Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Mar 29.
Published in final edited form as: Nat Rev Cardiol. 2011 Jan 18;8(5):266–277. doi: 10.1038/nrcardio.2010.200

Novel HDL-directed pharmacotherapeutic strategies

Emil M deGoma 1, Daniel J Rader 2
PMCID: PMC3315102  NIHMSID: NIHMS334680  PMID: 21243009

Abstract

The burden of atherothrombotic cardiovascular disease remains high despite currently available optimum medical therapy. To address this substantial residual risk, the development of novel therapies that attempt to harness the atheroprotective functions of HDL is a major goal. These functions include the critical role of HDL in reverse cholesterol transport, and its anti-inflammatory, antithrombotic, and antioxidant activities. Discoveries in the past decade have shed light on the complex metabolic and antiatherosclerotic pathways of HDL. These insights have fueled the development of HDL-targeted drugs, which can be classified among four different therapeutic approaches: directly augmenting apolipoprotein A-I (apo A-I) levels, such as with apo A-I infusions and upregulators of endogenous apo A-I production; indirectly augmenting apo A-I and HDL-cholesterol levels, such as through inhibition of cholesteryl ester transfer protein or endothelial lipase, or through activation of the high-affinity niacin receptor GPR109A; mimicking the functionality of apo A-I with apo A-I mimetic peptides; and enhancing steps in the reverse cholesterol transport pathway, such as via activation of the liver X receptor or of lecithin–cholesterol acyltransferase.

Introduction

Individuals with manifest atherosclerotic disease, or a collection of associated risk factors, continue to endure a high incidence of adverse cardiovascular events despite the use of aggressive combination medical therapy. In the best-case scenario of intensive treatment and vigilant monitoring, the incidence of ‘hard’ cardiovascular events in secondary prevention cohorts is 20% after 4–5 years.1,2 To address the substantial residual risk of atherothrombotic events that exists even after optimum medical therapy, the development of novel therapies that attempt to make use of the atheroprotective functions of HDL, from its critical role in reverse cholesterol transport to its anti-inflammatory, antithrombotic, and antioxidant activities (Figure 1),3 is a major goal. Discoveries in the past decade, including the identification of the high-affinity niacin receptor GPR109A, the development of assays that enable the assessment of HDL functionality (such as those that assess the efficiency of macrophage-specific reverse cholesterol transport), and the results of massive genome-wide association studies, have shed light on new and complex metabolic and antiatherosclerotic pathways in which HDL is involved.4,5 These insights, in turn, have fueled the development of new HDL-targeted drugs, which can be classified according to four different therapeutic approaches: directly augmenting the concentration of apolipoprotein A-I (apo A-I), the major protein constituent of HDL; indirectly augmenting the concentration of apo A-I and HDL cholesterol; mimicking the functionality of apo A-I; and enhancing reverse cholesterol transport (Box 1). In this Review, we aim to discuss the hypothesized mechanisms of these pharmacotherapies, changes in HDL mass and function and, where available, surrogate atherosclerosis end points from pre-clinical and clinical studies.

Figure 1.

Figure 1

HDL metabolism and targets of therapeutic intervention. Synthesized by the liver and the intestine, apo A-I acquires phospholipid to form nascent preβ-HDL. ABCA1 initiates the first step of reverse cholesterol transport, facilitating the efflux of free cholesterol from peripheral cells to nascent preβ-HDL. LCAT esterifies the cholesterol molecules to form cholesteryl esters, which migrate to the core of the HDL particle, resulting in formation of α-HDL. These mature HDL particles can acquire additional lipid via efflux mediated by ABCG1 and SR-BI. CETP mediates exchange of cholesteryl esters for triglycerides with VLDL or LDL, effecting depletion in cholesteryl esters and enrichment in triglycerides of HDL. The resulting HDL3 particles can be either taken up by the liver via SR-BI holoparticle uptake or modified by hepatic lipase and endothelial lipase. Metabolism by the latter releases lipid-poor apo A-I, which can be filtered by the glomeruli and degraded by cubilin/megalin in the proximal renal tubule. Targets of HDL-directed therapeutic interventions are indicated by red arrows and lines. Abbreviations: ABC, ATP-binding cassette transporter; Apo A-I, apolipoprotein A-I; CETP, cholesteryl ester transfer protein; CD36 and LIMPII analogous-1; LCAT, lecithin–cholesterol acyltransferase; LDL-R, LDL receptor; SR-BI, scavenger receptor class B type I.

Box 1. HDL-directed pharmacotherapeutic strategies.

Directly augmenting apo A-I

Intravenous apo A-I therapy

  • Recombinant apo A-I Milano/phospholipids (ETC-216)

  • Purified native apo A-I/phospholipids (CSL-111/112)

  • Autologous delipidated HDL

Oral upregulators of endogenous apo A-I production

  • RVX-208

Indirectly augmenting apo A-I and HDL-cholesterol

Cholesteryl ester transfer protein inhibitors

  • Dalcetrapib

  • Anacetrapib

Niacin receptor (GPR109A) agonists

Endothelial lipase inhibitors

Mimicking apo A-I functionality

Apo A-I mimetic peptides

Enhancing reverse cholesterol transport

Liver X receptor agonists

Lecithin–cholesterol acyltransferase activators

Abbreviation: ApoA-I, apolipoproteinA-I.

Directly augmenting apo A-I levels

Lipid-poor apo A-I, also termed nascent HDL or preβ-HDL, initiates reverse cholesterol transport by activating macrophage ATP-binding cassette sub-family A member 1 (ABCA1) and accepting effluxed cholesterol. From a pharmacodynamic standpoint, direct augmentation of lipid-poor apo A-I concentration arguably represents the most validated HDL-related therapeutic approach in terms of antiatherogenic potential. Lipid-poor apo A-I–phospholipid complexes, sometimes referred to as recombinant HDL (rHDL), have been studied extensively in animals and in preliminary studies in humans. Preclinical studies have demonstrated that the administration of apo A-I is associated with the inhibition or regression of atherosclerosis,610 enhanced macrophage-specific reverse cholesterol transport,5 and the inhibition of vascular inflammatory pathways,11 endothelial adhesion molecule expression12 and phospholipid oxidation.13 Moreover, short exploratory clinical studies of rHDL infusion have yielded decreases in coronary atherosclerosis, as assessed by coronary imaging, comparable with those obtained with long-term statin use at doses associated with improved clinical outcomes. These findings support the therapeutic potential of intravenous apo A-I infusion (Table 1).1417

Table 1.

Intravenous apo A-I for coronary atherosclerosis in humans1417

IVUS parameter ETC-216 CSL-111 Autologous HDL Atorvastatin 80 mg
Duration of therapy 5 weeks 4 weeks 7 weeks 18 months
Baseline atheroma volume (mm3) 268.4 151.0 229.3 184.4
Change in total atheroma volume (mm3; %) −14.1; −1.1 −5.3; −3.4 −12.2; −1.0 −0.4; −0.2

Abbreviation: IVUS, intravascular ultrasonography.

Recombinant apo A-I Milano/phospholipid

The recombinant apo A-I Milano protein differs from wild-type apo A-I by a cysteine to arginine substitution at amino acid 173. The effects of apo A-I Milano complexed with phospholipid (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine), also known as ETC-216, have been studied in animals and humans. The apo A-I Milano mutation was first identified in a cohort of Italian patients who exhibited a decreased prevalence of atherosclerosis despite very low levels of HDL cholesterol (10–20 mg/dl).18 In support of an antiatherogenic activity for this drug, infusions of ETC-216 in rabbit and mouse models of atherosclerosis were associated with considerable reductions in the lipid and macrophage content of plaque.10,19,20 The superiority of apo A-I Milano over native apo A-I, however, remains uncertain. Using genetic mouse models of atherosclerosis, cholesterol efflux assays in vitro, and examination of macrophage-specific reverse cholesterol transport in vivo, some studies suggested that apo A-I Milano had greater antiatherogenic effects than wild-type apo A-I,21,22 whereas others indicated equivalence to the wild-type protein.2326 Head-to-head intervention studies comparing administration of the two types of apo A-I are lacking.

In a small clinical study of 47 patients with acute coronary syndrome, five weekly doses of 15–45 mg/kg of ETC-216 significantly reduced total atheroma volume by 14.1 mm3 compared with baseline (4.2%; P <0.001), as measured by coronary intravascular ultrasonography (IVUS) (Table 1).14 Total atheroma volume did not change significantly with the administration of placebo (–0.2 mm3; P = 0.97), and HDL-cholesterol and apo A-I levels were not reported. Two of 15 patients (9%) in the high-dose ETC-216 group were withdrawn from the study owing to adverse events, one because of symptomatic cholelithiasis and the other because of a hypersensitivity reaction.14 A subsequent dose-ranging study in a rabbit model suggested that five infusions of ETC-216 at doses equivalent to 8–15 mg/kg in humans might be adequate to maximize plaque stabilization or regression.8 Substantial interest remains in investigating the therapeutic potential of rHDL infusion containing apo A-I Milano. Clinical development of this drug, which was halted owing to manufacturing difficulties, is scheduled to resume in 2011 following completion of technology transfer to a different pharmaceutical company.27

Purified native apo A-I/phospholipid

CSL-111 consists of complexes of native apo A-I and phosphatidylcholine, the former isolated from precipitates obtained by cold ethanol fractionation of human plasma and the latter derived from soybean.16,28 A randomized, placebo-controlled study in which serial intravascular ultrasonography was used to assess 145 patients with acute coronary syndrome revealed that administration of four weekly 40 mg/kg infusions of CSL-111 reduced atheroma volume by 5.3 mm3 compared with baseline (3.4%; P <0.001) (Table 1).16 No statistically significant difference in atheroma volume was observed when comparing patients receiving placebo and those treated with CSL-111 (P = 0.48). Quantitative coronary angiography, on the other hand, demonstrated that CSL-111 use was associated with less progression of coronary atherosclerosis than placebo (–0.039 mm and –0.071 mm, respectively; P = 0.03), as measured by the coronary score (per-patient mean of the minimal lumen diameter for all lesions measured) among patients with less-severe stenoses at baseline.16 Lower coronary scores have been associated with an increased risk of incidence of coronary events.29 By comparison, 40 mg daily doses of pravastatin or 40–80 mg daily doses of lovastatin administered for 2 years attenuated coronary atherosclerosis progression as assessed by coronary score to a similar degree as only 4 weeks of CSL-111 treatment.30,31 One-third of patients randomly assigned to receive 80 mg of CSL-111 had reversible alanine aminotransferase elevations exceeding 10 times the normal upper limit, but the group administered CSL-111 at the lower dose of 40 mg/kg did not differ from the placebo group with regard to transaminase levels. More participants randomly assigned to 40 mg/kg of CSL-111 exhibited hypotension compared with those receiving placebo (13.8% and 7.1%, respectively). Further details regarding the observed changes in blood pressure were not provided in the original publication.16

A subsequent study in patients with lower extremity peripheral artery disease demonstrated that treatment with a single CSL-111 infusion was associated with significant reductions in lipid content (P <0.05; assessed by oil red O staining) and endothelial adhesion molecule expression (P <0.05; assessed by vascular cell adhesion molecule 1 [VCAM-1] expression) in plaque excised by atherectomy.32 These changes were observed at the time of percutaneous superficial femoral artery revascularization, which occurred 5–7 days following CSL-111 infusion, indicating a rapid onset of action of the drug. In addition, CSL-111 considerably increased the levels of HDL cholesterol and enhanced the capacity of plasma depleted in apolipoprotein B (apo B) to promote cholesterol efflux from macrophages. Among patients with diabetes mellitus, infusion of CSL-111 increased the levels of HDL cholesterol up to 40%, inhibited platelet aggregation ex vivo, and reduced monocyte activation and neutrophil adhesion.33,34 A reformulated version of CSL-111, called CSL-112, has been reported in preclinical studies to provide greater cholesterol efflux capacity and have less hepatotoxicity than CSL-111.35 A phase I study of intravenous CSL-112 has been initiated.35

Autologous delipidated HDL

A novel HDL-directed pharmacotherapeutic approach utilizes the infusion of autologous delipidated HDL.17 The Plasma Delipidation System-2 (PDS-2) from Lipid Sciences involves the collection of 1 l of plasma by apheresis over 1.5–2.0 h followed by the selective removal of lipids from HDL particles using organic solvents; the lipid-poor preβ-HDL, an efficient acceptor of cholesterol from ABCA1, is subsequently reinfused over 1 h.

Preclinical evaluation of selective delipidated HDL in a nonhuman primate model of dyslipidemia achieved a significant 6.9% reduction in aortic atheroma volume, assessed by IVUS.36 In a small clinical study, among 28 patients with acute coronary syndrome, seven weekly infusions of autologous delipidated HDL increased preβ-HDL levels approximately 30-fold and decreased total atheroma volume by 12 mm3 (5.2%) from baseline (Table 1).17 As in the studies of intravenous apo A-I therapy discussed above, no statistically significant differences were observed between actively-treated patients and control groups. Autologous delipidated HDL infusions did not induce transaminitis or hypersensitivity reactions; however, apheresis resulted in hypotension in a third of the participants undergoing this treatment.

Oral upregulator of endogenous apo A-I

A small synthetic molecule (termed RVX-208) belonging to the quinazoline family, which are known primarily for their antimalarial properties, was reported in cellular screening assays to raise apo A-I production.37 In a monkey model of atherosclerosis, administration of a compound containing RVX-208 over approximately 2 months increased plasma apo A-I and HDL-cholesterol levels up to 60% and 97%, respectively, in a dose-dependent manner. Monkeys treated with RVX-208 had enhanced cholesterol efflux from macrophage foam cells ex vivo. A doubling in the levels of triglycerides was observed with the highest dose of the drug; however, the mid-range dose led to a 43% increase in apo A-I concentration without affecting triglyceride levels.37 No effect on apo B or LDL-cholesterol levels was observed with any dose of RVX-208. A short exploratory study in humans demonstrated increases in total plasma apo A-I and preβ-HDL levels (10% and 42%, respectively), as well as augmentation of cholesterol efflux with the use of RVX-208.37 Findings from the ASSERT study were presented at the 2010 AHA conference, and the full results are anticipated to be released in 2011. Modest changes in HDL-C and apo A-I were reported to have been observed in patients with stable coronary artery disease on chronic statin therapy.38 According to Dr Stephen J. Nicholls, a study of RVX-208 is being planned in patients with acute coronary syndrome using coronary intravascular ultrasonography and is currently scheduled to begin in 2011.38

Indirectly augmenting apo A-I levels

Increasing apo A-I and HDL-cholesterol levels can also be achieved indirectly by manipulating HDL metabolism, particularly through approaches that slow the rate of catabolism of apo A-I or HDL cholesterol, or both. Approaches within this category include inhibition of cholesteryl ester transfer protein (CETP) or endothelial lipase and, potentially, stimulation of the niacin receptor GPR109A.

CETP inhibitors

CETP shuttles cholesteryl esters from HDL to LDL and VLDL in exchange for triglycerides.39 Genetic CETP deficiency causes marked elevation in HDL-cholesterol levels and modest elevation in apo A-I levels. Some,4044 but not all,4547 epidemiological studies indicate that CETP has an atherogenic role, with reduced plasma CETP mass or activity associated with reduced incidence of atherothrombotic cardiovascular disease; however, this issue remains far from settled. Preclinical studies involving reduction in CETP activity through various means, including the use of small-molecule inhibitors, antisense oligonucleotides, and vaccine-induced antibodies, have generally demonstrated decreased plaque formation with these agents, lending further support to CETP inhibition as a viable therapeutic strategy for the prevention and management of atherothrombotic disease.4852

Despite these supporting data, the demise of the CETP inhibitor torcetrapib raised doubts about the concept of CETP inhibition. In the Investigation of Lipid Level Management to Understand its Impact in Atherosclerotic Events (ILLUMINATE),53 which involved patients receiving atorvastatin, torcetrapib worsened the primary combined cardiovascular end point (death from coronary heart disease, nonfatal myocardial infarction, stroke, and hospitalization for unstable angina) (hazard ratio [HR] 1.25, 95% CI 1.09–1.44, P = 0.001) and all-cause mortality (HR 1.58, 95% CI 1.14–2.19, P = 0.006) compared with placebo after 12 months despite increasing HDL-cholesterol levels by 72% and decreasing LDL-cholesterol levels by 25%.53 The worrisome results of ILLUMINATE, paralleled by negative imaging findings in trials in which coronary and carotid ultrasonography were used,54,55 have been at least partly attributed to the off-target effects of torcetrapib, such as the raising of systolic blood pressure by an average of 5.4 mmHg.56,57 As suggested by aldosterone measurements from participants in ILLUMINATE,53 findings from animal models58 and human adrenal cell assays59 indicate that these off-target effects are related to stimulation of aldosterone synthesis by torcetrapib via pathways independent of CETP inhibition.

Could inhibition of CETP activity explain some of the adverse effects observed in the clinical development program of torcetrapib? Initial concerns about CETP inhibition highlighted the possibility that the formation of large cholesterol-rich HDL particles resulting from CETP inhibition might be associated with impaired cholesterol efflux from peripheral macrophages to these particles.60 However, studies of patients either deficient in CETP or treated with a CETP inhibitor demonstrated not reduced, but rather enhanced, efflux of cholesterol via ATP-binding cassette sub-family G member 1 (ABCG1) to HDL.59,61,62 An important arm in the proximal step of reverse cholesterol transport, therefore, remains intact, if not improved, in the setting of CETP inhibition. On the other hand, the net effect of CETP inhibition on reverse cholesterol transport might depend, in part, on the effects of CETP on the hepatic uptake of HDL-derived cholesterol. The hepatic uptake of HDL cholesterol can be direct, or occur after transfer of HDL cholesterol via CETP to apo B-containing lipoproteins, which are then taken into the liver via the LDL receptor. As illustrated by macrophage-specific reverse cholesterol transport assays, in the setting of highly effective LDL clearance, CETP can actually enhance reverse cholesterol transport and have an atheroprotective role; however, when LDL clearance is impaired, CETP can slow down reverse cholesterol transport and thus be proatherogenic.63 The interdependence of terminal reverse cholesterol transport pathways suggests that the hypothesis that the individuals who are most likely to benefit from CETP inhibition are those with suboptimal LDL-receptor-mediated hepatic uptake of cholesterol. Fortunately, at least two novel compounds apparently lacking the off-target effects of torcetrapib enable further testing of the CETP-inhibition strategy.

Dalcetrapib

Dalcetrapib binds CETP irreversibly and is considerably less-potent than torcetrapib.64 A study in a hamster model suggested that dalcetrapib promotes reverse cholesterol transport.65 In a human study, among individuals with mean baseline HDL-cholesterol and LDL-cholesterol levels of 47 mg/dl and 144 mg/dl, respectively, mono-therapy with 600 mg of dalcetrapib daily increased HDL-cholesterol levels by 23% compared with placebo administration, after 4 weeks of treatment.66 Among patients with type II dyslipidemia receiving 40 mg of pravastatin daily whose baseline levels of HDL cholesterol and LDL cholesterol were 48 mg/dl and 120 mg/dl, respectively, the addition of 600 mg of dalcetrapib daily increased HDL-cholesterol levels by 28% and decreased LDL-cholesterol levels by 7% compared with placebo administration, after 4 weeks of treatment.67 After 24 weeks of therapy with 900 mg of dalcetrapib daily, HDL-cholesterol levels increased by 33% (mean baseline level of 42 mg/dl) in patients at high risk of coronary heart disease events who were treated with 10–80 mg of atorvastatin daily, when compared with HDL levels in participants receiving placebo plus atorvastatin.68 LDL-cholesterol levels, however, did not differ in the two patient groups (mean baseline level of 74 mg/dl).68 Importantly, no changes in blood pressure or aldosterone levels were observed with the use of high-dose dalcetrapib (900 mg).68

Two ongoing vascular imaging studies, DAL-Plaque69 and DAL-Plaque 2,70 will assess changes in atherosclerotic plaque (morphology, composition, and inflammatory activity) among patients with coronary heart disease treated with 600 mg of dalcetrapib daily. A wide variety of modalities will be used, including coronary and carotid ultrasonography, PET, and MRI. Another study, DAL-Vessel,71 will examine a different functional surrogate, arterial reactivity, by assessing brachial flow-mediated dilatation. Finally, the phase III clinical trial DAL-Outcomes72 will evaluate the effects of adding 600 mg of dalcetrapib daily to optimum pharmacotherapy in patients with acute coronary syndrome over a follow-up period of at least 2 years, with results expected in 2013.64

Anacetrapib

Anacetrapib potently inhibits CETP by forming a tight reversible bond with this protein.73 Healthy individuals with mean baseline HDL-cholesterol and LDL-cholesterol levels of 51 mg/dl and 138 mg/dl, respectively, exhibited changes in the levels of these lipoproteins of +129% and –38%, respectively, after receiving 300 mg of anacetrapib daily for 10 days.74 No changes in blood pressure, assessed through 24 h ambulatory monitoring, were observed. Among patients with dyslipidemia who had mean baseline HDL-cholesterol and LDL-cholesterol levels of 50 mg/dl and 141 mg/dl, respectively, addition of 300 mg of anacetrapib to 20 mg of atorvastatin daily for 8 weeks increased HDL-cholesterol levels by 120% and decreased LDL-cholesterol levels by 30% compared with statin monotherapy.75 Interestingly, lipoprotein(a) levels, which were unchanged by statin therapy, decreased by up to 50% following anacetrapib administration.75

Beyond a beneficial effect on standard lipid-mass parameters, anacetrapib can improve measures of HDL functionality. Compared with HDL isolated from patients receiving niacin or placebo, HDL sampled from patients who received 300 mg of anacetrapib daily for 8 weeks promoted greater cholesterol efflux from foam cells in culture, independent of HDL-cholesterol levels.62 In the same experimental set-up, lipid-A-induced cytokine and chemokine messenger RNA (mRNA) transcription (markers of inflammation) were comparable when the cells were treated with HDL from patients who received anacetrapib and when they were treated with control HDL, suggesting that the anti-inflammatory activity of HDL remained preserved with anacetrapib administration.

The phase III Determining the Efficacy and Tolerability of CETP Inhibition with Anacetrapib (DEFINE) randomized, placebo-controlled trial examined the effect of 100 mg of anacetrapib administered daily for 18 months to 1623 patients with coronary heart disease or equivalent conditions (peripheral artery disease, cerebrovascular disease, diabetes, or a 10-year Framingham risk score >20%) who had achieved LDL-cholesterol treatment goals with statin therapy.76 The primary end points were the percent change in LDL-cholesterol at 24 weeks and the safety profile of anacetrapib at 76 weeks. HDL-cholesterol and other lipid parameters including lipoprotein(a) were assessed as secondary endpoints.77 Treatment with anacetrapib was associated with a 40% reduction in LDL-cholesterol from 81 mg/dl to 45 mg/dl (P <0.001) and a 138% increase in HDL-cholesterol from 41 mg/dl to 101 mg/dl (P <0.001) compared with placebo.77 Lipoprotein(a) decreased 36% compared with placebo from 27 nmol/l to 15 nmol/l. No increases in clinic-based blood pressure, serum aldosterone levels, or cardiovascular events were observed following anacetrapib treatment at 76 weeks. Supported by these substantial improvements in LDL, HDL, and lipoprotein(a), as well as an apparently benign safety profile, the Randomized EValuation of the Effects of Anacetrapib Through Lipid-modification (REVEAL) is scheduled to begin in April 2011.78 This study will examine major coronary events, defined as coronary death, myocardial infarction and coronary revascularization procedures, in 30,000 patients with coronary heart disease, cerebrovascular atherosclerotic disease, or peripheral artery disease. The estimated study completion date is January 2017.78

Niacin receptor agonists

Niacin, the first antidyslipidemic agent identified, remains the most potent drug for increasing levels of HDL-cholesterol and apo A-I. Evidence suggests that HDL mediates the potential atheroprotective effects of niacin,79 although the role of beneficial changes in other lipid parameters, including a reduction in the concentration of LDL particles and lipoprotein(a), cannot be excluded. Randomized trials have shown a clinical benefit of niacin among patients with coronary heart disease, with low cardiovascular event rates80,81 and improvements in surrogate measures of vascular function, as assessed by imaging,79,82 associated with niacin administration. One of these studies, for example, revealed regression of carotid intima–media thickness (IMT), the primary imaging end point, associated with niacin treatment, whereas no change in IMT was observed among patients treated with the LDL-lowering drug ezetimibe.82 The niacin group achieved better outcomes in IMT compared with the ezetimibe group in the setting of 23% higher HDL-cholesterol levels, despite 7% higher LDL-cholesterol levels, lending further credence to an HDL-targeted approach.82 Definitive data on the incremental value of niacin when added to optimum medical therapy, which includes aggressive statin use, might emerge from two large trials, the Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides and Impact on Global Health Outcomes (AIM-HIGH) study83 and the Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2-THRIVE) study,84 which are anticipated to be communicated in 2012 or 2013.

After 50 years of clinical use,85 the mechanisms by which niacin raises the levels of HDL (and other lipoproteins) remain elusive. Studies of cultured hepatocytes suggest that these effects might be mediated by decreased catabolism of apo A-I owing to downregulation of the HDL holoparticle (protein plus lipids) receptor, which is a terminal step in the reverse cholesterol transport pathway.86 Kinetic studies in humans, on the other hand, demonstrate either decreased clearance87,88 or increased synthesis of apo A-I in patients receiving niacin,89 the latter finding being notably absent from analyses in vitro. The discovery of the niacin receptor GPR109A90 promised to usher in a new era in which the molecular mechanisms underlying the effects of niacin on lipids, and its adverse effects on the skin, such as flushing, could be clearly defined. This discovery might also open the way for the development of new synthetic agonists of GPR109A, which would ideally have the same therapeutic effects as niacin, but not the skin-related adverse effects of this drug, which substantially limit its therapeutic use. Activation of GPR109A on skin immune cells, such as Langerhans cells and keratinocytes, stimulates phospholipase A2, enhancing synthesis of prostaglandin D2 (PGD2) and prostaglandin E2 (PGE2), which subsequently induce cutaneous capillary vasodilation by binding to subtypes of the PGD2 receptor (DP1) and the PGE2 receptor (EP2 and EP4).4 Coadministration of the DP1 antagonist laropiprant reduces the incidence of the skin-related adverse effects of niacin.91 However, flushing still occurs in over half of the patients, and discontinuation of niacin treatment owing to severe skin-related symptoms remains a problem.91 A study in mice implicates keratinocyte-produced PGE2 as a key mediator of niacin-induced skin flushing, suggesting that this molecule might be another potential target to minimize flushing.92,93

Although activation of GPR109A on adipocytes clearly leads to an acute reduction in lipolysis and release of free fatty acids,90 the role of this receptor in mediating the effects of niacin on plasma lipoproteins remains uncertain. Partial agonism of GPR109A might differentially activate lipolysis and flushing pathways. A study in mice supports this dissociation and implicates ERK1/2 mitogen-activated protein kinase phosphorylation in vasodilation.94 Administration of the partial agonist MK-0354 in humans resulted in a lack of flushing; however, despite effective suppression of lipolysis and free-fatty-acid release, this partial agonist had no effect on HDL-cholesterol or other plasma lipoproteins.95 These results have brought into question the role of GPR109A agonism in mediating the effects of niacin therapy on plasma lipoproteins. This question is critical for the field, and will determine the future of GPR109A agonists as a therapeutic strategy.

Endothelial lipase inhibitors

Synthesized by and bound to vascular endothelial cells, endothelial lipase exhibits predominant phospholipase activity and affinity for HDL, unique characteristics among the lipoprotein–lipase family.96 An association between the expression of endothelial lipase and HDL-cholesterol levels was identified in overexpression and loss-of-function mouse models.9799 Genetic studies indicate a similar association in humans, with rare and low-frequency loss-of-function variants of endothelial lipase, identified through deep sequencing, resulting in elevated HDL-cholesterol levels.100 How changes in HDL-cholesterol levels attributed to endothelial lipase ultimately affect atherosclerosis remains uncertain. Some human studies point to an atherogenic role of endothelial lipase, with a positive association between plasma levels of this enzyme and coronary artery calcification, features of the metabolic syndrome (such as impaired fasting glucose, hypertriglyceridemia, low HDL-cholesterol, hypertension, and waist circumference), and inflammation.101,102 Carriers of endothelial-lipase variants associated with raised HDL-cholesterol levels have been reported to have a decreased risk of atherothrombotic disease,103 although this link has not been observed in other studies.104,105 Of note, animal studies have shown that HDL-cholesterol levels do not always correlate with physiological changes associated with decreased atherosclerotic burden, with an endothelial-lipase knockout mouse model failing to demonstrate improved macrophage reverse cholesterol transport with raised HDL-cholesterol levels.106 More worrisome, in the setting of hepatic-lipase deficiency, is that the absence of endothelial lipase resulted in an accumulation of small dense LDL,106 a particularly atherogenic subpopulation of LDL. These findings suggest that endothelial-lipase inhibition could potentially exert a detrimental effect on atherosclerosis, even though HDL-cholesterol levels are raised. Despite this uncertainty, endothelial lipase has been the object of substantial interest as a therapeutic target. High-throughput screening identified several compounds sharing a sulfonyl furan urea core as potent and selective endothelial-lipase inhibitors.107 Further evaluation of these and other small-molecule inhibitors of endothelial lipase has not been performed yet.

Mimicking apo A-I functionality

Another HDL-directed therapeutic approach utilizes small peptides that mimic one or more of the functions of apo A-I.108,109 The most well-studied apo A-I mimetic, 4F, consists of 18 amino acids designed to share the lipid-binding properties of apo A-I through a common secondary structure, the class A amphipathic helix.110 Although many studies have employed the parenteral administration of 4F made from L-amino acids (L-4F), the use of D-amino acids (D-4F) enables oral delivery of this compound by conferring resistance to mammalian gastrointestinal proteolytic enzymes.111 Studies in vitro112116 and in animals111,117119 indicate that 4F recapitulates many of the functions of apo A-I. In respect to reverse cholesterol transport, 4F has been demonstrated to promote cholesterol efflux from macrophages via ABCA1109,116 and offloading of cholesterol to hepatocytes via the scavenger receptor class B type I (SR-BI).114 In addition, 4F exhibits apo A-I-like anti-inflammatory properties, inducing a quiescent macrophage phenotype113 and inhibiting LDL-induced macrophage chemotaxis.111 The antioxidant capacity of 4F can surpass that of the native apo A-I, with 4F exhibiting a greater affinity for oxidized phospholipids and fatty acids,115 and retaining the ability to inhibit the production of chemokines from human artery walls despite coincubation with LDL.112 Finally, like apo A-I, 4F has antithrombotic properties, inhibiting platelet aggregation in hyperlipidemic mouse models.117

Animal studies suggest that these functions of 4F translate into atheroprotective effects.111,118,119 Several mouse models of accelerated atherosclerosis, including LDL-receptor-knockout and apolipoprotein E (apo E)-knockout mice, developed less atheroma when treated with 4F than controls.111,118 One short-term study in LDL receptor-null mice demonstrated no effect of the apo A-I mimetic on established aortic atheroma size and lipid content and a minimal decrease in macrophage content.111 Administration of 4F for 6 months in old apo E-null mice with coadministration of statins was associated with regression of established aortic lesions, which is, arguably, a more clinically relevant parameter than the effects on de novo plaque development.119

The only reported human study of D-4F to date hints at the possibility of a benefit for this compound in humans.120 Compared with HDL isolated from individuals who received placebo, HDL isolated from individuals treated with a single 300 mg or 500 mg dose of unformulated D-4F increased the inhibition of LDL-induced monocyte chemotaxis in cultures of human aortic endothelial cells. As in animal studies, neither changes in apo A-I or HDL-cholesterol levels nor serious toxicity were observed in treated individuals.

Several other apo A-I mimetics have been developed. Some, such as ETC-642 (also called RLT peptide), were designed to activate lecithin–cholesterol acyltransferase (LCAT; see below);121 others, such as oxpholipin 11, have antioxidant properties;122 and still others, such as ATI-5261, were engineered to maximize cholesterol efflux.123 The latter peptide increased macrophage-specific reverse cholesterol transport and also reduced the development of aortic atherosclerosis by up to 45% over 6 weeks of intravenous treatment in mice.123 As illustrated in a systematic study of 22 apo A-I mimetic peptides, structural elements that enhance one aspect of HDL functionality do not necessarily optimize other atheroprotective qualities that characterize apo A-I.124 The clinical development of apo A-I mimetic peptides is in its infancy, but with the current amount of activity in this area of research we can expect to see a growing number of additional peptides being developed. With the exception of D-4F, apo A-I mimetic peptides require parenteral administration and, therefore, are likely to be initially targeted to patients at high risk with acute coronary syndromes.

Enhancing reverse cholesterol transport

As reverse cholesterol transport is the most well-established atheroprotective function of HDL, particularly in its initial steps involving cholesterol efflux from peripheral cells, this process represents a major target of novel HDL-directed therapies.

Agonists of the liver X receptor

Liver X receptors (LXRs), which are members of the nuclear receptor superfamily, have a central role in lipid metabolism. Endogeneously activated by oxysterols, LXRs regulate the transcription of a myriad of target genes by binding to their promoters together with the retinoic acid receptor.125 With regard to HDL and reverse cholesterol transport, LXR activation has been demonstrated to promote mobilization of intracellular cholesterol,126 increase macrophage cholesterol efflux via macrophage ABCA1 and ABCG1,127 and augment intestinal HDL generation.128 Two LXR isoforms have been identified—LXRα and LXRβ. These isotypes have structural homology, but differ in tissue distribution, with the former being primarily expressed in the liver, macrophages, intestine, kidney, and adipose tissue, whereas the latter is found ubiquitously.129

Therapeutic development of LXR agonists has been hindered by hepatic steatosis and increased plasma tri-glyceride concentrations reported in preclinical studies of these drugs.130,131 Fortunately, dissociating LXR efficacy and toxicity might be possible owing to the differential effects of LXR agonism by receptor isoform and by tissue-specific effects. For example, gene knockout models in mice implicate LXRα,132 and not LXRβ,133 as the primary stimulus for hepatic lipogenesis and hypertriglyceridemia. Administration of a nonselective LXR agonist to LXRα-deficient mice increased HDL-cholesterol levels and stimulated macrophage ABCA1 expression and cholesterol efflux without inducing fatty liver and with minimal transcription of hepatic sterol regulatory element-binding protein isoform 1c (SREBP1c) mRNA, which upregulates the synthesis of triglycerides in the liver.74,134,135 Moreover, in a mouse model susceptible to atherosclerosis that lacked LXRα and apo E, treatment with the LXRα/β agonist GW3965 not only improved cholesterol in vitro (decreased total and unesterified cholesterol levels and increased HDL-cholesterol levels) but reduced aortic plaque development.136 These results indicate that a selective LXRβ agonist might augment reverse cholesterol transport without engendering hypertriglyceridemia or fatty liver.

A second approach to safer LXR development might be to selectively activate intestinal LXR. Fatty liver arises from activation of hepatic LXR, which, through upregulation of SREBP1c, stimulates lipogenesis.137,138 Elevation of triglyceride levels occurs via SREBP1c activation139 and the subsequent suppression of apolipoprotein A-V (apo A-V),140 which is an inhibitor of VLDL synthesis (apo A-V can also stimulate VLDL hydrolysis).141 On the other hand, LXR expression on both macrophages and the small intestine contributes to the regulation of reverse cholesterol transport. The absence of macrophage LXRs in a mouse model attenuated, but did not abolish, the increase in macrophage-specific reverse cholesterol transport observed following the administration of the systemic LXRα/β agonist GW3965, which indicates that macrophage LXR expression, but also nonmacrophage LXR expression, contribute to this transport.142 An intestine-specific LXRα/β agonist, GW6340, promoted macrophage-specific reverse cholesterol transport, augmenting the fecal excretion of radiolabeled sterol by 52% via increased intestinal HDL production and intestinal excretion of HDL-derived cholesterol.142 Consistent with these findings, a transgenic mouse model of intestine-specific LXRα overexpression demonstrated increased preβ-HDL levels, enhanced macrophage-specific reverse cholesterol transport, and decreased aortic atheroma volume.143 Intestine-specific expression of LXRα also protected mice taking a high-cholesterol diet from cholesterol and triglyceride accumulation in the liver.143 Intestine-specific LXR agonism, therefore, has potential antiatherogenic effects through the enhancement of reverse cholesterol transport and might avoid the toxicity associated with hepatic LXR activation.

LCAT activators

LCAT esterifies cholesterol, enabling its incorporation into the hydrophobic core of the HDL particle and transforming nascent HDL into spherical mature HDL in the process. Although the importance of LCAT in HDL metabolism is clear, with decreased activity of this enzyme leading to lower HDL-cholesterol levels, its role in reverse cholesterol transport and atherosclerosis in both animals144146 and humans147,148 has been increasingly controversial. The evidence further calls into question the relevance of LCAT in the development of atherosclerosis and throws doubt on the therapeutic potential of stimulating LCAT activity. A cross-sectional examination showed that homozygous and heterozygous carriers of mutations associated with LCAT deficiency have no increase in carotid IMT despite low HDL-cholesterol levels (homozygous 9 mg/dl, heterozygous 39 mg/dl).149 In a case–control analysis of the European Prospective Investigation of Cancer (EPIC)-Norfolk study150 population, low plasma levels of LCAT were not associated with the development of coronary artery disease, in contrast to results from a prior study by the same group examining families with LCAT gene mutations.151 Perhaps the most striking data on this issue are those from the Prevention of Renal and Vascular Endstage Disease (PREVEND) study,152 in which high LCAT activity, despite elevated HDL-cholesterol levels, independently predicted an increased risk of cardiovascular events. These findings are consistent with mouse studies of LCAT overexpression, which demonstrated attenuated macrophage-specific reverse cholesterol transport in vivo and ABCA1-mediated macrophage cholesterol efflux ex vivo.153 Only one LCAT activator has reached early clinical development, ETC-642, and little data are available on the outcomes of treatment with this agent.121 Despite these uncertainties, LCAT activation remains an interesting potential therapeutic target owing to its ability to raise HDL-cholesterol levels and its possible influence on reverse cholesterol transport.

Conclusions

A myriad of novel drugs whose development stems from attempts to leverage the atheroprotective activities of HDL is currently undergoing investigation. Comprehensive assessment of the effects of these drugs requires the analysis of HDL functionality, including HDL flux through reverse cholesterol transport pathways, and not simply the measurement of static circulating HDL-cholesterol levels. Niacin and the CETP inhibitors dalcetrapib and anacetrapib will provide the earliest wave of critical clinical data of HDL-directed strategies in large contemporary cohorts managed with aggressive medical therapy (Table 2). Time will tell whether the coming decade will witness the rise of the HDL-targeted approach or whether the promise of HDL as a viable therapy for atherosclerotic disease will remain unfulfilled.

Table 2.

Ongoing late-stage clinical trials of HDL-directed therapies

Drug Trial Patient population (n) Anticipated completion date
CETP inhibitors
Dalcetrapib DAL-Outcomes72 Acute coronary syndrome (15,600) 2013
Anacetrapib REVEAL78 Stable coronary heart disease or risk equivalent (30,000) 2017
Niacin
Niacin-simvastatin AIM-HIGH83 Stable vascular disease with atherogenic dyslipidemia (3,400) 2012
Niacin-laropiprant HPS2-THRIVE84 Stable vascular disease (25,000) 2013

Abbreviations: AIM-HIGH, Atherothrombosis Intervention in Metabolic Syndrome with Low HDL-C/High Triglycerides and Impact on Global Health Outcomes; CETP, cholesteryl ester transfer protein; DAL, dalcetrapib; HPS2-THRIVE, Heart Protection Study 2—Treatment of HDL to Reduce the Incidence of Vascular Events; REVEAL, Randomized EValuation of the Effects of Anacetrapib through Lipid-modification.

Key points.

  • Four HDL-targeted drug approaches exist: directly augmenting apolipoprotein A-I (apo A-I) levels, indirectly augmenting apo A-I and HDL-cholesterol levels, mimicking the effects of apo A-I, and enhancing reverse cholesterol transport

  • From a pharmacodynamic standpoint, direct augmentation of lipid-poor apo A-I levels arguably represents the most validated HDL therapeutic approach in terms of antiatherogenic potential

  • The clinical efficacy of interventions that raise HDL-cholesterol levels through slowing its metabolism, such as inhibition of cholesteryl ester transfer protein or endothelial lipase, remains to be established

  • The discovery of the niacin receptor GPR109A helped to define the mechanisms underlying the effect of niacin on free fatty acids and flushing, although it is unclear how niacin raises HDL-cholesterol levels

  • With the exception of D-4F, apo A-I mimetic peptides require parenteral administration and, therefore, are likely to be initially targeted to patients at high risk with acute coronary syndromes

  • Intestinal-specific liver X receptor (LXR) agonism or LXRβ-specific agonism might enhance reverse cholesterol transport while avoiding toxicity associated with nonselective LXR activation, namely, hepatic lipogenesis and hypertriglyceridemia

Review criteria.

A search for original, English-language articles published from 1990 to 2010 was performed in PubMed using the following search terms: “high-density lipoprotein”, “HDL”, “apolipoprotein A-I”, “apoA-I”, “reverse cholesterol transport”, “cholesteryl ester transfer protein”, “CETP”, “niacin”, “nicotinic acid receptor”, “lecithin cholesterol acyltransferase”, “LCAT”, “liver X receptor”, “endothelial lipase”. The reference lists of identified articles were also searched for further references. The NIH website ClinicalTrials.gov was searched using the search terms described above, as well as the following search terms: “CSL-111”, “ETC-216”, “dalcetrapib”, “anacetrapib”.

Acknowledgments

This work was supported by grants from the National Heart, Lung and Blood Institute (HL22633 and P50 HL70128) and the National Center for Research Resources (UL1-RR-024134). E. M. deGoma’s salary is partially funded by a National Heart, Lung and Blood Institute grant (K12 HL083772-01).

Footnotes

Competing interests

D. J. Rader declares associations with the following companies: Abbott, AstraZeneca, Bristol-Myers Squibb, Eli Lilly, Johnson & Johnson, Merck, Novartis, and Resverlogix. See the article online for full details of the relationships. E. M. deGoma declares no competing interests.

Author contributions

E. M. deGoma and D. J. Rader contributed equally to the discussion of content for the article, the research of data to include in the manuscript, writing the article and the reviewing and editing of the manuscript before submission.

Contributor Information

Emil M. deGoma, Division of Cardiovascular Medicine, University of Pennsylvania, Penn Tower, 6th Floor, 3400 Spruce Street, Philadelphia, PA 19104, USA

Daniel J. Rader, Department of Medicine, Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, 654 BRB II/III, 451 Curie Boulevard, Philadelphia, PA 19104, USA

References

  • 1.Boden WE, et al. Optimal medical therapy with or without PCI for stable coronary disease. N Engl J Med. 2007;356:1503–1516. doi: 10.1056/NEJMoa070829. [DOI] [PubMed] [Google Scholar]
  • 2.Frye RL, et al. A randomized trial of therapies for type 2 diabetes and coronary artery disease. N Engl J Med. 2009;360:2503–2515. doi: 10.1056/NEJMoa0805796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.deGoma EM, deGoma RL, Rader DJ. Beyond high-density lipoprotein cholesterol levels evaluating high-density lipoprotein function as influenced by novel therapeutic approaches. J Am Coll Cardiol. 2008;51:2199–2211. doi: 10.1016/j.jacc.2008.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wise A, et al. Molecular identification of high and low affinity receptors for nicotinic acid. J Biol Chem. 2003;278:9869–9874. doi: 10.1074/jbc.M210695200. [DOI] [PubMed] [Google Scholar]
  • 5.Zhang Y, et al. Overexpression of apolipoprotein A-I promotes reverse transport of cholesterol from macrophages to feces in vivo. Circulation. 2003;108:661–663. doi: 10.1161/01.CIR.0000086981.09834.E0. [DOI] [PubMed] [Google Scholar]
  • 6.Badimon JJ, Badimon L, Fuster V. Regression of atherosclerotic lesions by high density lipoprotein plasma fraction in the cholesterol-fed rabbit. J Clin Invest. 1990;85:1234–1241. doi: 10.1172/JCI114558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chiesa G, et al. Recombinant apolipoprotein A-I(Milano) infusion into rabbit carotid artery rapidly removes lipid from fatty streaks. Circ Res. 2002;90:974–980. doi: 10.1161/01.res.0000018422.31717.ee. [DOI] [PubMed] [Google Scholar]
  • 8.Parolini C, et al. Dose-related effects of repeated ETC-216 (recombinant apolipoprotein A-I Milano/1-palmitoyl-2-oleoyl phosphatidylcholine complexes) administrations on rabbit lipid-rich soft plaques: in vivo assessment by intravascular ultrasound and magnetic resonance imaging. J Am Coll Cardiol. 2008;51:1098–1103. doi: 10.1016/j.jacc.2007.12.010. [DOI] [PubMed] [Google Scholar]
  • 9.Badimon JJ, Badimon L, Galvez A, Dische R, Fuster V. High density lipoprotein plasma fractions inhibit aortic fatty streaks in cholesterol-fed rabbits. Lab Invest. 1989;60:455–461. [PubMed] [Google Scholar]
  • 10.Shah PK, et al. Effects of recombinant apolipoprotein A-I(Milano) on aortic atherosclerosis in apolipoprotein E-deficient mice. Circulation. 1998;97:780–785. doi: 10.1161/01.cir.97.8.780. [DOI] [PubMed] [Google Scholar]
  • 11.Barter PJ, et al. Antiinflammatory properties of HDL. Circ Res. 2004;95:764–772. doi: 10.1161/01.RES.0000146094.59640.13. [DOI] [PubMed] [Google Scholar]
  • 12.Dimayuga P, et al. Reconstituted HDL containing human apolipoprotein A-1 reduces VCAM-1 expression and neointima formation following periadventitial cuff-induced carotid injury in apoE null mice. Biochem Biophys Res Commun. 1999;264:465–468. doi: 10.1006/bbrc.1999.1278. [DOI] [PubMed] [Google Scholar]
  • 13.Nicholls SJ, et al. Reconstituted high-density lipoproteins inhibit the acute pro-oxidant and proinflammatory vascular changes induced by a periarterial collar in normocholesterolemic rabbits. Circulation. 2005;111:1543–1550. doi: 10.1161/01.CIR.0000159351.95399.50. [DOI] [PubMed] [Google Scholar]
  • 14.Nissen SE, 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. doi: 10.1001/jama.290.17.2292. [DOI] [PubMed] [Google Scholar]
  • 15.Nissen SE, et al. Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial. JAMA. 2004;291:1071–1080. doi: 10.1001/jama.291.9.1071. [DOI] [PubMed] [Google Scholar]
  • 16.Tardif JC, et al. Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial. JAMA. 2007;297:1675–1682. doi: 10.1001/jama.297.15.jpc70004. [DOI] [PubMed] [Google Scholar]
  • 17.Waksman R, et al. A first-in-man, randomized, placebo-controlled study to evaluate the safety and feasibility of autologous delipidated high-density lipoprotein plasma infusions in patients with acute coronary syndrome. J Am Coll Cardiol. 2010;55:2727–2735. doi: 10.1016/j.jacc.2009.12.067. [DOI] [PubMed] [Google Scholar]
  • 18.Sirtori CR, et al. Cardiovascular status of carriers of the apolipoprotein A-I(Milano) mutant: the Limone sul Garda study. Circulation. 2001;103:1949–1954. doi: 10.1161/01.cir.103.15.1949. [DOI] [PubMed] [Google Scholar]
  • 19.Ameli S, et al. Recombinant apolipoprotein A-I Milano reduces intimal thickening after balloon injury in hypercholesterolemic rabbits. Circulation. 1994;90:1935–1941. doi: 10.1161/01.cir.90.4.1935. [DOI] [PubMed] [Google Scholar]
  • 20.Shah PK, et al. High-dose recombinant apolipoprotein A-I(milano) mobilizes tissue cholesterol and rapidly reduces plaque lipid and macrophage content in apolipoprotein e-deficient mice. Potential implications for acute plaque stabilization. Circulation. 2001;103:3047–3050. doi: 10.1161/hc2501.092494. [DOI] [PubMed] [Google Scholar]
  • 21.Franceschini G, et al. Increased cholesterol efflux potential of sera from ApoA-IMilano carriers and transgenic mice. Arterioscler Thromb Vasc Biol. 1999;19:1257–1262. doi: 10.1161/01.atv.19.5.1257. [DOI] [PubMed] [Google Scholar]
  • 22.Wang L, et al. Bone marrow transplantation shows superior atheroprotective effects of gene therapy with apolipoprotein A-I Milano compared with wild-type apolipoprotein A-I in hyperlipidemic mice. J Am Coll Cardiol. 2006;48:1459–1468. doi: 10.1016/j.jacc.2006.07.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Weibel GL, et al. Wild-type ApoA-I and the Milano variant have similar abilities to stimulate cellular lipid mobilization and efflux. Arterioscler Thromb Vasc Biol. 2007;27:2022–2029. doi: 10.1161/ATVBAHA.107.148403. [DOI] [PubMed] [Google Scholar]
  • 24.Lebherz C, Sanmiguel J, Wilson JM, Rader DJ. Gene transfer of wild-type apoA-I and apoA-I Milano reduce atherosclerosis to a similar extent. Cardiovasc Diabetol. 2007;6:15. doi: 10.1186/1475-2840-6-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Parolini C, et al. Apolipoprotein A-I and the molecular variant apoA-I(Milano): evaluation of the antiatherogenic effects in knock-in mouse model. Atherosclerosis. 2005;183:222–229. doi: 10.1016/j.atherosclerosis.2005.03.008. [DOI] [PubMed] [Google Scholar]
  • 26.Alexander ET, et al. Macrophage reverse cholesterol transport in mice expressing ApoA-I Milano. Arterioscler Thromb Vasc Biol. 2009;29:1496–1501. doi: 10.1161/ATVBAHA.109.191379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tardif JC. Emerging high-density lipoprotein infusion therapies: fulfilling the promise of epidemiology? J Clin Lipidol. 2010;4:399–404. doi: 10.1016/j.jacl.2010.08.018. [DOI] [PubMed] [Google Scholar]
  • 28.Lerch PG, Förtsch V, Hodler G, Bolli R. Production and characterization of a reconstituted high density lipoprotein for therapeutic applications. Vox Sang. 1996;71:155–164. doi: 10.1046/j.1423-0410.1996.7130155.x. [DOI] [PubMed] [Google Scholar]
  • 29.Mack WJ, Xiang M, Selzer RH, Hodis HN. Serial quantitative coronary angiography and coronary events. Am Heart J. 2000;139:993–999. doi: 10.1067/mhj.2000.105702. [DOI] [PubMed] [Google Scholar]
  • 30.Jukema JW, et al. Effects of lipid lowering by pravastatin on progression and regression of coronary artery disease in symptomatic men with normal to moderately elevated serum cholesterol levels. The Regression Growth Evaluation Statin Study (REGRESS) Circulation. 1995;91:2528–2540. doi: 10.1161/01.cir.91.10.2528. [DOI] [PubMed] [Google Scholar]
  • 31.Waters D, et al. Effects of monotherapy with an HMG-CoA reductase inhibitor on the progression of coronary atherosclerosis as assessed by serial quantitative arteriography. The Canadian Coronary Atherosclerosis Intervention Trial. Circulation. 1994;89:959–968. doi: 10.1161/01.cir.89.3.959. [DOI] [PubMed] [Google Scholar]
  • 32.Shaw JA, et al. Infusion of reconstituted high-density lipoprotein leads to acute changes in human atherosclerotic plaque. Circ Res. 2008;103:1084–1091. doi: 10.1161/CIRCRESAHA.108.182063. [DOI] [PubMed] [Google Scholar]
  • 33.Patel S, et al. Reconstituted high-density lipoprotein increases plasma high-density lipoprotein anti-inflammatory properties and cholesterol efflux capacity in patients with type 2 diabetes. J Am Coll Cardiol. 2009;53:962–971. doi: 10.1016/j.jacc.2008.12.008. [DOI] [PubMed] [Google Scholar]
  • 34.Calkin AC, et al. Reconstituted high-density lipoprotein attenuates platelet function in individuals with type 2 diabetes mellitus by promoting cholesterol efflux. Circulation. 2009;120:2095–2104. doi: 10.1161/CIRCULATIONAHA.109.870709. [DOI] [PubMed] [Google Scholar]
  • 35.CSL Behring Research & Development Briefing [online] 2009 http://www.csl.com.au/docs/112/54/Print%20Version%20CSL%20Dec%202009%20RD%20Presentation.pdf.
  • 36.Sacks FM, et al. Selective delipidation of plasma HDL enhances reverse cholesterol transport in vivo. J Lipid Res. 2009;50:894–907. doi: 10.1194/jlr.M800622-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bailey D, et al. RVX-208: a small molecule that increases apolipoprotein A-I and high-density lipoprotein cholesterol in vitro and in vivo. J Am Coll Cardiol. 2010;55:2580–2589. doi: 10.1016/j.jacc.2010.02.035. [DOI] [PubMed] [Google Scholar]
  • 38.Miller R. Oral inducer of apoA-1 synthesis RVX-208 misses end point but provides reasons for optimism [online] 2010 http://www.theheart.org/article/1153279.doc.
  • 39.Neeli H, Rader DJ. Cholesteryl ester transfer protein (CETP) inhibitors: is there life after torcetrapib? Cardiol Clin. 2008;26:537–546. doi: 10.1016/j.ccl.2008.06.005. [DOI] [PubMed] [Google Scholar]
  • 40.Boekholdt SM, et al. Plasma levels of cholesteryl ester transfer protein and the risk of future coronary artery disease in apparently healthy men and women: the prospective EPIC (European Prospective Investigation into Cancer and nutrition)-Norfolk population study. Circulation. 2004;110:1418–1423. doi: 10.1161/01.CIR.0000141730.65972.95. [DOI] [PubMed] [Google Scholar]
  • 41.Brousseau ME, et al. Cholesteryl ester transfer protein TaqI B2B2 genotype is associated with higher HDL cholesterol levels and lower risk of coronary heart disease end points in men with HDL deficiency: Veterans Affairs HDL Cholesterol Intervention Trial. Arterioscler Thromb Vasc Biol. 2002;22:1148–1154. doi: 10.1161/01.atv.0000024566.57589.2e. [DOI] [PubMed] [Google Scholar]
  • 42.Lu B, et al. Causes of interscan variability of coronary artery calcium measurements at electron-beam CT. Acad Radiol. 2002;9:654–661. doi: 10.1016/s1076-6332(03)80310-0. [DOI] [PubMed] [Google Scholar]
  • 43.Ordovas JM, et al. Association of cholesteryl ester transfer protein-TaqIB polymorphism with variations in lipoprotein subclasses and coronary heart disease risk: the Framingham study. Arterioscler Thromb Vasc Biol. 2000;20:1323–1329. doi: 10.1161/01.atv.20.5.1323. [DOI] [PubMed] [Google Scholar]
  • 44.Thompson A, et al. Association of cholesteryl ester transfer protein genotypes with CETP mass and activity, lipid levels, and coronary risk. JAMA. 2008;299:2777–2788. doi: 10.1001/jama.299.23.2777. [DOI] [PubMed] [Google Scholar]
  • 45.Borggreve SE, et al. High plasma cholesteryl ester transfer protein levels may favour reduced incidence of cardiovascular events in men with low triglycerides. Eur Heart J. 2007;28:1012–1018. doi: 10.1093/eurheartj/ehm062. [DOI] [PubMed] [Google Scholar]
  • 46.Borggreve SE, et al. An increased coronary risk is paradoxically associated with common cholesteryl ester transfer protein gene variations that relate to higher high-density lipoprotein cholesterol: a population-based study. J Clin Endocrinol Metab. 2006;91:3382–3388. doi: 10.1210/jc.2005-2322. [DOI] [PubMed] [Google Scholar]
  • 47.Hirano K, et al. Genetic cholesteryl ester transfer protein deficiency is extremely frequent in the Omagari area of Japan. Marked hyperalphalipoproteinemia caused by CETP gene mutation is not associated with longevity. Arterioscler Thromb Vasc Biol. 1997;17:1053–1059. doi: 10.1161/01.atv.17.6.1053. [DOI] [PubMed] [Google Scholar]
  • 48.Gaofu Q, et al. Vaccinating rabbits with a cholesteryl ester transfer protein (CETP) B-Cell epitope carried by heat shock protein-65 (HSP65) for inducing anti-CETP antibodies and reducing aortic lesions in vivo. J Cardiovasc Pharmacol. 2005;45:591–598. doi: 10.1097/01.fjc.0000161402.91456.70. [DOI] [PubMed] [Google Scholar]
  • 49.Morehouse LA, et al. Inhibition of CETP activity by torcetrapib reduces susceptibility to diet-induced atherosclerosis in New Zealand White rabbits. J Lipid Res. 2007;48:1263–1272. doi: 10.1194/jlr.M600332-JLR200. [DOI] [PubMed] [Google Scholar]
  • 50.Okamoto H, et al. A cholesteryl ester transfer protein inhibitor attenuates atherosclerosis in rabbits. Nature. 2000;406:203–207. doi: 10.1038/35018119. [DOI] [PubMed] [Google Scholar]
  • 51.Rittershaus CW, et al. Vaccine-induced antibodies inhibit CETP activity in vivo and reduce aortic lesions in a rabbit model of atherosclerosis. Arterioscler Thromb Vasc Biol. 2000;20:2106–2112. doi: 10.1161/01.atv.20.9.2106. [DOI] [PubMed] [Google Scholar]
  • 52.Sugano M, et al. Effect of antisense oligonucleotides against cholesteryl ester transfer protein on the development of atherosclerosis in cholesterol-fed rabbits. J Biol Chem. 1998;273:5033–5036. doi: 10.1074/jbc.273.9.5033. [DOI] [PubMed] [Google Scholar]
  • 53.Barter PJ, et al. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007;357:2109–2122. doi: 10.1056/NEJMoa0706628. [DOI] [PubMed] [Google Scholar]
  • 54.Bots ML, et al. Torcetrapib and carotid intima-media thickness in mixed dyslipidaemia (RADIANCE 2 study): a randomised, double-blind trial. Lancet. 2007;370:153–160. doi: 10.1016/S0140-6736(07)61088-5. [DOI] [PubMed] [Google Scholar]
  • 55.Nicholls SJ, Tuzcu EM, Brennan DM, Tardif JC, Nissen SE. Cholesteryl ester transfer protein inhibition, high-density lipoprotein raising, and progression of coronary atherosclerosis: insights from ILLUSTRATE (Investigation of Lipid Level Management Using Coronary Ultrasound to Assess Reduction of Atherosclerosis by CETP Inhibition and HDL Elevation) Circulation. 2008;118:2506–2514. doi: 10.1161/CIRCULATIONAHA.108.790733. [DOI] [PubMed] [Google Scholar]
  • 56.Rader DJ. Illuminating HDL—is it still a viable therapeutic target? N Engl J Med. 2007;357:2180–2183. doi: 10.1056/NEJMe0707210. [DOI] [PubMed] [Google Scholar]
  • 57.Barter P. Lessons learned from the Investigation of Lipid Level Management to Understand its Impact in Atherosclerotic Events (ILLUMINATE) trial. Am J Cardiol. 2009;104 (10 Suppl):10E–15E. doi: 10.1016/j.amjcard.2009.09.014. [DOI] [PubMed] [Google Scholar]
  • 58.Matsuura F, Wang N, Chen W, Jiang XC, Tall AR. HDL from CETP-deficient subjects shows enhanced ability to promote cholesterol efflux from macrophages in an apoE- and ABCG1-dependent pathway. J Clin Invest. 2006;116:1435–1442. doi: 10.1172/JCI27602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Forrest MJ, 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. doi: 10.1038/bjp.2008.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ishigami M, et al. Large and cholesteryl ester-rich high-density lipoproteins in cholesteryl ester transfer protein (CETP) deficiency can not protect macrophages from cholesterol accumulation induced by acetylated low-density lipoproteins. J Biochem. 1994;116:257–262. doi: 10.1093/oxfordjournals.jbchem.a124516. [DOI] [PubMed] [Google Scholar]
  • 61.Yvan-Charvet L, et al. Inhibition of cholesteryl ester transfer protein by torcetrapib modestly increases macrophage cholesterol efflux to HDL. Arterioscler Thromb Vasc Biol. 2007;27:1132–1138. doi: 10.1161/ATVBAHA.106.138347. [DOI] [PubMed] [Google Scholar]
  • 62.Yvan-Charvet L, et al. Cholesterol efflux potential and antiinflammatory properties of high-density lipoprotein after treatment with niacin or anacetrapib. Arterioscler Thromb Vasc Biol. 2010;30:1430–1438. doi: 10.1161/ATVBAHA.110.207142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Tanigawa H, et al. Expression of cholesteryl ester transfer protein in mice promotes macrophage reverse cholesterol transport. Circulation. 2007;116:1267–1273. doi: 10.1161/CIRCULATIONAHA.107.704254. [DOI] [PubMed] [Google Scholar]
  • 64.Vergeer M, Stroes ES. The pharmacology and off-target effects of some cholesterol ester transfer protein inhibitors. Am J Cardiol. 2009;104 (10 Suppl):32E–38E. doi: 10.1016/j.amjcard.2009.09.017. [DOI] [PubMed] [Google Scholar]
  • 65.Niesor EJ, et al. Modulating cholesteryl ester transfer protein activity maintains efficient pre-β-HDL formation and increases reverse cholesterol transport. J Lipid Res. 2010;51:3443–3454. doi: 10.1194/jlr.M008706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.de Grooth GJ, et al. Efficacy and safety of a novel cholesteryl ester transfer protein inhibitor, JTT-705, in humans: a randomized phase II dose-response study. Circulation. 2002;105:2159–2165. doi: 10.1161/01.cir.0000015857.31889.7b. [DOI] [PubMed] [Google Scholar]
  • 67.Kuivenhoven JA, et al. Effectiveness of inhibition of cholesteryl ester transfer protein by JTT-705 in combination with pravastatin in type II dyslipidemia. Am J Cardiol. 2005;95:1085–1088. doi: 10.1016/j.amjcard.2004.12.064. [DOI] [PubMed] [Google Scholar]
  • 68.Stein EA, et al. Safety and tolerability of dalcetrapib (RO4607381/JTT-705): results from a 48-week trial. Eur Heart J. 2010;31:480–488. doi: 10.1093/eurheartj/ehp601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.ClinicalTrials.gov. A study of the effect of dalcetrapib on artherosclerotic disease in patients with coronary artery disease [online] 2010 http://clinicaltrials.gov/ct2/show/NCT01059682.
  • 70.ClinicalTrials.gov. A study of the effect of RO4607381 on atherosclerotic plaque in patients with coronary heart disease [online] 2010 http://clinicaltrials.gov/ct2/show/NCT00655473.
  • 71.ClinicalTrials.gov. A study assessing the effect of RO4607381 on vascular function in patients with coronary heart disease (CHD) or CHD-risk equivalent patients [online] 2010 http://clinicaltrials.gov/ct2/show/NCT00655538.
  • 72.ClinicalTrials.gov. A study of RO4607381 in stable coronary heart disease patients with recent acute coronary syndrome [online] 2010 http://clinicaltrials.gov/ct2/show/NCT00658515.
  • 73.Ranalletta M, et al. Biochemical characterization of cholesteryl ester transfer protein inhibitors. J Lipid Res. 2010;51:2739–2752. doi: 10.1194/jlr.M007468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Krishna R, et al. Effect of the cholesteryl ester transfer protein inhibitor, anacetrapib, on lipoproteins in patients with dyslipidaemia and on 24-h ambulatory blood pressure in healthy individuals: two double-blind, randomised placebo-controlled phase I studies. Lancet. 2007;370:1907–1914. doi: 10.1016/S0140-6736(07)61813-3. [DOI] [PubMed] [Google Scholar]
  • 75.Bloomfield D, et al. Efficacy and safety of the cholesteryl ester transfer protein inhibitor anacetrapib as monotherapy and coadministered with atorvastatin in dyslipidemic patients. Am Heart J. 2009;157:352–360. doi: 10.1016/j.ahj.2008.09.022. [DOI] [PubMed] [Google Scholar]
  • 76.Cannon CP, et al. Design of the DEFINE trial: determining the EFficacy and tolerability of CETP INhibition with AnacEtrapib. Am Heart J. 2009;158:513–519. doi: 10.1016/j.ahj.2009.07.028. [DOI] [PubMed] [Google Scholar]
  • 77.Cannon CP, et al. Safety of anacetrapib in patients with or at high risk for coronary heart disease. N Engl J Med. 2010;363:2406–2415. doi: 10.1056/NEJMoa1009744. [DOI] [PubMed] [Google Scholar]
  • 78.ClinicalTrials.gov. REVEAL: Randomized EValuation of the Effects of Anacetrapib through Lipid-modification [online] 2010 http://clinicaltrials.gov/ct2/show/NCT01252953.
  • 79.Taylor AJ, Sullenberger LE, Lee HJ, Lee JK, Grace KA. Arterial Biology for the Investigation of the Treatment Effects of Reducing Cholesterol (ARBITER) 2: a double-blind, placebo-controlled study of extended-release niacin on atherosclerosis progression in secondary prevention patients treated with statins. Circulation. 2004;110:3512–3517. doi: 10.1161/01.CIR.0000148955.19792.8D. [DOI] [PubMed] [Google Scholar]
  • 80.Brown BG, et al. Simvastatin and niacin, antioxidant vitamins, or the combination for the prevention of coronary disease. N Engl J Med. 2001;345:1583–1592. doi: 10.1056/NEJMoa011090. [DOI] [PubMed] [Google Scholar]
  • 81.Canner PL, et al. Fifteen year mortality in Coronary Drug Project patients: long-term benefit with niacin. J Am Coll Cardiol. 1986;8:1245–1255. doi: 10.1016/s0735-1097(86)80293-5. [DOI] [PubMed] [Google Scholar]
  • 82.Villines TC, et al. The ARBITER 6-HALTS Trial (Arterial Biology for the Investigation of the Treatment Effects of Reducing Cholesterol 6-HDL and LDL Treatment Strategies in Atherosclerosis): final results and the impact of medication adherence, dose, and treatment duration. J Am Coll Cardiol. 2010;55:2721–2726. doi: 10.1016/j.jacc.2010.03.017. [DOI] [PubMed] [Google Scholar]
  • 83.ClinicalTrials.gov. Niacin plus statin to prevent vascular events [online] 2010 http://clinicaltrials.gov/ct2/show/NCT00120289.
  • 84.ClinicalTrials.gov. Treatment of HDL to reduce the incidence of vascular events HPS2-THRIVE [online] 2010 http://clinicaltrials.gov/ct2/show/NCT00461630.
  • 85.Altschul R, Hoffer A, Stephen JD. Influence of nicotinic acid on serum cholesterol in man. Arch Biochem. 1955;54:558–559. doi: 10.1016/0003-9861(55)90070-9. [DOI] [PubMed] [Google Scholar]
  • 86.Jin FY, Kamanna VS, Kashyap ML. Niacin decreases removal of high-density lipoprotein apolipoprotein A-I but not cholesterol ester by Hep G2 cells. Implication for reverse cholesterol transport. Arterioscler Thromb Vasc Biol. 1997;17:2020–2028. doi: 10.1161/01.atv.17.10.2020. [DOI] [PubMed] [Google Scholar]
  • 87.Shepherd J, Packard CJ, Patsch JR, Gotto AM, Jr, Taunton OD. Effects of nicotinic acid therapy on plasma high density lipoprotein subfraction distribution and composition and on apolipoprotein A metabolism. J Clin Invest. 1979;63:858–867. doi: 10.1172/JCI109385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Blum CB, et al. High density lipoprotein metabolism in man. J Clin Invest. 1977;60:795–807. doi: 10.1172/JCI108833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lamon-Fava S, et al. Extended-release niacin alters the metabolism of plasma apolipoprotein (Apo) A-I and ApoB-containing lipoproteins. Arterioscler Thromb Vasc Biol. 2008;28:1672–1678. doi: 10.1161/ATVBAHA.108.164541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Tunaru S, et al. PUMA-G and HM74 are receptors for nicotinic acid and mediate its anti-lipolytic effect. Nat Med. 2003;9:352–355. doi: 10.1038/nm824. [DOI] [PubMed] [Google Scholar]
  • 91.Maccubbin D, et al. Flushing profile of extended-release niacin/laropiprant versus gradually titrated niacin extended-release in patients with dyslipidemia with and without ischemic cardiovascular disease. Am J Cardiol. 2009;104:74–81. doi: 10.1016/j.amjcard.2009.02.047. [DOI] [PubMed] [Google Scholar]
  • 92.Hanson J, et al. Nicotinic acid- and monomethyl fumarate-induced flushing involves GPR109A expressed by keratinocytes and COX-2-dependent prostanoid formation in mice. J Clin Invest. 2010;120:2910–2919. doi: 10.1172/JCI42273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Dunbar RL, Gelfand JM. Seeing red: flushing out instigators of niacin-associated skin toxicity. J Clin Invest. 2010;120:2651–2655. doi: 10.1172/JCI44098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Richman JG, et al. Nicotinic acid receptor agonists differentially activate downstream effectors. J Biol Chem. 2007;282:18028–18036. doi: 10.1074/jbc.M701866200. [DOI] [PubMed] [Google Scholar]
  • 95.Lai E, et al. Effects of a niacin receptor partial agonist, MK-0354, on plasma free fatty acids, lipids, and cutaneous flushing in humans. J Clin Lipidol. 2008;2:375–383. doi: 10.1016/j.jacl.2008.08.445. [DOI] [PubMed] [Google Scholar]
  • 96.Choi SY, Hirata K, Ishida T, Quertermous T, Cooper AD. Endothelial lipase: a new lipase on the block. J Lipid Res. 2002;43:1763–1769. doi: 10.1194/jlr.r200011-jlr200. [DOI] [PubMed] [Google Scholar]
  • 97.Ishida T, et al. Endothelial lipase is a major determinant of HDL level. J Clin Invest. 2003;111:347–355. doi: 10.1172/JCI16306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Jin W, Millar JS, Broedl U, Glick JM, Rader DJ. Inhibition of endothelial lipase causes increased HDL cholesterol levels in vivo. J Clin Invest. 2003;111:357–362. doi: 10.1172/JCI16146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jaye M, et al. A novel endothelial-derived lipase that modulates HDL metabolism. Nat Genet. 1999;21:424–428. doi: 10.1038/7766. [DOI] [PubMed] [Google Scholar]
  • 100.Edmondson AC, et al. Loss-of-function variants in endothelial lipase are a cause of elevated HDL cholesterol in humans. J Clin Invest. 2009;119:1042–1050. doi: 10.1172/JCI37176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Badellino KO, Wolfe ML, Reilly MP, Rader DJ. Endothelial lipase concentrations are increased in metabolic syndrome and associated with coronary atherosclerosis. PLoS Med. 2006;3:e22. doi: 10.1371/journal.pmed.0030022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Badellino KO, Wolfe ML, Reilly MP, Rader DJ. Endothelial lipase is increased in vivo by inflammation in humans. Circulation. 2008;117:678–685. doi: 10.1161/CIRCULATIONAHA.107.707349. [DOI] [PubMed] [Google Scholar]
  • 103.Tang NP, et al. Protective effect of an endothelial lipase gene variant on coronary artery disease in a Chinese population. J Lipid Res. 2008;49:369–375. doi: 10.1194/jlr.M700399-JLR200. [DOI] [PubMed] [Google Scholar]
  • 104.Vergeer M, et al. Lack of association between common genetic variation in endothelial lipase (LIPG) and the risk for CAD and DVT. Atherosclerosis. 2010;211:558–564. doi: 10.1016/j.atherosclerosis.2010.04.004. [DOI] [PubMed] [Google Scholar]
  • 105.Jensen MK, et al. The T111I variant in the endothelial lipase gene and risk of coronary heart disease in three independent populations. Eur Heart J. 2009;30:1584–1589. doi: 10.1093/eurheartj/ehp145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Brown RJ, et al. Impact of combined deficiency of hepatic lipase and endothelial lipase on the metabolism of both high-density lipoproteins and apolipoprotein B-containing lipoproteins. Circ Res. 2010;107:357–364. doi: 10.1161/CIRCRESAHA.110.219188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Goodman KB, et al. Discovery of potent, selective sulfonylfuran urea endothelial lipase inhibitors. Bioorg Med Chem Lett. 2009;19:27–30. doi: 10.1016/j.bmcl.2008.11.033. [DOI] [PubMed] [Google Scholar]
  • 108.Van Lenten BJ, et al. Apolipoprotein A-I mimetic peptides. Curr Atheroscler Rep. 2009;11:52–57. doi: 10.1007/s11883-009-0008-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Navab M, et al. The oxidation hypothesis of atherogenesis: the role of oxidized phospholipids and HDL. J Lipid Res. 2004;45:993–1007. doi: 10.1194/jlr.R400001-JLR200. [DOI] [PubMed] [Google Scholar]
  • 110.Anantharamaiah GM, et al. Structural requirements for antioxidative and anti-inflammatory properties of apolipoprotein A-I mimetic peptides. J Lipid Res. 2007;48:1915–1923. doi: 10.1194/jlr.R700010-JLR200. [DOI] [PubMed] [Google Scholar]
  • 111.Navab M, 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. doi: 10.1161/hc0302.103711. [DOI] [PubMed] [Google Scholar]
  • 112.Datta G, et al. Effects of increasing hydrophobicity on the physical-chemical and biological properties of a class A amphipathic helical peptide. J Lipid Res. 2001;42:1096–1104. [PubMed] [Google Scholar]
  • 113.Smythies LE, et al. Apolipoprotein A-I mimetic 4F alters the function of human monocyte-derived macrophages. Am J Physiol Cell Physiol. 2010;298:C1538–C1548. doi: 10.1152/ajpcell.00467.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Song X, Fischer P, Chen X, Burton C, Wang J. An apoA-I mimetic peptide facilitates off-loading cholesterol from HDL to liver cells through scavenger receptor BI. Int J Biol Sci. 2009;5:637–646. doi: 10.7150/ijbs.5.637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Van Lenten BJ, et al. Anti-inflammatory apoA-I-mimetic peptides bind oxidized lipids with much higher affinity than human apoA-I. J Lipid Res. 2008;49:2302–2311. doi: 10.1194/jlr.M800075-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Xie Q, Zhao SP, Li F. D-4F, an apolipoprotein A-I mimetic peptide, promotes cholesterol efflux from macrophages via ATP-binding cassette transporter A1. Tohoku J Exp Med. 2010;220:223–228. doi: 10.1620/tjem.220.223. [DOI] [PubMed] [Google Scholar]
  • 117.Buga GM, et al. L-4F alters hyperlipidemic (but not healthy) mouse plasma to reduce platelet aggregation. Arterioscler Thromb Vasc Biol. 2010;30:283–289. doi: 10.1161/ATVBAHA.109.200162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Li X, 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. doi: 10.1161/01.CIR.0000142857.79401.69. [DOI] [PubMed] [Google Scholar]
  • 119.Navab M, et al. D-4F and statins synergize to render HDL antiinflammatory in mice and monkeys and cause lesion regression in old apolipoprotein E-null mice. Arterioscler Thromb Vasc Biol. 2005;25:1426–1432. doi: 10.1161/01.ATV.0000167412.98221.1a. [DOI] [PubMed] [Google Scholar]
  • 120.Bloedon LT, et al. Safety, pharmacokinetics, and pharmacodynamics of oral apoA-I mimetic peptide D-4F in high-risk cardiovascular patients. J Lipid Res. 2008;49:1344–1352. doi: 10.1194/jlr.P800003-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Rader DJ. Molecular regulation of HDL metabolism and function: implications for novel therapies. J Clin Invest. 2006;116:3090–3100. doi: 10.1172/JCI30163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ruchala P, et al. Oxpholipin 11D: an anti-inflammatory peptide that binds cholesterol and oxidized phospholipids. PLoS One. 2010;5:e10181. doi: 10.1371/journal.pone.0010181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bielicki JK, et al. A new HDL mimetic peptide that stimulates cellular cholesterol efflux with high efficiency greatly reduces atherosclerosis in mice. J Lipid Res. 2010;51:1496–1503. doi: 10.1194/jlr.M003665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.D’Souza W, et al. Structure/function relationships of apolipoprotein a-I mimetic peptides: implications for antiatherogenic activities of high-density lipoprotein. Circ Res. 2010;107:217–227. doi: 10.1161/CIRCRESAHA.110.216507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Rader DJ. Liver X receptor and farnesoid X receptor as therapeutic targets. Am J Cardiol. 2007;100 (Suppl 1):S15–S19. doi: 10.1016/j.amjcard.2007.08.008. [DOI] [PubMed] [Google Scholar]
  • 126.Rigamonti E, et al. Liver X receptor activation controls intracellular cholesterol trafficking and esterification in human macrophages. Circ Res. 2005;97:682–689. doi: 10.1161/01.RES.0000184678.43488.9f. [DOI] [PubMed] [Google Scholar]
  • 127.Costet P, Luo Y, Wang N, Tall AR. Sterol-dependent transactivation of the ABC1 promoter by the liver X receptor/retinoid X receptor. J Biol Chem. 2000;275:28240–28245. doi: 10.1074/jbc.M003337200. [DOI] [PubMed] [Google Scholar]
  • 128.Brunham LR, et al. Intestinal ABCA1 directly contributes to HDL biogenesis in vivo. J Clin Invest. 2006;116:1052–1062. doi: 10.1172/JCI27352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Alberti S, Steffensen KR, Gustafsson JA. Structural characterisation of the mouse nuclear oxysterol receptor genes LXRα and LXRβ. Gene. 2000;243:93–103. doi: 10.1016/s0378-1119(99)00555-7. [DOI] [PubMed] [Google Scholar]
  • 130.Repa JJ, et al. Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRα and LXRβ. Genes Dev. 2000;14:2819–2830. doi: 10.1101/gad.844900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Grefhorst A, et al. Stimulation of lipogenesis by pharmacological activation of the liver X receptor leads to production of large, triglyceride-rich very low density lipoprotein particles. J Biol Chem. 2002;277:34182–34190. doi: 10.1074/jbc.M204887200. [DOI] [PubMed] [Google Scholar]
  • 132.Peet DJ, et al. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXRα. Cell. 1998;93:693–704. doi: 10.1016/s0092-8674(00)81432-4. [DOI] [PubMed] [Google Scholar]
  • 133.Alberti S, et al. Hepatic cholesterol metabolism and resistance to dietary cholesterol in LXRβ-deficient mice. J Clin Invest. 2001;107:565–573. doi: 10.1172/JCI9794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Lund EG, et al. Different roles of liver X receptor α and β in lipid metabolism: effects of an α-selective and a dual agonist in mice deficient in each subtype. Biochem Pharmacol. 2006;71:453–463. doi: 10.1016/j.bcp.2005.11.004. [DOI] [PubMed] [Google Scholar]
  • 135.Quinet EM, et al. Liver X receptor (LXR)-β regulation in LXRα-deficient mice: implications for therapeutic targeting. Mol Pharmacol. 2006;70:1340–1349. doi: 10.1124/mol.106.022608. [DOI] [PubMed] [Google Scholar]
  • 136.Bradley MN, et al. Ligand activation of LXRβ reverses atherosclerosis and cellular cholesterol overload in mice lacking LXRα and apoE. J Clin Invest. 2007;117:2337–2346. doi: 10.1172/JCI31909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Fiévet C, Staels B. Liver X receptor modulators: effects on lipid metabolism and potential use in the treatment of atherosclerosis. Biochem Pharmacol. 2009;77:1316–1327. doi: 10.1016/j.bcp.2008.11.026. [DOI] [PubMed] [Google Scholar]
  • 138.Yoshikawa T, et al. Identification of liver X receptor-retinoid X receptor as an activator of the sterol regulatory element-binding protein 1c gene promoter. Mol Cell Biol. 2001;21:2991–3000. doi: 10.1128/MCB.21.9.2991-3000.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Inaba T, et al. Angiopoietin-like protein 3 mediates hypertriglyceridemia induced by the liver X receptor. J Biol Chem. 2003;278:21344–21351. doi: 10.1074/jbc.M213202200. [DOI] [PubMed] [Google Scholar]
  • 140.Jakel H, et al. The liver X receptor ligand T0901317 down-regulates APOA5 gene expression through activation of SREBP-1c. J Biol Chem. 2004;279:45462–45469. doi: 10.1074/jbc.M404744200. [DOI] [PubMed] [Google Scholar]
  • 141.Schaap FG, et al. ApoAV reduces plasma triglycerides by inhibiting very low density lipoprotein-triglyceride (VLDL-TG) production and stimulating lipoprotein lipase-mediated VLDL-TG hydrolysis. J Biol Chem. 2004;279:27941–27947. doi: 10.1074/jbc.M403240200. [DOI] [PubMed] [Google Scholar]
  • 142.Yasuda T, et al. Tissue-specific liver X receptor activation promotes macrophage reverse cholesterol transport in vivo. Arterioscler Thromb Vasc Biol. 2010;30:781–786. doi: 10.1161/ATVBAHA.109.195693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Lo Sasso G, et al. Intestinal specific LXR activation stimulates reverse cholesterol transport and protects from atherosclerosis. Cell Metab. 2010;12:187–193. doi: 10.1016/j.cmet.2010.07.002. [DOI] [PubMed] [Google Scholar]
  • 144.Föger B, et al. Cholesteryl ester transfer protein corrects dysfunctional high density lipoproteins and reduces aortic atherosclerosis in lecithin cholesterol acyltransferase transgenic mice. J Biol Chem. 1999;274:36912–36920. doi: 10.1074/jbc.274.52.36912. [DOI] [PubMed] [Google Scholar]
  • 145.Hoeg JM, et al. Overexpression of lecithin:cholesterol acyltransferase in transgenic rabbits prevents diet-induced atherosclerosis. Proc Natl Acad Sci USA. 1996;93:11448–11453. doi: 10.1073/pnas.93.21.11448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Mertens A, et al. Increased low-density lipoprotein oxidation and impaired high-density lipoprotein antioxidant defense are associated with increased macrophage homing and atherosclerosis in dyslipidemic obese mice: LCAT gene transfer decreases atherosclerosis. Circulation. 2003;107:1640–1646. doi: 10.1161/01.CIR.0000056523.08033.9F. [DOI] [PubMed] [Google Scholar]
  • 147.Santamarina-Fojo S, Lambert G, Hoeg JM, Brewer HB., Jr Lecithin-cholesterol acyltransferase: role in lipoprotein metabolism, reverse cholesterol transport and atherosclerosis. Curr Opin Lipidol. 2000;11:267–275. doi: 10.1097/00041433-200006000-00007. [DOI] [PubMed] [Google Scholar]
  • 148.Rader DJ. Lecithin:cholesterol acyltransferase and atherosclerosis. Another high-density lipoprotein story that doesn’t quite follow the script. Circulation. 2009;120:549–552. doi: 10.1161/CIRCULATIONAHA.109.881979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Calabresi L, et al. Functional lecithin:cholesterol acyltransferase is not required for efficient atheroprotection in humans. Circulation. 2009;120:628–635. doi: 10.1161/CIRCULATIONAHA.108.818143. [DOI] [PubMed] [Google Scholar]
  • 150.Holleboom AG, et al. Plasma levels of lecithin:cholesterol acyltransferase and risk of future coronary artery disease in apparently healthy men and women: a prospective case-control analysis nested in the EPIC-Norfolk population study. J Lipid Res. 2010;51:416–421. doi: 10.1194/jlr.P900038-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hovingh GK, et al. Compromised LCAT function is associated with increased atherosclerosis. Circulation. 2005;112:879–884. doi: 10.1161/CIRCULATIONAHA.105.540427. [DOI] [PubMed] [Google Scholar]
  • 152.Dullaart RP, Perton F, van der Klauw MM, Hillege HL, Sluiter WJ. High plasma lecithin:cholesterol acyltransferase activity does not predict low incidence of cardiovascular events: possible attenuation of cardioprotection associated with high HDL cholesterol. Atherosclerosis. 2010;208:537–542. doi: 10.1016/j.atherosclerosis.2009.07.042. [DOI] [PubMed] [Google Scholar]
  • 153.Tanigawa H, et al. Lecithin:cholesterol acyltransferase expression has minimal effects on macrophage reverse cholesterol transport in vivo. Circulation. 2009;120:160–169. doi: 10.1161/CIRCULATIONAHA.108.825109. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES