lipoprotein lipase

  •  
  • TG

    triglyceride

  •  
  • VLDL

    very low-density lipoprotein.

References

1.

Keys
A
.
Cholesterol, “giant molecules,” and atherosclerosis
.
J Am Med Assoc
.
1951
;
147
:
1514
1519
.

2.

Gofman
JW
,
Lindgren
F
.
The role of lipids and lipoproteins in atherosclerosis
.
Science
.
1950
;
111
:
166
171
.

3.

Bilheimer
DW
,
Grundy
SM
,
Brown
MS
,
Goldstein
JL
.
Mevinolin stimulates receptor-mediated clearance of low density lipoprotein from plasma in familial hypercholesterolemia heterozygotes
.
Trans Assoc Am Physicians
.
1983
;
96
:
1
9
.

4.

Marais
AD
,
Naoumova
RP
,
Firth
JC
,
Penny
C
,
Neuwirth
CK
,
Thompson
GR
.
Decreased production of low density lipoprotein by atorvastatin after apheresis in homozygous familial hypercholesterolemia
.
J Lipid Res
.
1997
;
38
:
2071
2078
.

5.

Cholesterol Treatment Trialists (CTT) Collaborators
,
Mihaylova
B
,
Emberson
J
,
Blackwell
L
, et al. 
.
The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials
.
Lancet
.
2012
;
380
:
581
590
.

6.

Cholesterol Treatment Trialists (CTT) Collaborators
,
Fulcher
J
,
O'Connell
R
,
Voysey
M
, et al. 
.
Efficacy and safety of LDL-lowering therapy among men and women: meta-analysis of individual data from 174,000 participants in 27 randomised trials
.
Lancet
.
2015
;
385
:
1397
1405
.

7.

Amarenco
P
.
Effect of statins in stroke prevention
.
Curr Opin Lipidol
.
2005
;
16
:
614
618
.

8.

Morrone
D
,
Weintraub
WS
,
Toth
PP
, et al. .
Lipid-altering efficacy of ezetimibe plus statin and statin monotherapy and identification of factors associated with treatment response: a pooled analysis of over 21,000 subjects from 27 clinical trials
.
Atherosclerosis
.
2012
;
223
:
251
261
.

9.

Margolis
KL
,
Dunn
K
,
Simpson
LM
, et al. .
Coronary heart disease in moderately hypercholesterolemic, hypertensive black and non-black patients randomized to pravastatin versus usual care: the antihypertensive and lipid lowering to prevent heart attack trial (ALLHAT-LLT)
.
Am Heart J
.
2009
;
158
:
948
955
.

10.

Pyörälä
K
,
Pedersen
TR
,
Kjekshus
J
,
Faergeman
O
,
Olsson
AG
,
Thorgeirsson
G
.
Cholesterol lowering with simvastatin improves prognosis of diabetic patients with coronary heart disease. A subgroup analysis of the Scandinavian Simvastatin Survival Study (4S) [published erratum appears in Diabetes Care 1997 Jun;20(6):1048]
.
Diabetes Care
.
1997
;
20
:
614
620
.

11.

Haffner
S
,
Alexander
C
,
Cook
T
, et al. .
Reduced coronary events in simvastatin-treated subjects with coronary heart disease and diabetes or impaired fasting glucose: findings from the Scandinavian Simvastatin Survival Study (4S)
.
Arch Int Med
.
1999
;
159
:
2661
2667
.

12.

Scandinavian Simvastatin Survival Study Group
.
Randomized trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S)
.
Lancet
.
1994
;
344
:
1383
1389
.

13.

Khera
AV
,
Everett
BM
,
Caulfield
MP
, et al. .
Lipoprotein(a) concentrations, rosuvastatin therapy, and residual vascular risk: an analysis from the JUPITER Trial (Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin)
.
Circulation
.
2014
;
129
:
635
642
.

14.

Cooper
R
,
Cutler
J
,
Desvigne-Nickens
P
, et al. .
Trends and disparities in coronary heart disease, stroke, and other cardiovascular diseases in the United States: findings of the national conference on cardiovascular disease prevention
.
Circulation
.
2000
;
102
:
3137
3147
.

15.

O'Flaherty
M
,
Buchan
I
,
Capewell
S
.
Contributions of treatment and lifestyle to declining CVD mortality: why have CVD mortality rates declined so much since the 1960s?
Heart
.
2013
;
99
:
159
162
.

16.

Wiviott
SD
,
Cannon
CP
,
Morrow
DA
, et al. .
Can low-density lipoprotein be too low? The safety and efficacy of achieving very low low-density lipoprotein with intensive statin therapy: a PROVE IT-TIMI 22 substudy
.
J Am Coll Cardiol
.
2005
;
46
:
1411
1416
.

17.

O'Keefe
JH
Jr.,
Cordain
L
,
Harris
WH
,
Moe
RM
,
Vogel
R
.
Optimal low-density lipoprotein is 50 to 70 mg/dl: lower is better and physiologically normal
.
J Am Coll Cardiol
.
2004
;
43
:
2142
2146
.

18.

Garcia-Calvo
M
,
Lisnock
J
,
Bull
HG
, et al. .
The target of ezetimibe is Niemann-Pick C1-Like 1 (NPC1L1)
.
Proc Natl Acad Sci U S A
.
2005
;
102
:
8132
8137
.

19.

Cannon
CP
,
Blazing
MA
,
Giugliano
RP
, et al. ;
IMPROVE-IT Investigators
.
Ezetimibe added to statin therapy after acute coronary syndromes
.
N Engl J Med
.
2015
;
372
:
2387
2397
.

20.

Jarcho
JA
,
Keaney
JF
Jr
.
Proof that lower is better–LDL cholesterol and IMPROVE-IT
.
N Engl J Med
.
2015
;
372
:
2448
2450
.

21.

Seidah
NG
,
Benjannet
S
,
Wickham
L
, et al. .
The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation
.
Proc Natl Acad Sci U S A
.
2003
;
100
:
928
933
.

22.

Abifadel
M
,
Varret
M
,
Rabes
JP
, et al. .
Mutations in PCSK9 cause autosomal dominant hypercholesterolemia
.
Nat Genet
.
2003
;
34
:
154
156
.

23.

Poirier
S
,
Mayer
G
,
Benjannet
S
, et al. .
The proprotein convertase PCSK9 induces the degradation of low density lipoprotein receptor (LDLR) and its closest family members VLDLR and ApoER2
.
J Biol Chem
.
2008
;
283
:
2363
2372
.

24.

Poirier
S
,
Mayer
G
.
The biology of PCSK9 from the endoplasmic reticulum to lysosomes: new and emerging therapeutics to control low-density lipoprotein cholesterol
.
Drug Des Devel Ther
.
2013
;
7
:
1135
1148
.

25.

Colhoun
HM
,
Robinson
JG
,
Farnier
M
, et al. .
Efficacy and safety of alirocumab, a fully human PCSK9 monoclonal antibody, in high cardiovascular risk patients with poorly controlled hypercholesterolemia on maximally tolerated doses of statins: rationale and design of the ODYSSEY COMBO I and II trials
.
BMC Cardiovasc Disord
.
2014
;
14
:
121
.

26.

Koren
MJ
,
Scott
R
,
Kim
JB
, et al. .
Efficacy, safety, and tolerability of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 as monotherapy in patients with hypercholesterolaemia (MENDEL): a randomised, double-blind, placebo-controlled, phase 2 study
.
Lancet
.
2012
;
380
:
1995
2006
.

27.

Navarese
EP
,
Kolodziejczak
M
,
Schulze
V
, et al. .
Effects of proprotein convertase subtilisin/kexin type 9 antibodies in adults with hypercholesterolemia: a systematic review and meta-analysis
.
Ann Intern Med
.
2015
;
163
:
40
51
.

28.

Martin
MJ
,
Hulley
SB
,
Browner
WS
,
Kuller
LH
,
Wentworth
D
.
Serum cholesterol, blood pressure, and mortality: implications from a cohort of 361,662 men
.
Lancet
.
1986
;
2
:
933
936
.

29.

Hsia
J
,
MacFadyen
JG
,
Monyak
J
,
Ridker
PM
.
Cardiovascular event reduction and adverse events among subjects attaining low-density lipoprotein cholesterol <50 mg/dl with rosuvastatin. The JUPITER trial (Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin)
.
J Am Coll Cardiol
.
2011
;
57
:
1666
1675
.

30.

Robinson
JG
,
Farnier
M
,
Krempf
M
, et al. ;
Investigators ODYSSEY LONG TERM
.
Efficacy and safety of alirocumab in reducing lipids and cardiovascular events
.
N Engl J Med
.
2015
;
372
:
1489
1499
.

31.

Sabatine
MS
,
Giugliano
RP
,
Wiviott
SD
, et al. ;
Open-Label Study of Long-Term Evaluation against LDL Cholesterol (OSLER) Investigators
.
Efficacy and safety of evolocumab in reducing lipids and cardiovascular events
.
N Engl J Med
.
2015
;
372
:
1500
1509
.

32.

Lee
J
,
Hegele
RA
.
Abetalipoproteinemia and homozygous hypobetalipoproteinemia: a framework for diagnosis and management
.
J Inherit Metab Dis
.
2014
;
37
:
333
339
.

33.

Kostis
WJ
.
How low an LDL-C should we go with statin therapy?
Curr Atheroscler Rep
.
2014
;
16
:
388
.

34.

Welty
FK
.
Hypobetalipoproteinemia and abetalipoproteinemia
.
Curr Opin Lipidol
.
2014
;
25
:
161
168
.

35.

Berriot-Varoqueaux
N
,
Aggerbeck
LP
,
Samson-Bouma
M
,
Wetterau
JR
.
The role of the microsomal triglygeride transfer protein in abetalipoproteinemia
.
Annu Rev Nutr
.
2000
;
20
:
663
697
.

36.

Kayden
HJ
.
Abetalipoproteinemia
.
Annu Rev Med
.
1972
;
23
:
285
296
.

37.

Ballantyne
CM
,
Davidson
MH
,
Macdougall
DE
, et al. .
Efficacy and safety of a novel dual modulator of adenosine triphosphate-citrate lyase and adenosine monophosphate-activated protein kinase in patients with hypercholesterolemia: results of a multicenter, randomized, double-blind, placebo-controlled, parallel-group trial
.
J Am Coll Cardiol
.
2013
;
62
:
1154
1162
.

38.

Lemus
HN
,
Mendivil
CO
.
Adenosine triphosphate citrate lyase: emerging target in the treatment of dyslipidemia
.
J Clin Lipidol
.
2015
;
9
:
384
389
.

39.

Gutierrez
MJ
,
Rosenberg
NL
,
Macdougall
DE
, et al. .
Efficacy and safety of ETC-1002, a novel investigational low-density lipoprotein-cholesterol-lowering therapy for the treatment of patients with hypercholesterolemia and type 2 diabetes mellitus
.
Arterioscler Thromb Vasc Biol
.
2014
;
34
:
676
683
.

40.

Thompson
PD
,
Rubino
J
,
Janik
MJ
, et al. .
Use of ETC-1002 to treat hypercholesterolemia in patients with statin intolerance
.
J Clin Lipidol
.
2015
;
9
:
295
304
.

41.

Pinkosky
SL
,
Filippov
S
,
Srivastava
RA
, et al. .
AMP-activated protein kinase and ATP-citrate lyase are two distinct molecular targets for ETC-1002, a novel small molecule regulator of lipid and carbohydrate metabolism
.
J Lipid Res
.
2013
;
54
:
134
151
.

42.

Kastelein
JJ
,
Wedel
MK
,
Baker
BF
, et al. .
Potent reduction of apolipoprotein B and low-density lipoprotein cholesterol by short-term administration of an antisense inhibitor of apolipoprotein B
.
Circulation
.
2006
;
114
:
1729
1735
.

43.

Cuchel
M
,
Bloedon
LT
,
Szapary
PO
, et al. .
Inhibition of microsomal triglyceride transfer protein in familial hypercholesterolemia
.
N Engl J Med
.
2007
;
356
:
148
156
.

44.

Cuchel
M
,
Rader
DJ
.
Microsomal transfer protein inhibition in humans
.
Curr Opin Lipidol
.
2013
;
24
:
246
250
.

45.

Gordon
DJ
,
Rifkind
BM
.
High-density lipoprotein–the clinical implications of recent studies
.
N Engl J Med
.
1989
;
321
:
1311
1316
.

46.

Miller
GJ
,
Miller
NE
.
Plasma high density lipoprotein concentration and development of ischaemic heart disease
.
Lancet
.
1974
;
1
:
16
20
.

47.

Voight
BF
,
Peloso
GM
,
Orho-Melander
M
, et al. .
Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study
.
Lancet
.
2012
;
380
:
572
580
.

48.

Barter
PJ
,
Caulfield
M
,
Eriksson
M
, et al. ;
ILLUMINATE Investigators
.
Effects of torcetrapib in patients at high risk for coronary events
.
N Engl J Med
.
2007
;
357
:
2109
2122
.

49.

Forrest
MJ
,
Bloomfield
D
,
Briscoe
RJ
, et al. .
Torcetrapib-induced blood pressure elevation is independent of CETP inhibition and is accompanied by increased circulating levels of aldosterone
.
Br J Pharmacol
.
2008
;
154
:
1465
1473
.

50.

Schwartz
GG
,
Olsson
AG
,
Abt
M
, et al. .
Effects of dalcetrapib in patients with a recent acute coronary syndrome
.
N Engl J Med
2012
;
367
:
2089
2099
.

51.

Investigators
A-H
.
The role of niacin in raising high-density lipoprotein cholesterol to reduce cardiovascular events in patients with atherosclerotic cardiovascular disease and optimally treated low-density lipoprotein cholesterol rationale and study design. The Atherothrombosis Intervention in Metabolic syndrome with low HDL/high triglycerides: Impact on Global Health outcomes (AIM-HIGH)
.
Am Heart J
.
2011
;
161
:
471
477
.

52.

HPS2-THRIVE Collaborative Group
,
Landray
MJ
,
Haynes
R
,
Hopewell
JC
, et al. 
.
Effects of extended-release niacin with laropiprant in high-risk patients
.
N Engl J Med
.
2014
;
371
:
203
212
.

53.

Canner
PL
,
Berge
KG
,
Wenger
NK
, et al. .
Fifteen year mortality in Coronary Drug Project patients: long-term benefit with niacin
.
J Am Coll Cardiol
.
1986
;
8
:
1245
1255
.

54.

Coronary Drug Project report on clofibrate and niacin
.
Atherosclerosis
.
1978
;
30
:
239
240
.

55.

Rubin
EM
,
Ishida
BY
,
Clift
SM
,
Krauss
RM
.
Expression of human apolipoprotein A-I in transgenic mice results in reduced plasma levels of murine apolipoprotein A-I and the appearance of two new high density lipoprotein size subclasses
.
Proc Natl Acad Sci U S A
.
1991
;
88
:
434
438
.

56.

Tangirala
RK
,
Tsukamoto
K
,
Chun
SH
,
Usher
D
,
Pure
E
,
Rader
DJ
.
Regression of atherosclerosis induced by liver-directed gene transfer of apolipoprotein A-I in mice
.
Circulation
.
1999
;
100
:
1816
1822
.

57.

Hughes
SD
,
Verstuyft
J
,
Rubin
EM
.
HDL deficiency in genetically engineered mice requires elevated LDL to accelerate atherogenesis
.
Arterioscler Thromb Vasc Biol
.
1997
;
17
:
1725
1729
.

58.

Roheim
PS
.
Atherosclerosis and lipoprotein metabolism: role of reverse cholesterol transport
.
Am J Cardiol
1986
;
57
:
3C
10C
.

59.

Badimon
JJ
,
Fuster
V
,
Badimon
L
.
Role of high density lipoproteins in the regression of atherosclerosis
.
Circulation
.
1992
;
86
:
III86
III94
.

60.

Rosenson
RS
,
Brewer
HB
Jr.
,
Davidson
WS
, et al. .
Cholesterol efflux and atheroprotection: advancing the concept of reverse cholesterol transport
.
Circulation
.
2012
;
125
:
1905
1919
.

61.

Khera
AV
,
Cuchel
M
,
de la Llera-Moya
M
, et al. .
Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis
.
N Engl J Med
.
2011
;
364
:
127
135
.

62.

Rohatgi
A
,
Khera
A
,
Berry
JD
, et al. .
HDL cholesterol efflux capacity and incident cardiovascular events
.
N Engl J Med
.
2014
;
371
:
2383
2393
.

63.

Saleheen
D
,
Scott
R
,
Javad
S
, et al. .
Association of HDL cholesterol efflux capacity with incident coronary heart disease events: a prospective case-control study
.
Lancet Diabetes Endocrinol
.
2015
;
3
:
507
513
.

64.

Zhang
Y
,
Zanotti
I
,
Reilly
MP
,
Glick
JM
,
Rothblat
GH
,
Rader
DJ
.
Overexpression of apolipoprotein A-I promotes reverse transport of cholesterol from macrophages to feces in vivo
.
Circulation
.
2003
;
108
:
661
663
.

65.

Calabresi
L
,
Franceschini
G
.
High density lipoprotein and coronary heart disease: insights from mutations leading to low high density lipoprotein
.
Curr Opin Lipidol
.
1997
;
8
:
219
224
.

66.

Gualandri
V
,
Franceschini
G
,
Sirtori
CR
, et al. .
Almilano apoprotein: identification of the complete kindred and evidence of a dominant genetic transmission
.
Am J Hum Genet
.
1985
;
37
:
1083
1097
.

67.

Nissen
SE
,
Tsunoda
T
,
Tuzcu
EM
, et al. .
Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial
.
JAMA
.
2003
;
290
:
2292
2300
.

68.

Gille
A
,
Easton
R
,
D'Andrea
D
,
Wright
SD
,
Shear
CL
.
CSL112 enhances biomarkers of reverse cholesterol transport after single and multiple infusions in healthy subjects
.
Arterioscler Thromb Vasc Biol
.
2014
;
34
:
2106
2114
.

69.

Chattopadhyay
A
,
Navab
M
,
Hough
G
, et al. .
A novel approach to oral apoA-I mimetic therapy
.
J Lipid Res
.
2013
;
54
:
995
1010
.

70.

Navab
M
,
Anantharamaiah
GM
,
Hama
S
, et al. .
Oral administration of an Apo A-I mimetic Peptide synthesized from D-amino acids dramatically reduces atherosclerosis in mice independent of plasma cholesterol
.
Circulation
.
2002
;
105
:
290
292
.

71.

Li
X
,
Chyu
KY
,
Faria Neto
JR
,
Yano
J
, et al. .
Differential effects of apolipoprotein A-I-mimetic peptide on evolving and established atherosclerosis in apolipoprotein E-null mice
.
Circulation
.
2004
;
110
:
1701
1705
.

72.

Amar
MJ
,
D'Souza
W
,
Turner
S
, et al. .
5A apolipoprotein mimetic peptide promotes cholesterol efflux and reduces atherosclerosis in mice
.
J Pharmacol Exp Ther
.
2010
;
334
:
634
641
.

73.

Wool
GD
,
Cabana
VG
,
Lukens
J
, et al. .
4F Peptide reduces nascent atherosclerosis and induces natural antibody production in apolipoprotein E-null mice
.
FASEB J
.
2011
;
25
:
290
300
.

74.

Getz
GS
,
Reardon
CA
.
The structure/function of apoprotein A-I mimetic peptides: an update
.
Curr Opin Endocrinol Diabetes Obes
.
2014
;
21
:
129
133
.

75.

Picaud
S
,
Wells
C
,
Felletar
I
, et al. .
RVX-208, an inhibitor of BET transcriptional regulators with selectivity for the second bromodomain
.
Proc Natl Acad Sci U S A
.
2013
;
110
:
19754
19759
.

76.

Nicholls
SJ
,
Gordon
A
,
Johannson
J
, et al. .
ApoA-I induction as a potential cardioprotective strategy: rationale for the SUSTAIN and ASSURE studies
.
Cardiovasc Drugs Ther
.
2012
;
26
:
181
187
.

77.

Nicholls
SJ
,
Puri
R
,
Wolski
K
, et al. .
Effect of the BET protein inhibitor, RVX-208, on progression of coronary atherosclerosis: results of the phase 2b, randomized, double-blind, multicenter, ASSURE Trial
.
Am J Cardiovasc Drugs
.
In press
.

78.

Tzagournis
M
,
Chiles
R
,
Ryan
JM
,
Skillman
TG
.
Interrelationships of hyperinsulinism and hypertriglyceridemia in young patients with coronary heart disease
.
Circulation
.
1968
;
38
:
1156
1163
.

79.

Levy
RI
,
Glueck
CJ
.
Hypertriglyceridemia, diabetes mellitus, and coronary vessel disease
.
Arch Intern Med
.
1969
;
123
:
220
228
.

80.

Moberg
B
,
Wallentin
L
.
High density lipoprotein and other lipoproteins in normolipidaemic and hypertriglyceridaemic (type IV) men with coronary artery disease
.
Eur J Clin Invest
.
1981
;
11
:
433
440
.

81.

Sarwar
N
,
Danesh
J
,
Eiriksdottir
G
, 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
458
.

82.

Rapp
JH
,
Harris
HW
,
Hamilton
RL
, et al. .
Particle size distribution of lipoproteins from human atherosclerotic plaque: a preliminary report
.
J Vasc Surg
.
1989
;
9
:
81
88
.

83.

Eckel
RH
,
Grundy
SM
,
Zimmet
PZ
.
The metabolic syndrome
.
Lancet
.
2005
;
365
:
1415
1428
.

84.

Alberti
KG
,
Eckel
RH
,
Grundy
SM
, et al. ;
International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; International Association for the Study of Obesity
.
Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity
.
Circulation
.
2009
;
120
:
1640
1645
.

85.

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
.

86.

Mora
S
,
Rifai
N
,
Buring
JE
,
Ridker
PM
.
Fasting compared with nonfasting lipids and apolipoproteins for predicting incident cardiovascular events
.
Circulation
.
2008
;
118
:
993
1001
.

87.

Russo
GT
,
Meigs
JB
,
Cupples
LA
, et al. .
Association of the Sst-I polymorphism at the APOC3 gene locus with variations in lipid levels, lipoprotein subclass profiles and coronary heart disease risk: the Framingham offspring study
.
Atherosclerosis
.
2001
;
158
:
173
181
.

88.

Staels
B
,
Dallongeville
J
,
Auwerx
J
,
Schoonjans
K
,
Leitersdorf
E
,
Fruchart
JC
.
Mechanism of action of fibrates on lipid and lipoprotein metabolism
.
Circulation
.
1998
;
98
:
2088
2093
.

89.

Frick
MH
,
Elo
O
,
Haapa
K
, 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
1245
.

90.

Tepel
M
,
Aspelin
P
,
Lameire
N
.
Contrast-induced nephropathy: a clinical and evidence-based approach
.
Circulation
.
2006
;
113
:
1799
1806
.

91.

Rubins
HB
,
Robins
SJ
,
Collins
D
, 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
418
.

92.

van Walraven
C
,
Forster
AJ
,
Parish
DC
, et al. .
Validation of a clinical decision aid to discontinue in-hospital cardiac arrest resuscitations
.
JAMA
.
2001
;
285
:
1602
1606
.

93.

Bezafibrate Infarction Prevention Study
.
Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease
.
Circulation
.
2000
;
102
:
21
27
.

94.

Keech
A
,
Simes
RJ
,
Barter
P
, 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
1861
.

95.

ACCORD Study Group
,
Ginsberg
HN
,
Elam
MB
,
Lovato
LC
, et al. 
.
Effects of combination lipid therapy in type 2 diabetes mellitus
.
N Engl J Med
.
2010
;
362
:
1563
1574
.

96.

Results of the low-dose (20 mg) pravastatin GISSI Prevenzione trial in 4271 patients with recent myocardial infarction: do stopped trials contribute to overall knowledge? GISSI Prevenzione Investigators (Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardico)
.
Ital Heart J
.
2000
;
1
:
810
820
.

97.

Yokoyama
M
,
Origasa
H
,
Matsuzaki
M
, et al. ;
Japan EPA lipid intervention study (JELIS) Investigators
.
Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): a randomised open-label, blinded endpoint analysis
.
Lancet
.
2007
;
369
:
1090
1098
.

98.

Saito
Y
,
Yokoyama
M
,
Origasa
H
, et al. ;
Japan JELIS Investigators
.
Effects of EPA on coronary artery disease in hypercholesterolemic patients with multiple risk factors: sub-analysis of primary prevention cases from the Japan EPA Lipid Intervention Study (JELIS)
.
Atherosclerosis
.
2008
;
200
:
135
140
.

99.

Kwak
SM
,
Myung
SK
,
Lee
YJ
,
Seo
HG
,
Korean Meta-analysis Study
G
.
Efficacy of omega-3 fatty acid supplements (eicosapentaenoic acid and docosahexaenoic acid) in the secondary prevention of cardiovascular disease: a meta-analysis of randomized, double-blind, placebo-controlled trials
.
Arch Intern Med
.
2012
;
172
:
686
694
.

100.

Stone
NJ
,
Robinson
JG
,
Lichtenstein
AH
, et al. ;
American College of Cardiology/American Heart Association Task Force on Practice Guideline Panel
.
2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines
.
Circulation
.
2014
;
129
:
S1
S45
.

101.

Ballantyne
CM
,
Braeckman
RA
,
Soni
PN
.
Icosapent ethyl for the treatment of hypertriglyceridemia
.
Expert Opin Pharmacother
.
2013
;
14
:
1409
1416
.

102.

Jorgensen
AB
,
Frikke-Schmidt
R
,
Nordestgaard
BG
,
Tybjaerg-Hansen
A
.
Loss-of-function mutations in APOC3 and risk of ischemic vascular disease
.
N Engl J Med
.
2014
;
371
:
32
41
.

103.

TG and HDL Working Group of the Exome Sequencing Project, National Heart, Lung, Blood Institute
,
Crosby
J
,
Peloso
GM
,
Auer
PL
, et al. 
.
Loss-of-function mutations in APOC3, triglycerides, and coronary disease
.
N Engl J Med
.
2014
;
371
:
22
31
.

104.

Brown
WV
,
Baginsky
ML
.
Inhibition of lipoprotein lipase by an apoprotein of human very low density lipoprotein
.
Biochem Biophys Res Commun
.
1972
;
46
:
375
382
.

105.

Aalto-Setala
K
,
Fisher
EA
,
Chen
X
, et al. .
Mechanism of hypertriglyceridemia in human apolipoprotein (apo) CIII transgenic mice. Diminished very low density lipoprotein fractional catabolic rate associated with increased apo CIII and reduced apo E on the particles
.
J Clin Invest
.
1992
;
90
:
1889
1900
.

106.

Gaudet
D
,
Brisson
D
,
Tremblay
K
, et al. .
Targeting APOC3 in the familial chylomicronemia syndrome
.
N Engl J Med
.
2014
;
371
:
2200
2206
.

107.

Dijk
W
,
Kersten
S
.
Regulation of lipoprotein lipase by Angptl4
.
Trends Endocrinol Metab
.
2014
;
25
:
146
155
.

108.

Lee
EC
,
Desai
U
,
Gololobov
G
, et al. .
Identification of a new functional domain in angiopoietin-like 3 (ANGPTL3) and angiopoietin-like 4 (ANGPTL4) involved in binding and inhibition of lipoprotein lipase (LPL)
.
J Biol Chem
.
2009
;
284
:
13735
13745
.

109.

Desai
U
,
Lee
EC
,
Chung
K
, et al. .
Lipid-lowering effects of anti-angiopoietin-like 4 antibody recapitulate the lipid phenotype found in angiopoietin-like 4 knockout mice
.
Proc Natl Acad Sci U S A
.
2007
;
104
:
11766
11771
.

110.

Sahebkar
A
,
Chew
GT
,
Watts
GF
.
Recent advances in pharmacotherapy for hypertriglyceridemia
.
Prog Lipid Res
.
2014
;
56
:
47
66
.

111.

Miller
M
,
Stone
NJ
,
Ballantyne
C
, et al. ;
American Heart Association Clinical Lipidology, Thrombosis, and Prevention Committee of the Council on Nutrition, Physical Activity, and Metabolism; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Nursing; Council on the Kidney in Cardiovascular Disease
.
Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association
.
Circulation
.
2011
;
123
:
2292
2333
.

112.

Sandholzer
C
,
Hallman
DM
,
Saha
N
, et al. .
Effects of the apolipoprotein(a) size polymorphism on the lipoprotein(a) concentration in 7 ethnic groups
.
Hum Genet
.
1991
;
86
:
607
614
.

113.

Kukushkin
SK
,
Shamarin
VM
,
Manoshkina
EM
,
Berzak
NV
,
Petrosian
K
.
[New drug forms of molsidomine, Corvaton-forte and Corvaton-retard, and their antianginal efficacy in stenocardia patients]
.
Ter Arkh
.
1996
;
68
:
29
30
.

114.

Koschinsky
M
,
Boffa
M
.
Lipoprotein(a) as a therapeutic target in cardiovascular disease
.
Expert Opin Ther Targets
.
2014
;
18
:
747
757
.

115.

Nordestgaard
BG
,
Chapman
MJ
,
Ray
K
, et al. ;
European Atherosclerosis Society Consensus Panel
.
Lipoprotein(a) as a cardiovascular risk factor: current status
.
Eur Heart J
.
2010
;
31
:
2844
2853
.

116.

Emerging Risk Factors Collaboration
,
Erqou
S
,
Kaptoge
S
,
Perry
PL
, et al. 
.
Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality
.
JAMA
.
2009
;
302
:
412
423
.

117.

Albers
JJ
,
Slee
A
,
O'Brien
KD
, et al. .
Relationship of apolipoproteins A-1 and B, and lipoprotein(a) to cardiovascular outcomes: the AIM-HIGH trial (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglyceride and Impact on Global Health Outcomes)
.
J Am Coll Cardiol
.
2013
;
62
:
1575
1579
.

118.

Hoover-Plow
J
,
Huang
M
.
Lipoprotein(a) metabolism: potential sites for therapeutic targets
.
Metabolism
.
2013
;
62
:
479
491
.

119.

Tsimikas
S
,
Witztum
JL
.
The role of oxidized phospholipids in mediating lipoprotein(a) atherogenicity
.
Curr Opin Lipidol
.
2008
;
19
:
369
377
.

120.

Kolski
B
,
Tsimikas
S
.
Emerging therapeutic agents to lower lipoprotein (a) levels
.
Curr Opin Lipidol
.
2012
;
23
:
560
568
.

121.

Suk Danik
J
,
Rifai
N
,
Buring
JE
,
Ridker
PM
.
Lipoprotein(a), hormone replacement therapy, and risk of future cardiovascular events
.
J Am Coll Cardiol
.
2008
;
52
:
124
131
.

122.

Tsimikas
S
,
Viney
NJ
,
Hughes
SG
, et al. .
Antisense therapy targeting apolipoprotein(a): a randomised, double-blind, placebo-controlled phase 1 study
.
Lancet
.
2015
;
386
:
1472
1483
.

123.

Gaudet
D
,
Methot
J
,
Kastelein
J
.
Gene therapy for lipoprotein lipase deficiency
.
Curr Opin Lipidol
.
2012
;
23
:
310
320
.

124.

Grossman
M
,
Rader
DJ
,
Muller
DW
, et al. .
A pilot study of ex vivo gene therapy for homozygous familial hypercholesterolaemia
.
Nat Med
.
1995
;
1
:
1148
1154
.