Sunteți pe pagina 1din 17

QUARTERLY FOCUS ISSUE: PREVENTION/OUTCOMES State-of-the-Art Paper

High-Density Lipoprotein and Coronary Heart Disease


Current and Future Therapies
Pradeep Natarajan, MD,* Kausik K. Ray, MD, Christopher P. Cannon, MD*
Boston, Massachusetts; and Cambridge, England
Coronary heart disease remains a major cause of worldwide morbidity and mortality despite therapeutic ad-
vances that control many risk factors such as low-density lipoprotein cholesterol to levels lower than previously
possible. Population studies have consistently demonstrated an inverse association between high-density li-
poprotein cholesterol (HDL-C) levels with the risk of coronary heart disease. As a result, HDL-C is gaining increas-
ing interest as a therapeutic target. In this review, we explore the protective mechanisms of HDL and how cur-
rent and future therapies harness these benecial properties. We offer a biological framework to understand
treatment strategies as well as their resultant successes and failures to guide management and future direc-
tions. At present, raising HDL-C level holds great promise, on the basis of epidemiology and initial trials, but we
await the outcomes of the many large clinical outcomes trials currently under way to dene the clinical role of
older and novel therapies to raise HDL-C level. (J Am Coll Cardiol 2010;55:128399) 2010 by the American
College of Cardiology Foundation
Currently, our therapy for lipid modication for atheroscle-
rosis treatment and prevention focuses on lowering low-
density lipoprotein cholesterol (LDL-C). Lipid-lowering
treatment directed at LDL-C with standard doses of
3-hydroxy-3-methylglutaryl coenzyme A reductase inhibi-
tors (statins) has resulted in a relative risk reduction of
one-third in major vascular events as compared with placebo
(1). In patients at very high risk for vascular events, intensive
lipid-lowering has been shown to be benecial compared
with standard therapy (24). However, despite such notable
progress, cardiovascular disease continues to be the leading
cause of death and disease worldwide. As a result, novel
strategies to address this residual risk have been sought.
The Framingham Heart Study in the 1980s demon-
strated that the risk of coronary heart disease (CHD) was
signicantly lower among persons with higher levels of
high-density lipoprotein cholesterol (HDL-C) (normal
range 40 to 60 mg/dl) (5). A number of studies have
supported this inverse correlation between HDL-C and
CHD (59); hence, HDL-C has quickly evolved as one of
the traditional risk factors used by clinicians to predict risk
of incident CHD (4). An estimated 1 mg/dl higher
HDL-C is associated with a 2% lower risk of CHD for men
and a 3% lower risk for women (10). Current guidelines
recommend lowering nonHDL-C as a secondary target
after LDL-C lowering in recognition of the atherogenicity
of lipoproteins associated with hypertriglyceridemia (4).
Traditional lipid-lowering therapy appears to lower LDL-C
and nonHDL-C similarly, and these parameters are often
not distinguished in trials. Even after LDL-C is aggressively
controlled to very low levels with statin therapy, low
HDL-C still remains a signicant cardiovascular risk factor
(11,12). As a result, there has been increasing interest in
HDL-C as a therapeutic target (13).
Prevalence
The prevalence of low levels of HDL-C (e.g., 40 mg/dl)
varies geographically (14) and is particularly high in Latin
American countries, where the prevalence is as high as 46%
in men (15). Over a 7-year period, this prevalence has
continued to rise in Mexico by 2% to 3% (16). Approxi-
mately 25% of men in the United Kingdom have low levels
of HDL-C (17). A more recent Pan-European survey
suggests that the prevalence throughout Europe is similar to
that in the U.S., with low HDL-C levels in nearly one-half
of the Dutch (18). Although a signicant number in China
have low HDL-C levels, the prevalence is much lower than
in North America, with 7% of men and 2% of women
having low levels (19). In the international study evaluating
From the *Department of Medicine and the TIMI Study Group, Cardiovascular
Division, Brigham & Womens Hospital/Harvard Medical School, Boston, Massa-
chusetts; and the Department of Public Health and Primary Care, University of
Cambridge, Cambridge, England. Dr. Natarajan receives royalties from Artery
Therapeutics. Dr. Ray receives research grant support from Bristol-Myers Squibb and
Pzer, and honoraria for lectures and/or consultancy from Merck SP, MSD,
AstraZeneca, Pzer, Lilly, Daiichi Sankyo, Roche, Novartis, and Novo Nordisk. Dr.
Cannon receives research grant support from Accumetrics, AstraZeneca, Bristol-
Myers Squibb/Sano Partnership, GlaxoSmithKline, Intekrin Therapeutics, Merck,
Merck/Schering-Plough Partnership, Novartis, and Takeda, and is a clinical advisor
to and has equity holdings in Automedics Medical Systems.
Manuscript received November 25, 2009; revised manuscript received January 4,
2010, accepted January 4, 2010.
Journal of the American College of Cardiology Vol. 55, No. 13, 2010
2010 by the American College of Cardiology Foundation ISSN 0735-1097/10/$36.00
Published by Elsevier Inc. doi:10.1016/j.jacc.2010.01.008
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
risk factors for acute myocardial
infarction known as the INTER-
HEART study, it was recently
shown that there is specically a
higher prevalence of low HDL-C
levels in South Asians compared
with other Asians (20).
Biological Role of High-
Density Lipoprotein (HDL)
The equilibrium between athero-
genic lipoproteins, such as low-
density lipoprotein (LDL), and
antiatherogenic HDL can affect
endothelial dysfunction and in-
ammation (21). In the face of
elevated LDL-C levels, HDL-C
appears to progressively protect
against atherosclerosis (21).
Circulating HDL particles are
greatly heterogeneous with a very
complex metabolic prole (Table 1).
The various HDL subclasses vary
in quantitative and qualitative content of lipids, apolipopro-
teins, enzymes, and lipid transfer proteins, resulting in
differences in shape, density, size, charge, and antigenicity.
Discoid HDL particles are small, lipid-poor particles made
of apolipoprotein in a monolayer of phospholipids and free
cholesterol (2123). Spherical HDL particles are larger, are
made from discoid HDL, and contain a hydrophobic core of
cholesterol esters (2123).
Reverse cholesterol transport, the process of transporting
excess cholesterol from the arterial walls foam macrophages
to the liver, bile, and feces is one of HDLs antiatherogenic
properties (24,25) (Fig. 1). Lipid-free HDL, or apolipopro-
tein A-I, mediates this through the adenosine triphosphate
(ATP)-cassette binding transporter (ABC) A1. Then ester-
ication of HDL-C by lecithin-cholesterol acyltransferase
(LCAT) generates more mature HDL particles, including
small, dense spherical HDL3 and large, less dense spherical
HDL2 (22,24). These mature HDL particles may induce
further cholesterol efux through ABCG1 and ABCG4
(21,24,26). HDL-C measures the cholesterol content of
nascent HDL, HDL2, and HDL3 particles and is, there-
fore, a crude marker of reverse cholesterol transport. The
smaller HDL3 particles more efciently promote cholesterol
efux through the ABCA1 pathway than do their larger
counterparts, but they are equally as effective through the
ABCG1 pathway (2729).
After cholesterol efux from the plaque, reverse cholesterol
transport is completed by transport back to the liver. Through
the scavenger receptor B1, hepatocytes and steroid-producing
cells can uptake these mature HDL particles (25,30). Further-
more, cholesterol esters from these particles may be trans-
ferred to apolipoprotein B-containing particles, such as
LDL or very low-density lipoprotein (VLDL), through the
cholesterol ester transfer protein (CETP) (22,24,25). He-
patic LDL receptors can then scavenge these apolipoprotein
Bcontaining particles enriched with cholesterol esters,
further contributing to reverse cholesterol transport (25).
The subsequent HDL particle enriched with triglycerides
may regenerate small HDL particles through hydrolysis
from hepatic lipase (21,24,25).
In addition to HDL protecting against atherosclerosis
through reverse cholesterol transport, HDLs antioxidative ac-
tivity further protects against atherosclerosis (21,31) (Table 2).
Apolipoprotein A-I is a major factor in this process (31). In
addition to HDL-C esterication, LCAT can also hydrolyze
oxidized phospholipids of LDL (22,31,32).
For monocytes to gain entry into the subendothelial space
to promote atherosclerosis, their circulation must be ar-
rested, which is accomplished by binding to adhesion
molecules in the blood vessel wall (33,34). Expression of
these adhesion molecules depends on inammatory cyto-
kines and vascular injury, and is up-regulated in atheroscle-
rosis (3538). Animal models have demonstrated that HDL
inhibits the expression of adhesion molecules (39,40). HDL
can also directly inhibit the migration of these monocytes
into the subendothelial space (41). In the endothelium,
nitric oxide protects against inammation and activation
(42). HDL promotes vasoprotection by enhancing nitric
oxide synthase and thereby increasing the production of
nitric oxide (42,43). In addition to protection against
platelet activation through endothelial protection, HDL
inhibits the coagulation cascade through serine protease
protein C, which inactivates factors Va and VIIa (42,4446).
Dysfunctional HDL
Structural and functional changes accompany HDL in the
setting of acute or chronic inammation. Leukocyte myelo-
peroxidase may alter the function of normally atheroprotec-
tive molecules into so-called dysfunctional HDL with
proinammatory properties (47). During inammation,
HDL-C levels can be reduced because of increased HDL
catabolism, decreased apolipoprotein A-I synthesis, and
displacement of apolipoprotein A-I from HDL with serum
amyloid A, an acute-phase protein (48,49). Additionally,
platelet-activating acetylhydrolase and LCAT can become
dysfunctional or diminished during inammation, and
among persons with low HDL-C levels such as those with
Abbreviations
and Acronyms
ABC ATP-binding
cassette transporter
ACAT acyl-coenzyme A
cholesterol acyltransferase
ATP adenosine
triphosphate
CETP cholesterol ester
transfer protein
CHD coronary heart
disease
HDL-C high-density
lipoprotein cholesterol
LCAT lecithin-cholesterol
acyltransferase
LDL-C low-density
lipoprotein cholesterol
PPAR peroxisome
proliferator-activated
receptor
VLDL very low-density
lipoprotein
Physical Properties of HDL Particles
Table 1 Physical Properties of HDL Particles
Particle Diameter, nm Density, g/ml
Discoid HDL 8
Spherical HDL3 89 1.1251.21
Spherical HDL2 910 1.0631.125
Discoid high-density lipoprotein (HDL) is quickly degraded when extracted fromplasma. As a result,
when human HDL is processed through density gradient ultracentrifugation, the predominant
subfractions include HDL2 and HDL3. HDL3 is smaller and denser, and is relatively protein rich and
lipid poor (2123).
1284 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
type 2 diabetes mellitus and CHD (31,41,5052). Further-
more, in the metabolic syndrome, hypertriglyceridemic
states and the acute-phase response, largely through CETP,
enrich the triglyceride content of HDL-C (48,53). In turn,
triglyceride-rich HDL-C particles result in a lowering of
circulating HDL-C levels through their inherent instability
and subsequent degradation as well as through hydrolysis
from hepatic lipase (53). Furthermore, triglyceride-rich
HDL-C cannot be taken by hepatocytes through scavenger
receptor B1 as readily, resulting in reduced efcacy of reverse
cholesterol transport (53,54). Therefore, a combination of
metabolic abnormalities and inammatory milieu may result
in dysfunctional HDL (31,55).
Nonpharmacological Interventions
Observational and interventional studies have assessed the
relationship between lifestyle factors and circulating
HDL-C levels.
Aerobic exercise. In young adult women, moderate activity
was shown to signicantly increase HDL-C over the course
of 6 months (56). In a population of men and women
ranging from 50 to 65 years of age, however, an increased
frequency of exercise resulted in the highest HDL-C levels
Figure 1 Reverse Cholesterol Transport and Therapeutic Targets
Reverse cholesterol transport is one of the primary antiatherogenic properties of high-density lipoprotein (HDL) (24,25). This efux of cholesterol is largely facilitated
through adenosine triphosphate-binding cassette transporters (ABC) as lecithin-cholesterol acyltransferase (LCAT) helps modify these particles into more mature HDL2
and HDL3 particles (22,24). Furthermore, cholesterol ester transfer protein (CETP) transfers additional cholesterol esters from apolipoprotein B-containing particles to
HDL (22,24,25). Subsequently, these HDL particles are taken up by hepatocytes through scavenger receptor B1 (SRB1) (25,30). The various current pharmacologic
agents attempt to increase HDL-C levels by affecting different steps involved in reverse cholesterol transport. ACAT acyl-coenzyme A cholesterol acyltransferase;
CE cholesterol ester; FC free cholesterol; LDL low-density lipoprotein; PL phospholipid; PPAR peroxisome proliferator-activated receptor; TG triglycerides;
VLDL very low-density lipoprotein.
Properties of HDL
Table 2 Properties of HDL
Function Mechanism
Reverse cholesterol transport Cholesterol efux through ABCA1, ABCG1,
ABCG4 (21,22,2429)
LDL antioxidation Activation of LCAT (29,31,32)
Endothelial protection Inhibition of adhesion molecule expression
(39,40), prevention of monocyte
chemotaxis (41), nitric oxide synthase
stimulation (42,43)
Antiplatelet activity Protection against endothelial injury (42)
Anticoagulation Inhibition of factors Va and VIIa (4446)
ABC adenosine triphosphate-binding cassette transporter; HDL high-density lipoprotein;
LCAT lecithin-cholesterol acyltransferase; LDL low-density lipoprotein.
1285 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
(57). Meanwhile, over the short term, such as within the
rst month of aerobic exercise, the anti-inammatory ben-
ets of HDL predominate (58). For example, over the
course of only 3 weeks of exercise, although HDL-C levels
did not increase, HDL preferentially converted to an anti-
inammatory state (58).
Diet. The dietary consumption of saturated fats, unlike
polyunsaturated and monounsaturated fats, promotes ath-
erosclerosis (59). A high saturated fat diet has been shown
to diminish the ability of HDL to protect the endothelium
and reduces vasoreactivity (60,61). In contrast, a high
polyunsaturated fat diet enhances these features (62). In
vitro, HDL suppresses the chemokines that attract inam-
matory cells to the vessel wall (6365).
Purely low fat diets have been shown to decrease
HDL-C to a similar degree as LDL-C is reduced. How-
ever, when unsaturated fat replaces saturated fat and trans-
fats, LDL-C decreases proportionally more, thereby de-
creasing the LDL-to-HDL ratio (62,66). Diets high in
monounsaturated fats, including olive oil and canola oil,
have been shown to reduce LDL-C without adversely
affecting HDL (67). Certain polyunsaturated fats, including
n-3-polyunsaturated fatty acids, largely found in sh oil, can
elevate levels of HDL-C, especially among persons with high
triglycerides (68,69). Grapeseed oil, a byproduct of winemak-
ing, that contains a mixture of polyunsaturated vegetable oils
may increase HDL-C levels by nearly 13% (70).
Glycemic index measures how much a carbohydrate-
containing food increases blood sugar levels and is deter-
mined by ber content, glucose content, and digestibility.
Data suggest that the carbohydrate quality, or glycemic
index, may affect HDL-C levels (71). For instance, the
favorable effect of low glycemic index foods on increases in
HDL-C levels was observed in both the U.S. and Europe
(72,73). In the Nurses Health Study, persons particularly
susceptible to insulin resistance had increases in their
HDL-C when eating foods with low glycemic index (74).
Weight loss. Obesity is the central abnormality that con-
tributes to the metabolic syndrome and is associated with
low HDL-C and high triglyceride levels. Losing weight
through diet or aerobic exercise equally results in an increase
in HDL-C, without signicant differences between the 2
strategies (75). The effect of weight loss from caloric
restriction and regular exercise is particularly benecial for
overweight or obese men and women (76). In a meta-
analysis, subjects actively losing weight initially experience a
reduction in HDL-C levels, but subjects at a stabilized,
reduced weight have HDL-C levels inversely proportional
to their weight (77). This nding is consistent with the
observation that lipoprotein lipase decreases with acute
caloric restriction but increases with weight loss, especially
when the weight is stabilized (78,79). A sustained reduction
in weight can be achieved through various mechanisms, but
all have similar favorable effects on HDL-C levels.
Tobacco cessation. The Framingham study group showed
that cigarette smoking accounted for a drop in HDL-C by
4 mg/dl in men and 6 mg/dl in women (80). A meta-
analysis demonstrated that smokers generally have a 9%
lower HDL-C level and a 6% lower apolipoprotein A-I
level compared with matched nonsmokers (81). This inverse
association of cigarette smoking and HDL-C levels is dose
dependent (8184). Increased CETP activity and the re-
sultant transfer of cholesterol esters from HDL to apoli-
poprotein B-containing particles mediate tobacco smokings
atherogenic lipoprotein changes (85,86). Furthermore, in a
meta-analysis of 27 studies, it was observed that when
participants quit smoking tobacco, their HDL-C levels rose
by 4 mg/dl without a signicant effect on total cholesterol,
LDL-C, or triglyceride levels (87). Additionally, smoking
cessation results in an increase in apolipoprotein A-I (88).
These favorable effects on the lipid prole can be seen as
early as 30 days after quitting smoking (89).
Alcohol. While heavy alcohol intake is associated with
increased all-cause mortality and cardiovascular-related
mortality (9094), moderate alcohol intake appears to have
a protective effect on CHD (9599). Part of this protection
may be mediated by an associated increase in HDL-C
(100103). An increase in both the HDL2 and HDL3
subfractions, but with relatively larger increase in HDL2, is
observed with moderate alcohol consumption (103,104). A
systematic review showed that all forms of alcohol (e.g.,
beer, wine, and spirits) had a favorable cardioprotective
effect (105). However, data suggest that wine consumption
provides more cardioprotection (106). Some of this additive
benet may be from avonoids such as resveritrol that act
as lipoprotein antioxidants (107). The recommendation of
alcohol consumption for cardiovascular protection is limited
by the potential for abuse, dependence, and caloric intake.
Pharmacological Interventions
Various randomized controlled trials have evaluated the
efcacy and safety of pharmacologically modifying HDL
(Table 3).
Statins. The reduction of LDL-C with statins has a
positive effect on the occurrence of cardiovascular events
(108112). A decrease in LDL-C levels from statin therapy
is associated with a decrease in the progression of athero-
sclerosis (113115). Increases in HDL-C between 5% and
15% have been reported with statin-mediated therapy, with
an average increase of 9% (108,110,111,116). However,
the data regarding the clinical signicance of this are
unclear. The statin-induced HDL-C effects are relatively
small compared with the LDL-C effects, so the effects of
statins on HDL-C are likely to be small. Nevertheless, a
recent meta-analysis of statin therapy demonstrated that, in
patients with CHD in whom LDL-C levels are aggressively
reduced to 87.5 mg/dl, an increase in HDL-C by 7.5% is
independently associated with a regression in coronary
atherosclerosis (116). Statins also promote the formation of
a more favorable HDL subfraction prole by creating more
cholesterol-rich HDL particles, perhaps through the reduc-
1286 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
tion of cholesterol transfer from HDL (117120). That may
occur through the reduction in available apolipoprotein
B-containing atherogenic particles to accept cholesterol
esters and down-regulation of CETP expression (121,122).
Niacin. Currently, niacin (vitamin B3) is the most effective
available pharmacologic means to raise HDL-C levels.
Interest in niacin developed 50 years ago when it was
noted to decrease total cholesterol, lower LDL-C, and
raise HDL-C (123,124). Niacin increases HDL-C levels by
15% to 35% with a nonlinear dose-dependent response
(125129). Therefore, the majority of the benecial effects
of niacin occurs at doses of 1 to 1.5 g per day (126,129).
Niacin accomplishes this effect on HDL through multi-
ple mechanisms. It selectively inhibits hepatic clearance of
HDLs apolipoprotein A-I and not the cholesterol esters
(130,131). As a result, the cholesterol-decient apolipopro-
tein A-Icontaining HDL particles are recycled into the
circulation and freed for further reverse cholesterol transport
(130,132). Furthermore, niacin directly inhibits the transfer
of cholesterol esters through CTEP from HDL to apoli-
poprotein B-containing particles by directly inhibiting
CTEP activity as well as by reducing the hepatic synthesis
of CTEP (133). Niacin also targets this pathway by reduc-
ing the lipolysis in adipose tissue. That reduces the release of
free fatty acids into plasma, with a consequent decrease in
free fatty acid uptake by the liver that leads to a secondary
decrease in hepatic triglyceride synthesis. The resulting
decrease in concentration of plasma VLDL results in a
reduction in the transfer of cholesteryl esters from HDL to
VLDL, and hence an increase in concentration of HDL-C
(134). At a more local level, through the cyclic adenosine
monophosphate/protein kinase A secondary messengers,
niacin promotes the transcription of ABCA1 to provide
HDL particles with targets for reverse cholesterol transport
(135). Moreover, niacin tends to promote the formation of
HDL2 through its inhibition of hepatic lipase (136).
The CDP (Coronary Drug Project) evaluated the effect
of niacin on 8,341 men with a history of myocardial
infarction during 1966 to 1975 (137). Over a 5-year
follow-up period, the incidence of nonfatal reinfarction was
reduced by 27%. Even more compelling is that 15 years
later, niacin contributed to a signicant decrease in all-cause
mortality by 9% (138).
Additional clinical studies of niacin have typically been in
the setting of combination therapy. When combined with
colestipol, a bile-acid sequestrant, in the FATS (Familial
Pharmacotherapy and Effects on HDL-C
Table 3 Pharmacotherapy and Effects on HDL-C
Medication Class Mechanism Phase II/III Trials HDL-C Increase Ongoing Large Trials
Statins Reduced cholesterol transfer from HDL through
decreased ApoB-containing particles and
reduced CETP expression (117122)
* 5%15%
Niacin Inhibition of hepatic clearance of ApoA-I, CETP
inhibition, reduced CETP synthesis, reduced
adipose tissue lipolysis, ABCA1 transcription
promotion, hepatic lipase inhibition
(130136)
CDP (137), FATS (139), CLAS (141), Stockholm
Ischaemic Heart Disease Secondary
Prevention Study (143), ARBITER 2 (154),
ARBITER 6-HALTS (155)
15%35% AIM-HIGH (156);
HPS2-THRIVE (157)
Fibrates PPAR agonism resulting in ABCA1
up-regulation and synthesis, and ApoA-I
transcription (122,162167)
Helsinki Heart Study (168), VA-HIT (169),
BIP (171), FIELD (173)
10%15% ACCORD (188)
Thiazolidinediones PPAR agonism resulting in cellular
differentiation in vascular tissue and
adipocytes (190)
PROACTIVE (193), CHICAGO (194),
PERISCOPE (195), RECORD (197)
5%10%
Glitazars PPAR and PPAR agonism (199201) Kendall et al. (205) 15%30% Aleglitazar (211)
CETP inhibitors CETP inhibition (122,212219) ILLUMINATE (228), ILLUSTRATE (232),
RADIANCE1 (234), RADIANCE2 (235)
40%130% dal-OUTCOMES (241),
dal-VESSEL (242),
dal-PLAQUE (243),
DEFINE (246)
ACAT inhibitors ACAT-1 and ACAT-2 inhibition reducing
intracellular cholesterol esterication
(247,248)
ACTIVATE (253), CAPTIVATE (254) 0%5%
Recombinant
HDL infusion
Reverse cholesterol transport (259), increased
nitric oxide bioavailability (257,258)
ERASE (260)
ApoA-I Milano
infusion
Antioxidation from free cysteine residues
(264266)
Nissen et al. (270)
ApoA-I mimetics Saponication by amphipathic alpha-helices
(273,274), ABCA1-dependent reverse
cholesterol transport (273), antioxidation
(281)

ApoA-I up-regulators Promotion of ApoA-I transcription (287)
*There are several phase III trials with statins targeted specically to low-density lipoprotein cholesterol (LDL-C) lowering, which is outside the scope of this review. High-density lipoprotein cholesterol
(HDL-C) levels after infusions were not routinely measured in the ERASE study or in apolipoprotein (Apo) A-I trials (258,268). Only phase I safety data in humans are currently available for the Apo A-I
mimetic, 4F, and the Apo A-I up-regulator, RVX-208 (281,288,289).
ACAT acyl-coenzyme A cholesterol acyltransferase; CETP cholesterol ester transfer protein; PPAR peroxisome proliferator-activated receptor; other abbreviations as in Table 2.
1287 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
Atherosclerosis Treatment Study), HDL-C increased by
43%, with angiographic atherosclerotic regression in 39%
and a 73% reduction in CHD rates over a 2.5-year
follow-up period (139). After 10 years of niacin combined
with both colestipol and lovastatin, there was an 18.5%
absolute risk reduction in all-cause mortality (140). The
CLAS (Cholesterol-Lowering Atherosclerosis Study) also
evaluated the effect of combining colestipol with niacin in
162 men who had prior coronary artery bypass graft surgery,
and demonstrated an HDL-C increase by 37%, with an-
giographic regression of atherosclerosis of 16% at 2 years
and 18% at 4 years (141,142).
Combining niacin with a brate has also been shown to
be effective. In the Stockholm Ischaemic Heart Disease
Secondary Prevention Study, 554 post-myocardial infarc-
tion patients were randomly assigned to placebo or to a
combination of niacin and clobrate, and those in the active
treatment arm experienced a 26% reduction in all-cause
mortality and a 36% reduction in CHD mortality (143).
However, this benecial effect was more correlated with the
extent of reduction in serum triglyceride levels. When
examined among patients with CHD and low HDL-C
levels, triple therapy with cholestyramine, a bile-acid se-
questrant, gembrozil, and niacin resulted in a 36% increase
in HDL-C and a 13.7% reduction in CHD events (144).
A common limiting side effect of niacin is facial pruritis
and ushing caused by prostaglandin-mediated vasodilation
through the G-coupled protein receptor, GPR109A
(132,145). This pathway additionally, at least partially,
mediates the niacin-mediated antilipolytic effects in murine
models (145). Interestingly, ushing may be a marker of an
increased lipid response to niacin (146). Paradoxically, the
presence of ushing as a surrogate for niacins efcacy may
promote adherence through appropriate patient education.
Meanwhile, these ushing symptoms can be ameliorated
with the once-daily extended-release formulation, which is
equally as efcacious as the immediate-release formulation
(147). This ushing can also be addressed by taking aspirin
30 min before dosing and by avoiding spicy foods. Niacin in
its immediate-release or extended-release form is safe and
tolerable for diabetic patients as well (148). However, higher
doses of niacin resulted in a slight worsening of glycemic
control (p 0.048) in the ADVENT (Assessment of
Diabetes Control and Evaluation of the Efcacy of Niaspan
Trial) of type 2 diabetic patients randomly assigned to
niacin or placebo (148). When niacin is given at doses 2.5
g, it is associated with a 4% to 5% increase in ngerstick
glucose levels and may or may not have effects on hemo-
globin A1c, and these effects on hemoglobin A1c may be
transient only during uptitration of doses (149). Guidelines
from the National Cholesterol Education Program, Amer-
ican Heart Association, American Diabetes Association,
and National Lipid Association all consider niacin at doses
2 g daily safe for diabetic patients to manage dyslipidemia,
and the cardiovascular benets may outweigh the risks
(14,150152). Additionally, the National Lipid Association
further asserts that patients with impaired glucose tolerance
without overt diabetes mellitus (e.g., fasting glucose 110 to
125) can safely take niacin with careful monitoring of glycemic
control, similar to that for diabetic patients when niacin
therapy is initiated (152).
Both formulations have been evaluated in combination
with statins. In the HATS (HDL-Atherosclerosis Treat-
ment Study), patients with CHD, HDL-C 35 mg/dl, and
LDL-C 145 mg/dl treated with both simvastatin and
extended-release niacin had an increase in HDL-C levels by
26%, slight regression in proximal coronary artery stenosis
by angiography, and a 90% reduction in CHD events versus
dual placebo, although this represented 9 versus 1 events
(153). In the ARBITER 2 (Arterial Biology for the Inves-
tigation of Treatment Effects of Reducing Cholesterol 2)
trial, extended-release niacin was given to patients with
CHD, HDL-C 45 mg/dl, and LDL-C 120 mg/dl who
were already receiving statin therapy (154). This treatment
resulted in a 21% increase in HDL-C and a trend toward a
decrease in the progression of carotid intima media thick-
ness as assessed by ultrasonography over a 1-year follow-up
period (154). The follow-up study, ARBITER 6 (HDL and
LDL Treatment Strategies in Atherosclerosis), was de-
signed to compare the effects of HDL-Clowering therapy
with extended-release niacin versus additional LDL-C
lowering therapy with ezetimibe on atherosclerosis progres-
sion in patients already receiving statin therapy (155). This
trial was stopped early in 2009 with just 208 evaluable
patients when it was seen that patients receiving niacin had
a signicant reduction in carotid-intima media thickness at
both 8 months and 14 months (155).
Currently, the AIM-HIGH (Atherothrombosis Inter-
vention in Metabolic Syndrome with Low HDL/High
Triglycerides and Impact on Global Health Outcomes)
study has enrolled 3,300 patients with CHD and evidence
of the metabolic syndrome with HDL-C 40 mg/dl and
triglycerides 150 mg/dl not already given statin therapy
(156). In this U.S.Canadian, multicenter, randomized,
double-blind, parallel-group, controlled clinical trial, pa-
tients have been randomly allocated to therapy with simva-
statin alone or simvastatin and extended-release niacin and
are being evaluated over a 5-year period to better dene the
additive effect of HDL-raising therapies. Another trial, the
HPS2-THRIVE (Heart Protection Study 2 Treatment on
HDL to Reduce the Incidence of Vascular Events) is
recruiting 25,000 patients with a history of CHD, stroke, or
peripheral arterial disease and randomizing them to placebo
or a new tablet containing extended-release niacin plus a
specic blocker of prostaglandin D2, called laropiprant, to
prevent the ushing associated with niacin (157).
Fibrates. Fibrates are peroxisome proliferator-activated re-
ceptor (PPAR)- agonists that lower LDL-C by 10% to
20%, lower triglycerides by 25% to 45%, and increase
HDL-C modestly by 10% to 15% (158161). Through the
activation of PPAR, ABCA1 is up-regulated as well as
hepatic HDL synthesis (162,163). The HDL production is
1288 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
largely mediated through PPARs stimulation of the apo-
lipoprotein A-I genes promoter (164166). PPAR ago-
nism may also decrease CETP gene expression, resulting in
lower levels of plasma CETP (122). Additionally, by reduc-
ing triglyceride-rich lipoproteins available to accept choles-
terol esters, HDL-C levels are boosted (122,167).
In the Helsinki Heart study, 4,081 healthy men without
CHD with nonHDL-C 200 mg/dl were randomly
assigned to gembrozil versus placebo (168). LDL-C de-
creased by 11%, triglycerides decreased by 35%, and
HDL-C increased by 11%. Although the more pronounced
effect was on triglycerides, the 34% reduction in CHD
events was signicantly associated with both LDL-C low-
ering and HDL-C elevation. The VA-HIT (Veterans
Affairs High-Density Lipoprotein Cholesterol Intervention
Trial) evaluated patients with CHD, LDL-C 140 mg/dl,
triglycerides 300 mg/dl, and HDL-C 40 mg/dl on
gembrozil (160). LDL-C levels did not differ signicantly
between the study groups over the 5-year study period, so
the 22% reduction in CHD events was attributed to the 6%
increase in HDL-C in a subsequent multivariable analysis
(160,169). It was the rst trial that demonstrated a relation-
ship between HDL-C increase and CHD event reduction
in patients with moderate LDL-C levels. Another analysis
of the VA-HIT study demonstrated that gembrozil ther-
apy for patients with CHD and low HDL-C was cost
effective in terms of the cost of quality-adjusted life-years
saved (170).
The effects of the brates on coronary events have been
variable. Although bezabrate therapy in patients with
CHD and low HDL-C levels increases HDL-C by 18%,
there was only a trend toward the reduction of CHD events
in the BIP (Bezabrate Infarction Protection) study (171).
A 16-year follow-up of this trial demonstrated that subjects
with the highest HDL-C response to therapy had a signif-
icant reduction in the risk of death (172). In the FIELD
(Fenobrate Intervention and Event Lowering in Diabetes)
study, the authors studied the effect of fenobrate therapy
on 9,795 randomized patients with type 2 diabetes mellitus
not receiving statin therapy and with a total cholesterol level
of 251 mg/dl (173). However, fenobrate did not signif-
icantly alter HDL-C levels but it did lower LDL-C and
triglyceride levels. There was no signicant effect on total
coronary events, but a signicant 24% reduction in nonfatal
myocardial infarction was observed. Overall, the FIELD
trial failed to demonstrate a signicant benet of an agent
that predominantly reduces triglycerides in a high-risk
population. That may have resulted from a greater use of
open-label statin therapy for subjects allocated to placebo.
Studies of clobrate have been plagued with concerns
regarding potential safety (174176).
The brates are primarily excreted renally, and there are
retrospective observational data suggesting that they may
result in reversible worsening renal function in patients with
baseline renal impairment (177,178). In severe renal impair-
ment, the drugs half-life is prolonged and is not dialyzable,
and there is concern regarding permanent renal dysfunction
in kidney transplant patients (177,179). Although the
mechanism of inducing renal dysfunction is unclear, inter-
esting data propose that brates result in increased creati-
nine levels from increased creatinine production without an
actual change in creatinine clearance (180). Renal function
in patients with severe nondialysis-dependent renal disease
should be monitored during the initiation of brate therapy
for dyslipidemia, but brates are not contraindicated in
patients with baseline renal impairment.
Given the benets of statin therapy for diabetic patients,
there is little evidence to recommend brates as an alternative
rst-line treatment to statins for the management of dyslipi-
demia among diabetic patients (181,182). The combination of
statins and brates bears the risk of myotoxicity with an
incidence of 0.12%, but there are some data suggesting the
benets of combined statin and brate therapy, particularly for
diabetic patients (14,183185). All brates have been associ-
ated with reports of creatinine kinase elevations and myopathy
when given with statins, but that appears to occur more
frequently with gembrozil than with fenobrate (186). Gem-
brozil, more than fenobrate, inhibits hepatic glucoronida-
tion of various statins, thereby preventing statin clearance and
increasing serum levels of statins in their active form (187).
The ACCORD (Action to Control Cardiovascular Risk in
Diabetes) study is a large-scale trial that is currently prospec-
tively evaluating the safety and efcacy of the combination of
simvastatin and fenobrate in managing dyslipidemia in type 2
diabetes (188).
Thiazolidinediones. The insulin-sensitizing thiazolidinediones,
which include pioglitazone and rosiglitazone, were approved
in the U.S. in the late 1990s for the treatment of type 2
diabetes mellitus. Troglitazone was also a member of this
class but was withdrawn because of idiosyncratic liver injury
(189). These agents are agonists of peroxisome proliferator-
activated receptor (PPAR)-, a nuclear receptor involved in
cellular differentiation, largely in vascular tissue and adipo-
cytes (190). Insulin resistance contributes to the metabolic
syndrome, which includes hyperglycemia, elevation in tri-
glycerides, and reductions in HDL-C levels. As a result,
insulin-sensitizers such as thiazolidinediones were proposed
to not only correct dysglycemia but also to potentially
correct lipid abnormalities (189).
A meta-analysis of 23 randomized trials demonstrated
that pioglitazone increased HDL-C levels by 4.6 mg/dl and
that rosiglitazone increased HDL-C levels by 2.7 mg/dl
(191). However, unlike pioglitazone, rosiglitazone increased
LDL-C and total cholesterol and did not decrease triglyc-
erides (191). Even in patients without overt diabetes mel-
litus but with evidence of insulin resistance, pioglitazone
increases HDL-C levels (192). The PROACTIVE (Pro-
spective Pioglitazone Clinical Trial in Macrovascular
Events) assessed the benet of pioglitazone in a higher risk
population, type 2 diabetes with a hemoglobin A1c level
6.5%, and stable macrovascular disease (193). Pioglita-
zone increased HDL-C by 8.9% and decreased triglycerides
1289 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
by 9.6% without a signicant reduction in the primary end
point but with a 16% reduction in the combined secondary
end point of mortality and nonfatal myocardial infarction
and stroke (193). Given the modest 0.6% decrease in
hemoglobin A1c, many have speculated that part of the
clinical benets of these agents could be related to the
HDL-Craising benets of these agents.
Pioglitazone has been recently tested against other oral
hypoglycemic agents, including glimepiride, a sulfonylurea
that acts as an insulin secretagogue. In the CHICAGO
(Carotid Intima Media Thickness in Atherosclerosis Using
Pioglitazone) trial, type 2 diabetic patients treated with
pioglitazone had a decrease in the progression of carotid
intima media thickness compared with type 2 diabetic
patients treated with glimepiride (194). More recently, the
PERISCOPE (Pioglitazone Effect on Regression of Intra-
vascular Sonographic Coronary Obstruction Prospective
Evaluation) study randomly allocated type 2 diabetic pa-
tients with CHD to pioglitazone or glimepiride (195).
Patients treated with pioglitazone had a 4.6 mg/dl increase
in HDL-C levels and a decrease in CHD progression as
evaluated by intravascular ultrasonography (195).
The effects of pioglitazone may not extend to thiazo-
lidinediones, in general. Rosiglitazone does not result in the
same favorable lipid prole (191). Furthermore, a recent
meta-analysis suggested that rosiglitazone treatment results
in a signicant risk of myocardial infarction (196). How-
ever, in the large, randomized RECORD (Rosiglitazone
Evaluated for Cardiac Outcomes and Regulation of Gly-
caemia in Diabetes) trial, an interim analysis subsequently
showed no difference in myocardial infarction or cardiovas-
cular deaths (197). A meta-analysis of pioglitazone, largely
involving the PROACTIVE trial, demonstrated a decrease
in mortality, myocardial infarction, or stroke (198). With
the currently available data, this class is generally not used
for HDL modication.
Glitazars. A new class of agents called glitazars with
combined PPAR- and PPAR- agonism is being evalu-
ated (199201). These drugs would theoretically combine
the lipid benets of PPAR activation with the insulin-
sensitizing effects of PPAR activation to optimally treat
the metabolic syndrome.
Ragaglitazar increased HDL-Cby 31%, decreased triglycerides
by 62%, and decreased hemoglobin A1c by 1.3%, but the
adverse events of edema, anemia, and leukopenia have
drawn concern (202,203). Muraglitazar increases HDL-C
by as much as 16% in type 2 diabetic patients, but, as with
ragaglitazar and the thiazolidinediones, weight gain and
edema were more common with muraglitazar therapy
(204,205). An analysis of muraglitazars phase 2 and 3 data
revealed an increase in risk of death, cardiovascular events,
and congestive heart failure associated with muraglitazar
(206). Tesaglitazar, a third agent in this drug class, can
increase HDL-C by 13% (207209). Recently, a phase 2
trial of aleglitazar was shown to increase HDL-C by 20%
and also decrease hemoglobin A1c in a dose-dependent
manner, with an increase in edema but not congestive heart
failure or myocardial infarction (210). As a result, a phase 3
study of aleglitazar in type 2 diabetic patients with a recent
acute coronary syndrome has been announced (211).
CETP inhibitors. Humans with CETP deciency due to
molecular defects in the CETP gene have markedly elevated
plasma levels of HDL-C and apolipoprotein A-I (212,213).
Studies regarding the association of lower CETP levels and
atherosclerosis are mixed, but patients with CETP de-
ciency and higher HDL-C levels tend to have lower rates of
atherosclerotic disease (214217). There was initial concern
that the larger, cholesterol-rich HDL particles formed
through CETP inhibition would not as efciently allow for
reverse cholesterol transport (218). In response, a study
showed that these larger particles promote cholesterol efux
through the ABCG transporter (219). Such observations
led to the concept that pharmacological CETP inhibition
might favorably increase HDL-C levels (122).
Animal models of CETP inhibitors using a variety of
different pharmacological strategies have resulted in an increase
in HDL-C and an attenuation of atherosclerosis (220226).
Early small studies of torcetrapib, a direct CETP inhibitor, in
healthy subjects and patients with HDL-C 40 mg/dl dem-
onstrated signicant increases in HDL-C levels of 50% to 60%
(218,227). Torcetrapib was evaluated on a much larger scale
through the ILLUMINATE (Investigation of Lipid Level
Management to Understand its Impact in Atherosclerotic
Events) trial (228). In that trial, 15,067 patients with a history
of cardiovascular disease or type 2 diabetes mellitus receiving
atorvastatin were randomly assigned to torcetrapib or placebo.
Patients receiving torcetrapib experienced a 72.1% increase in
HDL-C, a 24.9% decrease in LDL-C, and a 9% decrease in
triglycerides over 1 year. However, the ILLUMINATE trial
was prematurely terminated because of an increase in cardio-
vascular events (hazard ratio: 1.25; 95% condence interval:
1.09 to 1.44) and all-cause mortality (hazard ratio: 1.58; 95%
condence interval: 1.14 to 2.19). Off-target toxic effects
including hyperaldosteronism as reected by the observed
hypokalemia, hypernatremia, and increase in systolic blood
pressure by a median of 5.4 mm Hg specic to the molecule
itself may have been the culprits (228,229). Additionally, there
is speculation that the HDL particles formed by torcetrapib
may not participate in reverse cholesterol transport and other
cardioprotective qualities, but recent data suggest that these
particles do participate in reverse cholesterol transport
(229231).
The ILLUSTRATE (Investigation of Lipid Level Man-
agement Using Coronary Ultrasound to Assess Reduction
of Atherosclerosis by CETP Inhibition and HDL Eleva-
tion) study concurrently did not show an effect of torce-
trapib on coronary atherosclerosis (232). A post-hoc analysis
suggested that patients who experienced the highest level of
HDL-C increase did have some regression of coronary
atherosclerosis (233). Patients with familial hypercholester-
olemia treated with torcetrapib in the RADIANCE1 (Rat-
ing Atherosclerotic Disease Change by Imaging With a
1290 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
New CETP Inhibitor 1) study did not demonstrate a
change in carotid intima media thickness despite a 55.5%
increase in HDL-C (234). There was also progression of
disease in the common carotid segment, raising the concern
of a directly toxic effect of torcetrapib on the endothelium
(229,234). In the RADIANCE2 trial, patients with mixed
dyslipidemia also had a similar increase in HDL-C, without
an effect on carotid intima media thickness (235). Similar to
in the ILLUMINATE trial, torcetrapib was associated with
a small increase in blood pressure in the ILLUSTRATE,
RADIANCE1, and RADIANCE2 studies (232,234,235).
Dalcetrapib (also known as R1658, JTT-705, and
RO460731) is another CETP inhibitor (236). A phase 2
trial involving healthy subjects demonstrated a 34% increase
in HDL-C (237). Another phase 2 trial involving patients
with dyslipidemia on pravastatin therapy showed an associ-
ated 28% increase in HDL-C with dalcetrapib (238).
Neither study had an increase in blood pressure or cardio-
vascular adverse events, as seen in the torcetrapib trials
(237239). In vitro, dalcetrapib does not activate the renin-
angiotensin-aldosterone pathway (240). Currently, the
phase 3 dal-OUTCOMES trial is recruiting 15,600
patients with recent acute coronary syndromes to further
study dalcetrapibs effect on cardiovascular morbidity and
mortality as well as its safety prole (241). Also, the phase
2b dal-VESSEL study will further evaluate dalcetrapibs
favorable effect on endothelial function in hypercholester-
olemic patients with a low baseline HDL-C level (242).
And the phase 2b dal-PLAQUE study plans to characterize
its effects on atherosclerotic plaque with magnetic resonance
imaging and positron emission tomography/computed to-
mography (243).
Anacetrapib (also known as MK-0859) is a novel, potent
CETP inhibitor also being studied. A phase 1 trial involving
healthy subjects documented an increase in HDL-C up to
129% without an effect on blood pressure (244). Recently, a
phase 2 trial showed that 589 patients with dyslipidemia
receiving atorvastatin randomly assigned to anacetrapib had
a 130% increase in their HDL-C levels and a decrease of
40% in their LDL-C levels without an effect on blood
pressure (245). The DEFINE (Efcacy and Tolerability of
CETP Inhibition With Anacetrapib) trial has recruited
1,623 patients with CHD already receiving statin therapy
for an anacetrapib phase 3 safety trial, and a larger outcomes
study is planned (246).
ACAT inhibitors. A target for treating atherosclerosis has
been the inhibition of acyl-coenzyme A cholesterol acyl-
transferase (ACAT), which esteries cholesterol in a range
of tissues (247,248). Theoretically ACAT, specically
ACAT-1, inhibition would slow the progression of foam
macrophage cells in theory (249). Subsequently, free cho-
lesterol would be available for reverse cholesterol transport
(250). ACAT-2, the other identied ACAT isoform, is
present in the liver and intestine, so inhibition of this
enzyme would reduce LDL formation (251).
Avasimibe, a nonselective ACAT inhibitor, was assessed
in 639 randomized patients with CHD in the phase 2
A-PLUS (Avasimibe and Progression of Lesions on Ultra-
Sound) study (252). Unfortunately, avasimibe did not alter
HDL-C levels, and it did not have an impact on coronary
atherosclerosis as assessed by intravascular ultrasonogra-
phy (252). Pactimibe, another nonselective ACAT inhib-
itor, was evaluated by intravascular ultrasonography in
the ACTIVATE (ACAT Intravascular Atherosclerosis
Treatment Evaluation) trial of 408 patients with CHD
(253). Pactimibe resulted in a 5.4% (p 0.04) increase in
HDL-C, but although there was no difference compared
with placebo with respect to atherosclerosis progression,
secondary variables suggested that pactimibe limited athero-
sclerosis regression compared with placebo (253). Further-
more, the more recent CAPTIVATE (Carotid Atheroscle-
rosis Progression Trial Investigating Vascular ACAT
Inhibition Treatment Effects) evaluated the effects of pac-
timibe on carotid atherosclerosis using ultrasonography of
carotid intima media thickness in 892 patients with familial
atherosclerosis (254). There was no change in HDL-C
levels, and although there was no change in maximum
carotid intima media thickness, but there was an increase in
mean carotid intima media thickness by 2.9% (p 0.05)
after 1 year, and, notably, there was a signicant absolute
risk increase of 2.1% of cardiovascular death, myocardial
infarction, and stroke with pactimibe (254). Potential ex-
planations for this proatherogenic effect of ACAT inhibi-
tion is that the accumulation of free cholesterol in macro-
phages results in ABCA1 degradation, limiting reverse
cholesterol transport, and such free cholesterol abundance
causes cellular death, stimulating and intensifying the de-
velopment of atherosclerosis (255,256).
Reconstituted HDL infusion. Short-term infusions of
reconstituted HDL have been a target of reverse cholesterol
transport therapy. The HDL infusions also increase nitric
oxide bioavailability for endothelial protection (257,258). In
a small study of healthy subjects, these intravenous infusions
promoted reverse cholesterol transport (259).
On the basis of these data, the ERASE (Effect of rHDL
on AtherosclerosisSafety and Efcacy) study randomly
assigned 183 patients within 2 weeks of an acute coronary
syndrome to 4 weekly infusions of CSL-111, reconstituted
HDL made of puried human apolipoprotein A-I and
soybean phosphatidylcholine (260). There was a nonsignif-
icant trend toward an improvement in coronary atheroma
volume when measured by intravascular ultrasonography,
but the study was prematurely terminated because of a high
incidence of liver function test abnormalities (260). None-
theless, there is some evidence that reconstituted HDL
infusion promotes a favorable change in the quality of
coronary atheroma (260,261). In type 2 diabetic patients,
recent data suggest that reconstituted HDL infusion not
only suppresses the expression of inammatory markers and
enhances reverse cholesterol efux, but also augments gly-
cemic control by increasing plasma insulin and activating
1291 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
adenosine monophosphate-activated protein kinase in skel-
etal muscle (262,263).
Apolipoprotein A-I Milano infusion. In 1980, a group in
Milano described 40 persons in a small village in northern
Italy called Limone sul Garda who, despite very low
HDL-C levels (10 to 30 mg/dl), had a reduced atheroscle-
rotic disease burden and longer lives (264266). This was
attributed to an arginine to cysteine mutation at position
173 of apolipoprotein A-I, termed apolipoprotein A-I
Milano, allowing for disulde bond dimer formation
(264266). Not all of the cysteine residues dimerize, and
the free thiol groups have antioxidant properties (267).
Recombinant apolipoprotein A-I Milano infusions in ani-
mals demonstrated evidence of atherosclerosis regression,
which can occur in as little as 48 h (268,269).
This therapy was piloted in humans when ETC-216,
recombinant apolipoprotein A-I Milano complexed with
phospholipid, was randomly infused in 57 patients within 2
weeks of an acute coronary syndrome over 5 weekly treat-
ments (270). There was signicant reduction in intravascu-
lar ultrasound-measured coronary atheroma burden with
ETC-216, with 1 patient reported to have a signicant rise
in transaminases (270). In a trial of 47 patients after an acute
coronary syndrome, recombinant apolipoprotein A-I Mi-
lano infusion was associated with reverse coronary remod-
eling whereby atheroma burden was reduced but luminal
size was unchanged (271).
Apolipoprotein A-I mimetics. Apolipoprotein A-I is a
protein with 243 amino acids and consists of a tandem
assortment of amphipathic helices (272). Reverse choles-
terol transport by the ABCA1 pathway is mediated by the
detergent-like properties of these amphipathic helical seg-
ments (273,274).
The development of smaller peptides that retain such
activity began with the development of peptide mimetic
18A, consisting of 18 amino acids, which does not share
sequence homology of apolipoprotein A-I but can form a
similar amphipathic helix (275). Multiple modications
have been studied, but the most studied is 4F, which is a
dimerized version of 18A with 2 phenylalanine substitutions
(273,275278).
An orally active 4F peptide made with D-amino acids,
D-4F, given to mice decreases atherosclerotic lesions and
promotes reverse cholesterol transport (279,280). Phase I data
suggest that D-4F may be safe for humans with CHD, and
although it does not alter lipoprotein levels, it may improve the
HDL anti-inammatory index, but further clinical studies are
needed to elucidate this signicance (281).
Given concerns regarding possible cytotoxicity through
ABCA1-independent lipid efux, additional peptide mi-
metics have been engineered (282). Twenty-two amino acid
peptides based on domains of apolipoprotein A-I that have
a higher afnity for ABCA1 have been shown to promote
cholesterol efux without cytopathic effects (282284). Fur-
thermore, such domains are conserved across other apoli-
poproteins, and similarly designed peptides from apoli-
poprotein E promote ABCA1-mediated reverse cholesterol
transport (285). Recently, 5A, an asymmetric bihelical
peptide based on 2F, with 1 of the domains containing more
alanine residues and thereby reducing its helical content, has
been constructed to more closely reect the combination of
low- and high-afnity helices on apolipoprotein A-I (286).
The 5A peptide had increased ABCA1-dependent choles-
terol efux and decreased hemolysis compared with its
parent compound (286).
The strategy of using peptide mimetics to treat athero-
sclerotic disease is in its infancy but carries the potential to
treat disease through specic, endogenous pathways.
Apolipoprotein A-I up-regulators. Recently, a novel par-
ticle called RVX-208 that increases the transcription of
apolipoprotein A-I to promote reverse cholesterol transport
has been announced (287). This transport was facilitated
through the ABCA1, ABCG1, and SR-B1 pathways
through apolipoprotein A-I up-regulation (288). Phase I
data from 24 healthy subjects have shown a dose-dependent
increase in apolipoprotein A-I, and thereby HDL function-
ality (288,289). Further data and investigation are needed to
fully examine the ability of small molecules up-regulating
apolipoprotein A-I to impact the burden of atherosclerosis.
Conclusions
There is overwhelming evidence that high levels of HDL-C
in nature are associated with a lower risk of CHD. Unlike
LDL-C, the mechanisms controlling HDL-C are more
complex. Lifestyle interventions are safe but only modestly
increase HDL-C. The best treatments available currently
are the niacin derivatives, although the newer CETP
inhibitors, apolipoprotein mimetics, and apolipoprotein up-
regulators hold much promise. The next 5 years should
provide information on whether the ux or cycling of HDL
is more important than HDL-C levels and also whether we
should target raising specic HDL subclasses rather than
HDL-C itself.
Acknowledgment
The authors would like to thank Philip Barter, MD, PhD,
for his thoughtful and helpful review of this manuscript.
Reprint requests and correspondence: Dr. Christopher P. Can-
non, Brigham and Womens Hospital, Cardiovascular Division,
TIMI Study, 350 Longwood Avenue, First Floor Ofce, Boston,
Massachusetts 02115. E-mail: cpcannon@partners.org.
REFERENCES
1. Downs JR, Cleareld M, Weis S, et al. Primary prevention of acute
coronary events with lovastatin in men and women with average
cholesterol levels: results of AFCAPS/TexCAPS. JAMA 1998;279:
161522.
2. Cannon CP, Braunwald E, McCabe CH, et al. Intensive versus
moderate lipid lowering with statins after acute coronary syndromes.
N Engl J Med 2004;350:1495504.
1292 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
3. LaRosa JC, Grundy SM, Waters DD, et al. Intensive lipid lowering
with atorvastatin in patients with stable coronary disease. N Engl
J Med 2005;352:142535.
4. Grundy SM, Cleeman JI, Merz CN, et al. Implications of recent
clinical trials for the National Cholesterol Education Program Adult
Treatment Panel III guidelines. Circulation 2004;110:22739.
5. Castelli WP, Garrison RJ, Wilson PWF, et al. Incidence of coronary
heart disease and lipoprotein cholesterol levels: the Framingham
Study. JAMA 1986;256:28358.
6. Assmann G, Schulte H, von Eckardstein A, Huang Y. High-density
lipoprotein cholesterol as a predictor of coronary heart disease risk:
the PROCAM experience and pathophysiological implications for
reverse cholesterol transport. Atherosclerosis 1996;124 Suppl:1120.
7. Curb JD, Abbott RD, Rodriguez BL, et al. A prospective study of
HDL-C and cholesteryl ester transfer protein gene mutations and the
risk of coronary heart disease in the elderly. J Lipid Res 2004;45:
94853.
8. Sharrett AR, Ballantyne CM, Coady SA, et al. Coronary heart
disease prediction from lipoprotein cholesterol levels, triglycerides,
lipoprotein(a)l, apolipoproteins A-I and B, and HDL density sub-
fractions: the Atherosclerosis Risk in Communities (ARIC) study.
Circulation 2001;104:110813.
9. Turner RC, Millins H, Neil HA, et al. Risk factors for coronary
artery disease in non-insulin dependent diabetes mellitus: United
Kingdom Prospective Diabetes Study (UKPDS:23). BMJ 1998;316:
8238.
10. Gordon DJ, Probsteld JL, Garrison RJ, et al. High-density lipopro-
tein cholesterol and cardiovascular disease: four prospective American
studies. Circulation 1989;79:815.
11. Barter P, Gotto AM, LaRosa JC, et al. HDL cholesterol, very low
levels of LDL cholesterol, and cardiovascular events. N Engl J Med
2007;357:130110.
12. Baigent C, Keech A, Kearney PM, et al. Efcacy and safety of
cholesterol-lowering treatment: prospective meta-analysis of data
from 90,056 participants in 14 randomised trials of statins. Lancet
2005;366:126778.
13. Barter PJ, Cauleld M, Eriksson M, et al. Effects of torcetrapib in
patients at high risk for coronary events. N Engl J Med 2007;357:
210922.
14. Expert Panel on Detection, Evaluation, and Treatment of High
Blood Cholesterol in Adults. Executive summary of the third report
of the National Cholesterol Education Program (NCEP) Expert
Panel on Detection, Evaluation, and Treatment of High Blood
Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001;285:
248697.
15. Passos VM, Barreto SM, Diniz LM, Lima-Costa MF. Type 2 diabetes:
prevalence and associated factors in a Brazilian communitythe Bambui
health and aging study. Sao Paulo Med J 2005;123:6671.
16. Aguilar-Salinas CA, Olaiz G, Valles V, et al. High prevalence of low
HDL cholesterol concentrations and mixed hyperlipidemia in a
Mexican nationwide survey. J Lipid Res 2001;42:1298307.
17. Patel JV, Kirby M, Hughes EA. The lipid audit: analysis of lipid
management in two centres in Britain 2003. Br J Cardiol 2004;11:
2147.
18. Bruckert E. Epidemiology of low HDL-cholesterol: results of studies
and surveys. Eur Heart J 2006;8 Suppl:F1722.
19. Li Z, Yang R, Xu G, Xia T. Serum lipid concentrations and
prevalence of dyslipidemia in a large professional population in
Beijing. Clin Chem 2005;51:14450.
20. Karthikeyan G, Teo KK, Islam S, et al. Lipid prole, plasma
apolipoproteins, and risk of a rst myocardial infarction among
Asians: an analysis from the INTERHEART study. J Am Coll
Cardiol 2009;53:24453.
21. Kontush A, Chapman MJ. Antiatherogenic small, dense HDL
guardian angel of the arterial wall? Nat Clin Pract Cardiovasc Med
2006;3:14453.
22. Barter PJ. The regulation and remodelling of HDL by plasma factors.
Atherosclerosis 2002;3 Suppl:3947.
23. Kontush A, Gurin M, Chapman MJ. Spotlight on HDL-raising
therapies: insights from the torcetrapib trials. Nat Clin Pract Car-
diovasc Med 2008;5:32936.
24. von Eckardstein A, Nofer JR, Assmann G. High density lipoproteins
and arteriosclerosis: role of cholesterol efux and reverse cholesterol
transport. Arterioscler Thromb Vasc Biol 2001;21:1327.
25. Lewis GF, Rader DJ. New insights into the regulation of HDL
metabolism and reverse cholesterol transport. Circ Res 2005;96:
122132.
26. Wang N, Lan D, Chen W, Matsuura F, Tall AR. ATP-binding
cassette transporters G1 and G4 mediate cellular cholesterol efux to
high-density lipoproteins. Proc Natl Acad Sci U S A 2004;101:
97749.
27. Ohta T, Saku K, Takata K, Nakamura R, Ikeda Y, Matsuda I.
Different effects of subclasses of HDL containing apoA-I but not
apoA-II (LpA-I) on cholesterol esterication in plasma and net
cholesterol efux from foam cells. Arterioscler Thromb Vasc Biol
1995;15:95662.
28. Sasahara T, Nestel P, Fidge N, Sviridov D. Cholesterol transport
between cells and high density lipoprotein subfractions from obese
and lean subjects. J Lipid Res 1998;39:54454.
29. Asztalos B, Zhang W, Roheim PS, Wong L. Role of free apoli-
poprotein A-I in cholesterol efux: formation of pre-alpha-migrating
high-density lipoprotein particles. Arterioscler Thromb Vasc Biol
1997;17:16306.
30. Meyers CD, Kashyap ML. Pharmacologic elevation of high-density
lipoproteins: recent insights on mechanism of action and atheroscle-
rosis protection. Curr Opin Cardiol 2004;19:36673.
31. Navab M, Anantharamaiah GM, Reddy ST, et al. The oxidation
hypothesis of atherogenesis: the role of oxidized phospholipids and
HDL. J Lipid Res 2004;45:9931007.
32. Kontush A, Chantepie S, Chapman MJ. Small, dense HDL particles
exert potent protection of atherogenic LDL against oxidative stress.
Arterioscler Thromb Vasc Biol 2003;23:18818.
33. Lawrence MB, Springer TA. Leukocytes roll on a selectin at
physiologic ow rates: distinction from and prerequisite for adhesion
through integrins. Cell 1991;65:85973.
34. Davies MJ, Gordon JL, Gearing AJ, et al. The expression of the
adhesion molecules ICAM-1, VCAM-1, PECAM, and E-selectin
in human atherosclerosis. J Pathol 1993;171:2239.
35. Kume N, Cybulsky MI, Gimbrone MA Jr. Lysophosphatidylcholine,
a component of atherogenic lipoproteins, induces mononuclear leu-
kocyte adhesion molecules in cultured human and rabbit arterial
endothelial cells. J Clin Invest 1992;90:113844.
36. Krejcy K, Schwarzacher S, Ferber W, Plesch C, Cybulsky MI,
Weidinger FF. Expression of VCAM-1 in rabbit iliac arteries is
associated with vasodilator dysfunction of regenerated endothelium
following balloon injury. Atherosclerosis 1996;122:5967.
37. Rohde LE, Lee RT, Rivero J, et al. Circulating cell adhesion
molecules are correlated with ultrasound-based assessment of carotid
atherosclerosis. Arterioscler Thromb Vasc Biol 1998;18:176570.
38. Ridker PM, Hennekens CH, Roitman-Johnson B, Stampfer MJ,
Allen J. Plasma concentration of soluble intercellular adhesion mol-
ecule 1 and risks of future myocardial infarction in apparently healthy
men. Lancet 1998;351:8892.
39. Nicholls SJ, Dusting GJ, Cutri B, et al. Reconstituted HDL inhibits
the acute pro-oxidant and proinammatory vascular changes induced
by a periarterial collar in normocholesterolemic rabbits. Circulation
2005;111:154350.
40. Dimayuga P, Zhu J, Oguchi S, et al. Reconstituted HDL containing
human apolipoprotein A-1 reduces VCAM-1 expression and neoin-
tima formation following periadventitial cuff-induced carotid injury
in apoE null mice. Biochem Biophys Res Commun 1999;264:4658.
41. Ansell BJ, Navab M, Hama S, et al. Inammatory/antiinammatory
properties of high-density lipoprotein distinguish patients from
control subjects better than high-density lipoprotein cholesterol levels
and are favorably affected by simvastatin treatment. Circulation
2003;108:27516.
42. Mineo C, Deguchi H, Grifn JH, Shaul PW. Endothelial and
antithrombotic actions of HDL. Circ Res 2006;98:135264.
43. Yuhanna IS, Zhu Y, Cox BE, et al. High-density lipoprotein binding
to scavenger receptor-BI activates endothelial nitric oxide synthase.
Nat Med 2001;7:8537.
44. Eitzman DT, Westrick RJ, Shen Y, et al. Homozygosity for factor V
Leiden leads to enhanced thrombosis and atherosclerosis in mice.
Circulation 2005;111:18225.
45. Grifn JH, Fernandez JA, Deguchi H. Plasma lipoproteins, hemo-
stasis and thrombosis. Thromb Haemost 2001;86:38694.
46. Grifn JH, Kojima K, Banka CL, Curtiss LK, Fernandez JA.
High-density lipoprotein enhancement of anticoagulant activities of
1293 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
plasma protein S and activated protein C. J Clin Invest 1999;103:
21927.
47. Zheng L, Nukuna B, Brennan ML, et al. Apolipoprotein A-I is a
selective target for myeloperoxidase catalyzed oxidation and func-
tional impairment in subjects with cardiovascular disease. J Clin
Invest 2004;114:52941.
48. Khovidhunkit W, Kim MS, Memon RA, et al. Effects of infection
and inammation on lipid and lipoprotein metabolism: mechanisms
and consequences to the host. J Lipid Res 2004;45:116996.
49. Esteve E, Ricart W, Fernandez-Real JM. Dyslipidemia and inam-
mation: an evolutionary conserved mechanism. Clin Nutr 2005;24:
1631.
50. Kontush A, de Faria EC, Chantepie S, Chapman MJ. A normotri-
glyceridemic, low HDL-cholesterol phenotype is characterised by
elevated oxidative stress and small, dense HDL particles with
attenuated antioxidative activity. Atherosclerosis 2005;182:27785.
51. Nobecourt E, Jacqueminet S, Hansel B, et al. Defective antioxidative
activity of small, dense HDL particles in type 2 diabetes: relationship
to elevated oxidative stress and hyperglycemia. Diabetologia 2005;48:
52938.
52. Hansel B, Giral P, Nobecourt E, et al. Metabolic syndrome is
associated with elevated oxidative stress and dysfunctional dense
high-density lipoprotein particles displaying impaired antioxidative
activity. J Clin Endocrinol Metab 2004;89:496371.
53. Lamarche B, Rashid S, Lewis GF. HDL metabolism in hypertri-
glyceridemic states: an overview. Clin Chim Acta 1999;286:14561.
54. Greene DJ, Skeggs JW, Morton RE. Elevated triglyceride content
diminishes the capacity of high density lipoprotein to deliver cho-
lesteryl esters via the scavenger receptor class B type I (SR-BI). J Biol
Chem 2001;276:480411.
55. Navab M, Reddy ST, Van Lenten BJ, Anantharamaiah GM,
Fogelman AM. The role of dysfunctional HDL in atherosclerosis. J
Lipid Res 2009;50 Suppl:1459.
56. Duncan JJ, Gordon NF, Scott CB. Women walking for health and
tness: how much is enough? JAMA 1991;266:32959.
57. Kraus WE, Houmard JA, Duscha BD, et al. Effects of the amount
and intensity of exercise on plasma lipoproteins. N Engl J Med
2002;347:148392.
58. Roberts CK, Ng C, Hama S, Eliseo AJ, Barnard RJ. Effect of a
short-term diet and exercise intervention on inammatory/anti-
inammatory properties of HDL in overweight/obese men with
cardiovascular risk factors. J Appl Physiol 2006;101:172732.
59. Kushi LH, Lew RA, Stare FJ. Diet and 20-year mortality from
coronary heart disease. The Ireland-Boston Diet-Heart Study.
N Engl J Med 1985;312:8118.
60. West SG. Effect of diet on vascular reactivity: an emerging marker for
vascular risk. Curr Atheroscler Rep 2001;3:44655.
61. Perez-Jimenez F, Lopez-Miranda J, Mata P. Protective effect of
dietary monounsaturated fat on arteriosclerosis: beyond cholesterol.
Atherosclerosis 2002;163:38598.
62. Nicholls SJ, Lundman P, Harmer JA, et al. Consumption of
saturated fat impairs the anti-inammatory properties of high-density
lipoproteins and endothelial function. J Am Coll Cardiol 2006;48:
71520.
63. Cockerill GW, Rye K-A, Gamble JR, Vadas MA, Barter PJ.
High-density lipoproteins inhibit cytokine-induced expression of
endothelial cell adhesion molecules. Arterioscler Thromb Vasc Biol
1995;15:198794.
64. Baker PW, Rye K-A, Gamble JR, Vadas MA, Barter PJ. Ability of
reconstituted high density lipoproteins to inhibit cytokine-induced
expression of vascular cell adhesion molecule-1 in human umbilical
vein endothelial cells. J Lipid Res 1999;40:34553.
65. De Caterina R, Spiecker M, Solaini G, et al. The inhibition of
endothelial activation by unsaturated fatty acids. Lipids 1999;34
Suppl:1914.
66. Mensink RP, Katan MB. Effect of dietary fatty acids on serum lipids
and lipoproteins. A meta-analysis of 27 trials. Arterioscler Thromb
1992;12:9119.
67. Kris-Etherton PM, Pearson TA, Wan Y, et al. High-monounsaturated
fatty acid diets lower both plasma cholesterol and triacylglycerol concen-
trations. Am J Clin Nutr 1999;70:100915.
68. Lichtenstein AH. Dietary fat and cardiovascular disease risk: quantity
or quality? J Womens Health (Larchmt) 2003;12:10914.
69. Kromhout D, Bosschieter EB, de Lezenne Coulander C. The inverse
relation between sh consumption and 20-year mortality from
coronary heart disease. N Engl J Med 1985;312:12059.
70. Nash DT, Nash SD. Grapeseed oil, a natural agent which raises
serum HDL levels. J Am Coll Cardiol 1993;21:31820.
71. Mosdl A, Witte DR, Frost G, Marmot MG, Brunner EJ. Dietary
glycemic index and glycemic load are associated with high-density-
lipoprotein cholesterol at baseline but not with increased risk of
diabetes in the Whitehall II study. Am J Clin Nutr 2007;86:98894.
72. Frost G, Leeds A, Dore D, Madeiros S, Brading S, Dornhorst A.
Glycemic index as a determinant of serum HDL-cholesterol concen-
tration. Lancet 1999;353:10458.
73. Ford E, Liu S. Glycemic index and serum high-density lipoprotein
cholesterol concentration among United States adults. Arch Int Med
2001;161:5726.
74. Liu S, Manson JE, Stampfer MJ, et al. Dietary glycemic load assessed
by food-frequency questionnaire in relation to plasma high-density-
lipoprotein cholesterol and fasting plasma triacylglycerols in post-
menopausal women. Am J Clin Nutr 2001;73:5606.
75. Wood PD, Stefanick ML, Dreon DM, et al. Changes in plasma
lipids and lipoproteins in overweight men during weight loss through
dieting as compared with exercise. N Engl J Med 1988;319:11739.
76. Wood PD, Stefanick ML, Williams PT, Haskell WL. The effects on
plasma lipoproteins of a prudent weight-reducing diet, with or
without exercise, in overweight men and women. N Engl J Med
1991;325:4616.
77. Dattilo AM, Kris-Etherton PM. Effects of weight reduction on
blood lipids and lipoproteins: a meta-analysis. Am J Clin Nutr
1992;56:3208.
78. Schwartz RS, Brunzell JD. Increase of the adipose tissue lipoprotein
lipase activity of weight loss. J Clin Invest 1981;67:142530.
79. Taskinen MR, Nikkila EA. Effects of caloric restriction on lipid
metabolism in man. Atherosclerosis 1979;32:28999.
80. Garrison RJ, Kannel WB, Feinleib M, Castelli WP, McNamara PM,
Padgett SJ. Cigarette smoking and HDL cholesterol: the Framing-
ham Offspring Study. Atherosclerosis 1978;30:1725.
81. Craig WY, Palomaki GE, Haddow JE. Cigarette smoking and serum
lipid and lipoprotein concentrations: an analysis of published data.
BMJ 1989;298:7848.
82. Criqui MH, Wallace RB, Heiss G, Mishkel M, Schonfeld G, Jones
GT. Cigarette smoking and plasma high-density lipoprotein choles-
terol. The Lipid Research Clinics Program Prevalence Study. Circu-
lation 1980;62:IV706.
83. Brischetto CS, Connor WE, Connor SL, Matarazzo JD. Plasma
lipid and lipoprotein proles of cigarette smokers from randomly
selected families: enhancement of hyperlipidemia and depression of
high-density lipoprotein. Am J Cardiol 1983;52:67580.
84. Sirisali K, Kanluan T, Poungvarin N, Prabhant C. Serum lipid,
lipoprotein-cholesterol and apolipoproteins A-I and B of smoking
and non-smoking males. J Med Assoc Thai 1992;75:70913.
85. Dullaart RP, Hoogenberg K, Dikkeschei BD, van Tol A. Higher
plasma lipid transfer protein activities and unfavorable lipoprotein
changes in cigarette-smoking men. Arterioscler Thromb 1994;14:
15815.
86. Mero N, Van Tol A, Scheek LM, et al. Decreased postprandial high
density lipoprotein cholesterol and apolipoproteins A-I and E in
normolipidemic smoking men: relations with lipid transfer proteins
and LCAT activities. J Lipid Res 1998;39:1493502.
87. Maeda K, Noguchi Y, Fukui T. The effects of cessation from
cigarette smoking on the lipid and lipoprotein proles: a meta-
analysis. Prev Med 2003;37:28390.
88. Richard F, Marcaux N, Dallongeville J, et al. Effect of smoking
cessation on lipoprotein A-I and lipoprotein A-I:A-II levels. Metab-
olism 1997;46:7115.
89. Moffatt RJ. Effects of cessation of smoking on serum lipids and high
density lipoprotein-cholesterol. Atherosclerosis 1988;74:859.
90. Klatsky AL, Friedman GD, Siegelaub AB. Alcohol and mortality: a
ten-year Kaiser-Permanente experience. Ann Intern Med 1981;95:
13945.
91. Pell S, DAlonzo CA. A ve-year mortality study of alcoholics. J
Occup Med 1973;15:1205.
92. Rosengren A, Wilhelmsen L, Wedel H. Separate and combined
effects of smoking and alcohol abuse in middle-aged men. Acta Med
Scand 1988;223:1118.
1294 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
93. Thorarinsson AA. Mortality among men alcoholics in Iceland,
195174. J Stud Alcohol 1979;40:70418.
94. Deutscher S, Rockette HE, Krishnaswami V. Evaluation of habitual
excessive alcohol consumption on myocardial infarction risk in
coronary disease patients. Am Heart J 1984;108:98895.
95. Colditz GA, Branch LG, Lipnick RJ, et al. Moderate alcohol and
decreased cardiovascular mortality in an elderly cohort. Am Heart J
1985;109:8869.
96. Friedman LA, Kimball AW. Coronary heart disease mortality and
alcohol consumption in Framingham. Am J Epidemiol 1986;124:
4819.
97. Stampfer MJ, Colditz GA, Willett WC, Speizer FE, Hennekens
CH. A prospective study of moderate alcohol consumption and the
risk of coronary disease and stroke in women. N Engl J Med
1988;319:26773.
98. Rimm EB, Giovannucci EL, Willett WC, et al. Prospective study of
alcohol consumption and risk of coronary disease in men. Lancet
1991;338:4648.
99. Mukamal KJ, Jensen MK, Grnbaek M, et al. Drinking frequency,
mediating biomarkers, and risk of myocardial infarction in women
and men. Circulation 2005;112:140613.
100. Hartung GH, Foreyt JP, Mitchell RE, Mitchell JG, Reeves RS, Gotto
AM Jr. Effect of alcohol intake on high-density lipoprotein cholesterol
levels in runners and inactive men. JAMA 1983;249:74750.
101. Langer RD, Criqui MH, Reed DM. Lipoproteins and blood pressure
as biological pathways for effect of moderate alcohol consumption on
coronary heart disease. Circulation 1992;85:9105.
102. Suh I, Shaten BJ, Cutler JA, Kuller LH. Alcohol use and mortality
from coronary heart disease: the role of high-density lipoprotein
cholesterol: the Multiple Risk Factor Intervention Trial Research
Group. Ann Intern Med 1992;116:8817.
103. Gaziano JM, Buring JE, Breslow JL, et al. Moderate alcohol intake,
increased levels of high-density lipoprotein and its subfractions, and
decreased risk of myocardial infarction. N Engl J Med 1993;329:
182934.
104. Schfer C, Parlesak A, Eckoldt J, et al. Beyond HDL-cholesterol
increase: phospholipid enrichment and shift from HDL3 to HDL2
in alcohol consumers. J Lipid Res 2007;48:15508.
105. Rimm EB, Klatsky A, Grobbee D, Stampfer MJ. Review of moderate
alcohol consumption and reduced risk of coronary heart disease: is the
effect due to beer, wine, or spirits. BMJ 1996;312:7316.
106. Grnbaek M, Becker U, Johansen D, et al. Type of alcohol consumed
and mortality from all causes, coronary heart disease, and cancer. Ann
Intern Med 2000;133:4119.
107. Constant J. Alcohol, ischemic heart disease, and the French paradox.
Cor Artery Dis 1997;8:6459.
108. Long-Term Intervention With Pravastatin in Ischaemic Disease
(LIPID) Study Group. Prevention of cardiovascular events and death
with pravastatin in patients with coronary heart disease and a broad
range of initial cholesterol levels. N Engl J Med 1998;339:134957.
109. Shepherd J, Cobbe SM, Ford I, et al. Prevention of coronary heart
disease with pravastatin in men with hypercholesterolemia. N Engl
J Med 1995;333:13017.
110. Sacks FM, Pfeffer MA, Moye LA, et al. The effect of pravastatin on
coronary events after myocardial infarction in patients with average
cholesterol levels. N Engl J Med 1996;335:10019.
111. Downs JR, Cleareld M, Weis S, et al. Primary prevention of acute
coronary events with lovastatin in men and women with average
cholesterol levels: results of AFCAPS/TexCAPS. AirForce/Texas Cor-
onary Atherosclerosis Prevention Study. JAMA 1998;279:161522.
112. Violi F, Micheletta F, Iuliano L. MRC/BHF heart protection study
of cholesterol lowering with simvastatin in 20,536 high-risk individ-
uals: a randomised placebo-controlled trial. Lancet 2002;360:722.
113. Nissen SE, Tuzcu EM, Schoenhagen P, et al. Effect of intensive
compared with moderate lipid-lowering therapy on progression of
coronary atherosclerosis: a randomized controlled trial. JAMA 2004;
291:107180.
114. Jensen LO, Thayssen P, Pedersen KE, Stender S, Haghfelt T.
Regression of coronary atherosclerosis by simvastatin: a serial intra-
vascular ultrasound study. Circulation 2004;110:26570.
115. Okazaki S, Yokoyama T, Miyauchi K, et al. Early statin treatment in
patients with acute coronary syndrome: demonstration of the bene-
cial effect on atherosclerotic lesions by serial volumetric intravascular
ultrasound analysis during half a year after coronary event: the
ESTABLISH study. Circulation 2004;110:10618.
116. Nicholls SJ, Tuzcu EM, Sipahi I, et al. Statins, high-density
lipoprotein cholesterol, and regression of coronary atherosclerosis.
JAMA 2007;297:499508.
117. Asztalos BF, Roheim PS, Milani RL, et al. Distribution of ApoA-
I-containing HDL subpopulations in patients with coronary heart
disease. Arterioscler Thromb Vasc Biol 2000;20:26706.
118. Asztalos BF. HDL Atherosclerosis Treatment Study. High-density
lipoprotein metabolism and progression of atherosclerosis: new in-
sights from the HDL Atherosclerosis Treatment Study. Curr Opin
Cardiol 2004;19:38591.
119. Asztalos BF, Collins D, Cupples LA, et al. Value of high-density
lipoprotein (HDL) subpopulations in predicting recurrent cardiovas-
cular events in the Veterans Affairs HDL Intervention Trial. Arte-
rioscler Thromb Vasc Biol 2005;25:218591.
120. Asztalos BF, Le Maulf F, Dallal GE, et al. Comparison of the effects
of high doses of rosuvastatin versus atorvastatin on the subpopula-
tions of high density lipoproteins. Am J Cardiol 2007;99:6815.
121. Guerin M, Lassel TS, Le Goff W, Farnier M, Chapman MJ. Action
of atorvastatin in combined hyperlipidemia: preferential reduction of
cholesteryl ester transfer from HDL to VLDL1 particles. Arterioscler
Thromb Vasc Biol 2000;20:18997.
122. Chapman MJ, Le Goff W, Guerin M, Kontush A. Cholesteryl ester
transfer protein: at the heart of the action of lipid-modulating therapy
with statins, brates, niacin, and cholesteryl ester transfer protein
inhibitors. Eur Heart J 2010;31:14964.
123. Altschul R, Hoffer A, Stephen JD. Inuence of nicotinic acid on
serum cholesterol in man. Arch Biochem 1955;54:5589.
124. Parsons WB Jr., Flinn JH. Reduction of serum cholesterol levels and
betalipoprotein cholesterol levels by nicotinic acid. Arch Intern Med
1959;103:78390.
125. Carlson LA, Hamsten A, Asplund A. Pronounced lowering of serum
levels of lipoprotein Lp(a) in hyperlipidaemic subjects treated with
nicotinic acid. J Intern Med 1989;226:2716.
126. Capuzzi DM, Guyton JR, Morgan JM, et al. Efcacy and safety of an
extended-release niacin (Niaspan): a long-term study. Am J Cardiol
1998;82:U7481.
127. Guyton JR, Goldberg AC, Kreisberg RA, Sprecher DL, Superko
HR, OConnor CM. Effectiveness of once-nightly dosing of
extended-release niacin alone and in combination for hypercholes-
terolemia. Am J Cardiol 1998;82:73743.
128. McKenney JM, McCormick LS, Weiss S, Koren M, Kafonek S,
Black DM. A randomized trial of the effects of atorvastatin and
niacin in patients with combined hyperlipidemia or isolated hyper-
triglyceridemia. Collaborative Atorvastatin Study Group. Am J Med
1998;104:13743.
129. Knopp RH. Drug treatment of lipid disorders. N Engl J Med
1999;341:498511.
130. 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;17:20208.
131. Blum CB, Levy RI, Eisenberg S, Hall M III, Goebel RH, Berman
M. High density lipoprotein metabolism in man. J Clin Invest
1977;60:795807.
132. Piepho RW. The pharmacokinetics and pharmacodynamics of agents
proven to raise high-density lipoprotein cholesterol. Am J Cardiol
2000;86:L3540.
133. van der Hoorn JW, de Haan W, Berbe JF, et al. Niacin increases
HDL by reducing hepatic expression and plasma levels of cholesteryl
ester transfer protein in APOE*3Leiden CETP mice. Arterioscler
Thromb Vasc Biol 2008;28:201622.
134. Karpe F, Frayn KN. The nicotinic acid receptora new mechanism
for an old drug. Lancet 2004;363:18924.
135. Rubic T, Trottmann M, Lorenz RL. Stimulation of CD36 and the
key effector of reverse cholesterol transport ATP-binding cassette A1
in monocytoid cells by niacin. Biochem Pharmacol 2004;67:4119.
136. Zambon A, Hokanson JE, Brown BG, Brunzell JD. Evidence for a
new pathophysiological mechanism for coronary artery disease regres-
sion: hepatic lipase-mediated changes in LDL density. Circulation
1999;99:195964.
137. Coronary Drug Project Research Group. Clobrate and niacin in
coronary heart disease. JAMA 1975;231:36081.
1295 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
138. Canner PL, Berge KG, Wenger NK, et al. Fifteen year mortality in
Coronary Drug Project patients: long-term benet with niacin. J Am
Coll Cardiol 1986;8:124555.
139. Brown G, Albers JJ, Fisher LD, et al. Regression of coronary artery
disease as a result of intensive lipid-lowering therapy in men with
high levels of apolipoprotein B. N Engl J Med 1990;323:128998.
140. Brown BG, Zambon A, Poulin D, et al. Use of niacin, statins, and
resins in patients with combined hyperlipidemia. Am J Cardiol
1998;81:52B9B.
141. Blankenhorn DH, Nessim SA, Johnson RL, Sanmarco ME, Azen
SP, Cashin-Hemphill L. Benecial effects of combined colestipol-
niacin therapy on coronary atherosclerosis and coronary venous
bypass grafts. JAMA 1987;257:323340.
142. Cashin-Hemphill L, Mack WJ, Pogoda JM, Sanmarco ME, Azen
SP, Blankenhorn DH. Benecial effects of colestipol-niacin on
coronary atherosclerosis: a 4-year follow-up. JAMA 1990;264:
30137.
143. Carlson LA, Rosenhamer G. Reduction of mortality in the Stock-
holm Ischaemic Heart Disease Secondary Prevention Study by
combined treatment with clobrate and nicotinic acid. Acta Med
Scand 1988;223:40518.
144. Whitney EJ, Krasuski RA, Personius BE, et al. A randomized trial of
a strategy for increasing high-density lipoprotein cholesterol levels:
effects on progression of coronary heart disease and clinical events.
Ann Intern Med 2005;142:95104.
145. Benyo Z, Gille A, Kero J, et al. GPR109A (PUMA-G/HM74A)
mediates nicotinic acid-induced ushing. J Clin Invest 2005;115:
363440.
146. Taylor AJ, Stanek EJ. Flushing and the HDL-C response to
extended-release niacin. J Clin Lipidol 2008;2:2858.
147. Knopp RH, Alagona P, Davidson M, et al. Equivalent efcacy of a
time-release form of niacin (Niaspan) given once-a-night versus plain
niacin in the management of hyperlipidemia. Metabolism 1998;47:
1097104.
148. Grundy SM, Vega GL, McGovern ME, et al. Efcacy, safety, and
tolerability of once-daily niacin for the treatment of dyslipidemia
associated with type 2 diabetes: results of the assessment of diabetes
control and evaluation of the efcacy of Niaspan trial. Arch Intern
Med 2002;162:156876.
149. Goldberg RB, Jacobson TA. Effects of niacin on glucose control in
patients with dyslipidemia. Mayo Clin Proc 2008;83:4708.
150. Buse JB, Ginsberg HN, Bakris GL, et al. Primary prevention of
cardiovascular diseases in people with diabetes mellitus: a scientic
statement from the American Heart Association and the American
Diabetes Association. Circulation 2007;115:11426.
151. American Diabetes Association. Standards of medical care in diabetes
2007. Diabetes Care 2007;30 Suppl 1:441.
152. Guyton JR, Bays HE. Safety considerations with niacin therapy.
Am J Cardiol 2007;99:C2231.
153. Brown BG, Zhao XQ, Chait A, et al. Simvastatin and niacin,
antioxidant vitamins, or the combination for the prevention of
coronary disease. N Engl J Med 2001;345:158392.
154. Taylor AJ, Sullenberger LE, Lee HJ, et al. 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;110:35127.
155. Taylor AJ, Villines TC, Stanek EJ, et al. Extended-release niacin or
ezetimibe and carotid intima-media thickness. N Engl J Med
2009;361:211322.
156. Brown BG, Boden WE. Niacin Plus Statin to Reduce Vascular Events.
Available at: http://clinicaltrials.gov/ct/show/NCT00120289. Accessed
March 7, 2009.
157. Armitage J, Baigent C, Chen Z, Landray M. A Randomized Trial of
the Long-Term Clinical Effects of Raising HDL Cholesterol With
Extended Release Niacin/Laropiprant. Available at: http://
clinicaltrials.gov/ct2/show/NCT00461630. Accessed March 7, 2009.
158. Saku K, Gartside PS, Hynd BA, et al. Mechanism of action of
gembrozil on lipoprotein metabolism. J Clin Invest 1985;75:170212.
159. Frick MH, Elo O, Haapa K, et al. Helsinki Heart Study: primary-
prevention trial with gembrozil in middle-aged men with dyslipi-
demia. Safety of treatment, changes in risk factors, and incidence of
coronary heart disease. N Engl J Med 1987;317:123745.
160. Rubins HB, Robins SJ, Collins D, et al. Gembrozil for the
secondary prevention of coronary heart disease in men with low levels
of high-density lipoprotein cholesterol. N Engl J Med 1999;341:
4108.
161. Birjmohun RS, Hutten BA, Kastelein JJ, Stroes ES. Efcacy and
safety of high-density lipoprotein cholesterol-increasing compounds:
a meta-analysis of randomized controlled trials. J Am Coll Cardiol
2005;45:18597.
162. Hossain MA, Tsujita M, Gonzalez FJ, Yokoyama S. Effects of brate
drugs on expression of ABCA1 and HDL biogenesis in hepatocytes.
J Cardiovasc Pharmacol 2008;51:25866.
163. Arakawa R, Tamehiro N, Nishimaki-Mogami T, Ueda K, Yokoyama
S. Fenobric acid, an active form of fenobrate, increases apolipopro-
tein A-I-mediated high-density lipoprotein biogenesis by enhancing
transcription of ATP-binding cassette transporter A1 gene in a liver
X receptor-dependent manner. Arterioscler Thromb Vasc Biol 2005;
25:11937.
164. Vu-Dac N, Schoonjans K, Laine B, Fruchart J, Auwerx J, Staels B.
Negative regulation of the human apolipoprotein A-I promoter by
brates can be attenuated by the interaction of the peroxisome
proliferator-activated receptor with its response element. J Biol Chem
1994;269:310128.
165. Berthou L, Duverger N, Emmanuel F, et al. Opposite regulation of
human versus mouse apolipoprotein A-I by brates in human apo
A-I transgenic mice. J Clin Invest 1996;97:240816.
166. Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leitersdorf E,
Fruchart JC. Mechanism of action of brates on lipid and lipoprotein
metabolism. Circulation 1998;98:208893.
167. Guerin M, Bruckert E, Dolphin PJ, Turpin G, Chapman MJ.
Fenobrate reduces plasma cholesteryl ester transfer from HDL to
VLDL and normalizes the atherogenic, dense LDL prole in
combined hyperlipidemia. Arterioscler Thromb Vasc Biol 1996;16:
76372.
168. Manninen V, Elo MO, Frick MH, et al. Lipid alterations and decline
in the incidence of coronary heart disease in the Helsinki Heart
Study. JAMA 1988;260:64151.
169. Robins SJ, Collins D, Wittes JT, et al. Relation of gembrozil
treatment and lipid levels with major coronary events. VA-HIT: a
randomized controlled trial. JAMA 2001;285:158591.
170. Nyman JA, Martinson MS, Nelson D, et al. Cost-effectiveness of
gembrozil for coronary heart disease patients with low levels of
high-density lipoprotein cholesterol: the Department of Veterans
Affairs High-Density Lipoprotein Cholesterol Intervention Trial.
Arch Intern Med 2002;162:17782.
171. The Bezabrate Infarction Prevention (BIP) Studies. Secondary
prevention by raising HDL cholesterol and reducing triglycerides in
patients with coronary artery disease. Circulation 2000;102:217.
172. Goldenberg I, Boyko V, Tennenbaum A, Tanne D, Behar S, Guetta
V. Long-term benet of high-density lipoprotein cholesterol-raising
therapy with bezabrate: 16-year mortality follow-up of the beza-
brate infarction prevention trial. Arch Intern Med 2009;169:50814.
173. Keech A, Simes RJ, Barter P, et al. Effects of long-term fenobrate
therapy on cardiovascular events in 9795 people with type 2 diabetes
mellitus (the FIELD study): randomised controlled trial. Lancet
2005;366:184961.
174. Committee of Principal Investigators. WHO cooperative trial on
primary prevention of ischaemic heart disease with clobrate to lower
serum cholesterol: nal mortality follow-up. Report of the Commit-
tee of Principal Investigators. Lancet 1984;2:6004.
175. Arthur JB, Ashby DWR, Bremer C, et al. Trial of clobrate in the
treatment of ischaemic heart disease: ve-year study by a group of
physicians of the Newcastle upon Tyne region. BMJ 1971;4:76775.
176. Ischaemic heart disease: a secondary prevention trial using clobrate.
Report by a Research Committee of the Scottish Society of Physi-
cians. BMJ 1971;4:77584.
177. Broeders N, Knoop C, Antoine M, Tielemans C, Abramowicz D.
Fibrate-induced increase in blood urea and creatinine: is gembrozil
the only innocuous agent? Nephrol Dial Transplant 2000;15:19939.
178. Lipscombe J, Lewis GF, Cattran D, Bargman JM. Deterioration in
renal function associated with brate therapy. Clin Nephrol 2001;55:
3944.
179. Miller DB, Spence JD. Clinical pharmacokinetics of bric acid
derivatives (brates). Clin Pharmacokinet 1998;34:15562.
1296 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
180. Hottelart C, El Esper N, Rose F, Achard JM, Fournier A. Feno-
brate increases creatininemia by increasing metabolic production of
creatinine. Nephron 2002;92:53641.
181. Colhoun HM, Betteridge DJ, Durrington PN, et al. Primary pre-
vention of cardiovascular disease with atorvastatin in type 2 diabetes
in the Collaborative Atorvastatin Diabetes Study (CARDS): multi-
centre randomised placebo-controlled trial. Lancet 2004;364:68596.
182. Collins R, Armitage J, Parish S, Sleigh P, Peto R. MRC/BHF heart
protection study of cholesterol-lowering with simvastatin in 5963
people with diabetes: a randomised placebo-controlled trial. Lancet
2003;361:200516.
183. Grundy SM, Vega GL, Yuan Z, et al. Effectiveness and tolerability
of simvastatin plus fenobrate for combined hyperlipidemia (the
SAFARI trial). Am J Cardiol 2005;95:4628.
184. Athyros VG, Papageorgiou AA, Athyrou VV, et al. Atorvastatin and
micronized fenobrate alone and in combination in type 2 diabetes
with combined hyperlipidemia. Diabetes Care 2002;25:1198202.
185. Shek A, Ferrill MJ. Statin-brate combination therapy. Ann Phar-
macother 2001;35:90817.
186. Omar MA, Wilson JP. FDA adverse event reports on statin-
associated rhabdomyolysis. Ann Pharmacother 2002;36:28895.
187. Prueksaritanont T, Tang C, Qiu Y, et al. Effect of brates on
metabolism of statins in humans hepatocytes. Drug Metab Dispos
2002;30:12807.
188. Ginsberg HN, Bonds DE, Lovato LC, et al. Evolution of the lipid
trial protocol of the Action to Control Cardiovascular Risk in
Diabetes (ACCORD) trial. Am J Cardiol 2007;99 Suppl:5667.
189. Parulkar AA, Pendergrass ML, Granda-Ayala R, Lee TR, Fonseca
VA. Nonhypoglycemic effects of thiazolidinediones. Ann Intern Med
2001;134:6171.
190. Auwerx J. PPAR, the ultimate thrifty gene. Diabetologia 1999;42:
103349.
191. Chiquette E, Ramirez G, Defronzo R. A meta-analysis comparing
the effect of thiazolidinediones on cardiovascular risk factors. Arch
Intern Med 2004;164:2097104.
192. Szapary PO, Bloedon LT, Samaha FF, et al. Effects of pioglitazone
on lipoproteins, inammatory markers, and adipokines in nondiabetic
patients with metabolic syndrome. Arterioscler Thromb Vasc Biol
2006;26:1828.
193. Dormandy JA, Charbonnel B, Eckland DJ, et al. Secondary preven-
tion of macrovascular events in patients with type 2 diabetes in the
PROactive Study (Prospective Pioglitazone Clinical Trial in Macro-
vascular Events): a randomised controlled trial. Lancet 2005;366:
127989.
194. Mazzone T, Meyer PM, Feinsein SB, et al. Effect of pioglitazone
compared with glimepiride on carotid intima-media thickness in type
2 diabetes; a randomized trial. JAMA 2006;296:257281.
195. Nissen SE, Nicholls SJ, Wolski K, et al. Comparison of pioglitazone
vs glimepiride on progression of coronary atherosclerosis in patients
with type 2 diabetes: the PERISCOPE randomized controlled trial.
JAMA 2008;299:156173.
196. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocar-
dial infarction and death from cardiovascular causes. N Engl J Med
2007;356:245771.
197. Home PD, Pocock SJ, Beck-Nielsen H, et al. Rosiglitazone evaluated
for cardiovascular outcomesan interim analysis. N Engl J Med
2007;357:2838.
198. Lincoff AM, Wolski K, Nicholls SJ, Nissen SE. Pioglitazone and risk
of cardiovascular events in patients with type 2 diabetes mellitus: a
meta-analysis of randomized trials. JAMA 2007;298:11808.
199. Lohray BB, Lohray VB, Bajji AC, et al. ()3-[4-[2-(Phenoxazin-
10-yl) ethoxyl]-2-ethoxypropanoic acid [()DRF2725]: a dual
PPAR agonist with potent antihyperglycemic and lipid modulating
activity. J Med Chem 2001;44:26758.
200. Sauerberg P, Pettersson I, Jeppesen L, et al. Novel tricyclic-
alkyloxyphenylpropionic acid: dual PPAR/agonists with hypolipi-
demic and antidiabetic activity. J Med Chem 2002;45:789804.
201. Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, et al. PPAR and
PPAR activators direct a distinct tissue-specic transcriptional
response via a PPRE in the lipoprotein lipase gene. EMBO J
1996;15:533648.
202. Saad MF, Greco S, Osei K, et al. Ragaglitazar improves glycemic
control and lipid prole in type 2 diabetic subjects: a 12-week,
double-blind, placebo-controlled dose-ranging study with an open
pioglitazone arm. Diabetes Care 2004;27:13249.
203. Skrumsager BK, Nielsen KK, Muller M, Pabst G, Drake PG,
Edsberg B. Ragaglitazar: the pharmacokinetics, pharmacodynamics,
and tolerability of a novel dual PPAR alpha and gamma agonist in
healthy subjects and patients with type 2 diabetes. J Clin Pharmacol
2003;43:124456.
204. Buse JB, Rubin CJ, Frederich R, et al. Muraglitazar, a dual (alpha/
gamma) PPAR activator: a randomized, double- blind, placebo-
controlled, 24-week monotherapy trial in adult patients with type 2
diabetes. Clin Ther 2005;27:118195.
205. Kendall DM, Rubin CJ, Mohideen P, et al. Improvement of glycemic
control, triglycerides, and HDL cholesterol levels with muraglitazar,
a dual (alpha/gamma) peroxisome proliferator-activated receptor
activator, in patients with type 2 diabetes inadequately controlled
with metformin monotherapy: a double-blind, randomized,
pioglitazone-comparative study. Diabetes Care 2006;29:101623.
206. Nissen SE, Wolski K, Topol EJ. Effect of muraglitazar on death and
major adverse cardiovascular events in patients with type 2 diabetes
mellitus. JAMA 2005;294:25816.
207. Goldstein BJ, Rosenstock J, Anzalone D, Tou C, Ohman KP. Effect
of tesaglitazar, a dual PPAR alpha/gamma agonist, on glucose and
lipid abnormalities in patients with type 2 diabetes: a 12-week
dose-ranging trial. Curr Med Res Opin 2006;22:257590.
208. Wilding JP, Gause-Nilsson I, Persson A. Tesaglitazar, as add-on
therapy to sulphonylurea, dose-dependently improves glucose and
lipid abnormalities in patients with type 2 diabetes. Diab Vasc Dis
Res 2007;4:194203.
209. Ratner RE, Parikh S, Tou C. Efcacy, safety and tolerability of
tesaglitazar when added to the therapeutic regimen of poorly con-
trolled insulin-treated patients with type 2 diabetes. Diab Vasc Dis
Res 2007;4:21421.
210. Henry RR, Lincoff AM, Mudaliar S, Rabbia M, Chognot C, Herz
M. Effect of the dual peroxisome proliferator-activated receptor-
alpha/gamma agonist aleglitazar on risk of cardiovascular disease in
patients with type 2 diabetes (SYNCHRONY): a phase II, random-
ised, dose-ranging study. Lancet 2009;374:12635.
211. Hoffman-La Roche. Roche to Commence Phase III trials With
Innovative Treatment Designed to Lower Cardiovascular Risk in
Diabetes Patients With Recent Heart attack. Media Release. Avail-
able at: http://www.roche.com/med-cor-2009-06-09. Accessed Oc-
tober 24, 2009.
212. Brown ML, Inazu A, Hesler CB, et al. Molecular basis of lipid
transfer protein deciency in a family with increased high-density
lipoproteins. Nature 1989;342:44851.
213. Inazu A, Brown ML, Hesler CB, et al. Increased high-density
lipoprotein levels caused by a common cholesteryl-ester transfer
protein gene mutation. N Engl J Med 1990;323:12348.
214. Barzilai N, Atzmon G, Schechter C, et al. Unique lipoprotein
phenotype and genotype associated with exceptional longevity.
JAMA 2003;290:203040.
215. Boekholdt SM, Sacks FM, Jukema JW, et al. Cholesteryl ester
transfer protein TaqIB variant, high-density lipoprotein cholesterol
levels, cardiovascular risk, and efcacy of pravastatin treatment:
individual patient meta-analysis of 13 677 subjects. Circulation
2005;111:27887.
216. Hirano K, Yamashita S, Kuga Y, et al. Atherosclerotic disease in
marked hyperalphalipoproteinemia: combined reduction of cho-
lesteryl ester transfer protein and hepatic triglyceride lipase. Arterio-
scler Thromb Vasc Biol 1995;15:184956.
217. Zhong S, Sharp DS, Grove JS, et al. Increased coronary heart disease
in Japanese-American men with mutation in the cholesteryl ester
transfer protein gene despite increased HDL levels. J Clin Invest
1996;97:291723.
218. Brousseau ME, Schaefer EJ, Wolfe ML, et al. Effects of an inhibitor
of cholesteryl ester transfer protein on HDL cholesterol. N Engl
J Med 2004;350:150515.
219. Matsuura F, Wang N, Chen W, Jiang XC, Tall AR. HDL from
CETP-decient subjects shows enhanced ability to promote choles-
terol efux from macrophages in an apoE- and ABCG1-dependent
pathway. J Clin Invest 2006;116:143542.
220. Whitlock ME, Swenson TL, Ramakrishnan R, et al. Monoclonal
antibody inhibition of cholesteryl ester transfer protein activity in the
1297 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
rabbit: effects on lipoprotein composition and high density lipoprotein
cholesteryl ester metabolism. J Clin Invest 1989;84:12937.
221. Gaynor BJ, Sand T, Clark RW, Aiello RJ, Bamberger MJ, Moberly
JB. Inhibition of cholesteryl ester transfer protein activity in hamsters
alters HDL lipid composition. Atherosclerosis 1994;110:1019.
222. Sugano M, Makino N. Changes in plasma lipoprotein cholesterol
levels by antisense oligodeoxynucleotides against cholesteryl ester
transfer protein in cholesterol-fed rabbits. J Biol Chem 1996;271:
190803.
223. Sugano M, Makino N, Sawada S, et al. Effect of antisense oligonu-
cleotides against cholesteryl ester transfer protein on the development
of atherosclerosis in cholesterol-fed rabbits. J Biol Chem 1998;273:
50336.
224. Okamoto H, Yonemori F, Wakitani K, Minowa T, Maeda K,
Shinkai H. A cholesteryl ester transfer protein inhibitor attenuates
atherosclerosis in rabbits. Nature 2000;406:2037.
225. Kothari HV, Poirier KJ, Lee WH, Satoh Y. Inhibition of cholesteryl
ester transfer protein by CGS 25159 and changes in lipoproteins in
hamsters. Atherosclerosis 1997;128:5966.
226. Rittershaus CW, Miller DP, Thomas LJ, 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;20:210612.
227. Clark RW, Sutn TA, Ruggeri RB, et al. Raising high-density
lipoprotein in humans through inhibition of cholesteryl ester transfer
protein: an initial multidose study of torcetrapib. Arterioscler
Thromb Vasc Biol 2004;24:4907.
228. Barter PJ, Cauleld M, Eriksson M, et al. Effects of torcetrapib in
patients at high risk for coronary events. N Engl J Med 2007;357:
210922.
229. Tall AR, Yvan-Charvet L, Wang N. The failure of torcetrapib: was
it the molecule or the mechanism? Arterioscler Thromb Vasc Biol
2007;27:25760.
230. Yvan-Charvet L, Matsuura F, Wang N, et al. Inhibition of cho-
lesteryl ester transfer protein by torcetrapib modestly increases mac-
rophage cholesterol efux to HDL. Arterioscler Thromb Vasc Biol
2007;27:11328.
231. Matsuura F, Wang N, Chen W, Jiang XC, Tall AR. HDL from
CETP-decient subjects shows enhanced ability to promote choles-
terol efux from macrophages in an apoE- and ABCG1-dependent
pathway. J Clin Invest 2006;116:143542.
232. Nissen SE, Tardif JC, Nicholls SJ, et al. Effect of torcetrapib on the
progression of coronary atherosclerosis. N Engl J Med 2007;356:
130416.
233. Nicholls SJ, Tuzcu EM, Brennan DM, Tardif JC, Nissen SE. Cholesteryl
ester transfer protein inhibition, high-density lipoprotein raising, and pro-
gression of coronary atherosclerosis: insights from ILLUSTRATE (In-
vestigation of Lipid Level Management Using Coronary Ultrasound to
Assess Reduction of Atherosclerosis by CETP Inhibition and HDL
Elevation). Circulation 2008;118:250614.
234. Kastelein JJ, van Leuven SI, Burgess L, et al. Effect of torcetrapib on
carotid atherosclerosis in familial hypercholesterolemia. N Engl
J Med 2007;356:162030.
235. Bots ML, Visseren FL, Evans GW, et al. Torcetrapib and carotid
intima-media thickness in mixed dyslipidaemia (RADIANCE 2
study): a randomised, double-blind trial. Lancet 2007;370:15360.
236. Okamoto H, Yonemori F, Wakitani K, et al. A cholesteryl ester
transfer protein inhibitor attenuates atherosclerosis in rabbits. Nature
2000;406:2037.
237. De Grooth GJ, Kuivenhoven JA, Stalenhoef AF, et al. Efcacy and
safety of a novel cholesteryl ester transfer protein inhibitor, JTT-705,
in humans: a randomized phase II dose-response study. Circulation
2002;105:215965.
238. Kuivenhoven JA, de Grooth GJ, Kawamura H, et al. Effectiveness of
inhibition of cholesteryl ester transfer protein by JTT-705 in com-
bination with pravastatin in type II dyslipidemia. Am J Cardiol
2005;95:10858.
239. Stein EA, Stroes ESG, Steiner G, et al. Safety and tolerability of
dalcetrapib. Am J Cardiol 2009;104:8291.
240. Capponi AM, Clerc RG, Campos L, et al. No increase in the in vitro
production of aldosterone or the expression of CYP11B2 with the
CETP modulator dalcetrapib (RO4607381/JTT-705), in contrast
with torcetrapib (abstr). Circulation 2008;118 Suppl:452.
241. Hoffman-La Roche. A Study of RO4607381 in Stable Coronary
Heart Disease Patients With Recent Acute Coronary Syndrome.
Available at: http://clinicaltrials.gov/ct2/show/NCT00658515. Ac-
cessed October 24, 2009.
242. Hermann F, Enseleit F, Spieker LE, et al. Cholesteryl ester transfer
protein inhibition and endothelial function in type II hyperlipidemia.
Thromb Res 2009;123:4605.
243. Hoffman-La Roche. A Study of the Effect of RO4607381 on Athero-
sclerotic Plaque in Patients With Coronary Heart Disease. Available
at: http://clinicaltrials.gov/ct2/show/study/NCT00655473?term
RO4607381&rank4. Accessed October 24, 2009.
244. Krishna R, Anderson MS, Bergman AJ, et al. Effect of the cholesteryl
ester transfer protein inhibitor, anacetrapib, on lipoproteins in pa-
tients with dyslipidaemia and on 24-h ambulatory blood pressure in
healthy individuals: two double-blind, randomised placebo-
controlled phase I studies. Lancet 2007;370:190714.
245. Bloomeld D, Carlson GL, Sapre A, et al. Efcacy and safety of the
cholesterol ester transfer protein inhibitor anacetrapib as mono-
therapy and coadministered with atorvastatin in dyslipidemic pa-
tients. Am Heart J 2009;157:35260.e2.
246. Cannon CP, Dansky HM, Davidson M, et al. Design of the
DEFINE trial: Determining the Efcacy and Tolerability of CETP
Inhibition With Anacetrapib. Am Heart J 2009;158:5139.e3.
247. Miyazaki A, Sakai M, Sakamoto Y, et al. Acyl-coenzyme A:choles-
terol acyltransferase inhibitors for controlling hypercholesterolemia
and atherosclerosis. Curr Opin Invest Drugs 2003;4:10959.
248. Heinonen TM. Inhibition of acyl coenzyme A-cholesterol acyltrans-
ferase: a possible treatment of atherosclerosis? Curr Atheroscler Rep
2002;4:6570.
249. Bocan TM, Mueller SB, Uhlendorf PD, et al. Inhibition of acyl-CoA
cholesterol O-acyltransferase reduces the cholesteryl ester enrichment
of atherosclerotic lesions in the Yucatan micropig. Atherosclerosis
1993;99:17586.
250. An S, Jang YS, Park JS, et al. Inhibition of acyl-coenzyme A:cho-
lesterol acyltransferase stimulates cholesterol efux from macrophages
and stimulates farnesoid X receptor in hepatocytes. Exp Mol Med
2008;40:40717.
251. Anderson RA, Joyce C, Davis M. et al. Identication of a form of
acyl-CoA:cholesterol acyltransferase specic to liver and intestine in
nonhuman primates. J Biol Chem 1998;273:2674754.
252. Tardif JC, Grgoire J, LAllier PL, et al. Effects of the acyl coenzyme
A:cholesterol acyltransferase inhibitor avasimibe on human athero-
sclerotic lesions. Circulation 2004;110:33727.
253. Nissen SE, Tuzcu EM, Brewer HB, et al. Effect of ACAT inhibition
on the progression of coronary atherosclerosis. N Engl J Med
2006;354:125363.
254. Meuwese MC, de Groot E, Duivenvoorden R, et al. ACAT
inhibition and progression of carotid atherosclerosis in patients with
familial hypercholesterolemia: the CAPTIVATE randomized trial.
JAMA 2009;301:11319.
255. Feng B, Tabas I. ABCA1-mediated cholesterol efux is defective in
free cholesterol-loaded macrophages. Mechanism involves enhanced
ABCA1 degradation in a process requiring full NPC1 activity. J Biol
Chem 2002;277:4327180.
256. Warner GJ, Stoudt G, Bamberger M, Johnson WJ, Rothblat GH.
Cell toxicity induced by inhibition of acyl coenzyme A:cholesterol
acyltransferase and accumulation of unesteried cholesterol. J Biol
Chem 1995;270:57728.
257. Spieker LE, Sudano I, Hrlimann D, et al. High-density lipoprotein
restores endothelial function in hypercholesterolemic men. Circula-
tion 2002;105:1399402.
258. Bisoendial RJ, Hovingh GK, Levels JH, et al. Restoration of
endothelial function by increasing high-density lipoprotein in sub-
jects with isolated low high-density lipoprotein. Circulation 2003;
107:29448.
259. Nanjee MN, Cooke CJ, Garvin R, et al. Intravenous apoA-I/lecithin
discs increase pre-beta-HDL concentration in tissue uid and stim-
ulate reverse cholesterol transport in humans. J Lipid Res 2001;42:
158693.
260. Tardif JC, Grgoire J, LAllier PL, et al. Effects of reconstituted
high-density lipoprotein infusions on coronary atherosclerosis: a
randomized controlled trial. JAMA 2007;297:167582.
1298 Natarajan et al. JACC Vol. 55, No. 13, 2010
HDL Therapies March 30, 2010:128399
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014
261. Shaw JA, Bobik A, Murphy A, et al. Infusion of reconstituted
high-density lipoprotein leads to acute changes in human atheroscle-
rotic plaque. Circ Res 2008;103:108491.
262. Patel S, Drew BG, Nakhla S, et al. Reconstituted high-density
lipoprotein increases plasma high-density lipoprotein anti-
inammatory properties and cholesterol efux capacity in patients
with type 2 diabetes. J Am Coll Cardiol 2009;53:96271.
263. Drew BG, Duffy SJ, Formosa MF, et al. High-density lipoprotein
modulates glucose metabolism in patients with type 2 diabetes
mellitus. Circulation 2009;119:210311.
264. Franceschini G, Sirtori CR, Capurso A, Weisgraber KH, Mahley
RW. A-I Milano apolipoprotein: decreased high density lipoprotein
cholesterol levels with signicant lipoprotein modications and with-
out clinical atherosclerosis in an Italian family. J Clin Invest 1993;
91:144552.
265. Gualandri V, Franceschini G, Sirtori SR. Identication of the
complete kindred and evidence of a dominant genetic transmission.
Am J Hum Genet 1985;37:108397.
266. Sirtori CR, Calabresi L, Franceschini G, et al. Cardiovascular status
of carriers of the apolipoprotein A-I (Milano) mutant: the Limone
sul Garda Study. Circulation 2001;103:194954.
267. Bielicki JK, Oda MN. Apolipoprotein A-I (Milano) and apolipopro-
tein A-I (Paris) exhibit an antioxidant activity distinct from that of
wild-type apolipoprotein A-I. Biochemistry 2002;41:208996.
268. Chiesa G, Sirtori CR. Recombinant apolipoprotein A-I (Milano): a
novel agent for the induction of regression of atherosclerotic plaques.
Ann Med 2003;35:26773.
269. Shah PK, Yano J, Reyes O, et al. High-dose recombinant apoli-
poprotein A-I Milano mobilizes tissue cholesterol and rapidly reduces
plaque lipid and macrophage content in apolipoprotein e-decient
mice. Circulation 2001;103:304750.
270. 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:
2292300.
271. Nicholls SJ, Tuzcu EM, Sipahi I, et al. Relationship between
atheroma regression and change in lumen size after infusion of
apolipoprotein A-I Milano. J Am Coll Cardiol 2006;47:9927.
272. Phoenix DA, Harris F, Daman OA, Wallace J. The prediction of
amphiphilic alpha-helices. Curr Protein Pept Sci 2002;3:20121.
273. Remaley AT, Thomas F, Stonik JA, et al. Synthetic amphipathic
helical peptides promote lipid efux from cells by an ABCA1-
dependent and an ABCA1-independent pathway. J Lipid Res 2003;
44:82836.
274. Segrest JP, Jones MK, De Loof H, Brouillette CG, Venkatachalapa-
thi YV, Anantharamaiah GM. The amphipathic helix in the ex-
changeable apolipoproteins: a review of secondary structure and
function. J Lipid Res 1992;33:14166.
275. Anantharamaiah GM, Jones JL, Brouillette CG, et al. Studies of
synthetic peptide analogs of amphipathic helix I: structure of peptide/
DMPC complexes. J Biol Chem 1985;260:1024855.
276. Van Lenten BJ, Wagner AC, Jung CL, et al. Anti-inammatory
apoA-I-mimetic peptides bind oxidized lipids with much higher
afnity than human apoA-I. J Lipid Res 2008;49:230211.
277. Datta G, Chaddha M, Hama S, et al. Effects of increasing hydro-
phobicity on the physical-chemical and biological properties of class
A amphipathic helical peptide. J Lipid Res 2001;42:1096104.
278. Navab M, Anantharamaiah GM, Reddy ST, et al. Apolipoprotein A-I
mimetic peptides. Arterioscler Thromb Vasc Biol 2005;25:132531.
279. 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;105:2902.
280. Navab M, Anantharamaiah GM, Reddy ST, et al. Oral D-4F causes
formation of pre-beta high-density lipoprotein and improves high-
density lipoprotein-mediated cholesterol efux and reverse cholesterol
transport from macrophages in apolipoprotein E-null mice. Circula-
tion 2004;109:321520.
281. Bloedon LT, Dunbar R, Duffy D, et al. Safety, pharmacokinetics,
and pharmacodynamics of oral apoA-I mimetic peptide D-4F in
high-risk cardiovascular patients. J Lipid Res 2008;49:134452.
282. Palgunachari MN, Mishra VK, Lund-Katz S, et al. Only the two end
helixes of eight tandem amphipathic helical domains of human apo
A-I have signicant lipid afnity. Implications for HDL assembly.
Arterioscler Thromb Vasc Biol 1996;16:32838.
283. Gillotte KL, Zaiou M, Lund-Katz S, et al. Apolipoprotein-mediated
plasma membrane microsolubilization. Role of lipid afnity and
membrane penetration in the efux of cellular cholesterol and
phospholipid. J Biol Chem 1999;274:20218.
284. Natarajan P, Forte TM, Chu B, et al. Identication of an apolipopro-
tein A-I structural element that mediates cellular cholesterol efux
and stabilizes ATP binding cassette transporter A1. J Biol Chem
2004;279:2404452.
285. Vedhachalam C, Narayanaswami V, Neto N, et al. The C-terminal
lipid-binding domain of apolipoprotein Eis a highly efcient mediator of
ABCA1-dependent cholesterol efux that promotes the assembly of
high-density lipoproteins. Biochemistry 2007;46:258393.
286. Sethi AA, Stonik JA, Thomas F, et al. Asymmetry in the lipid afnity
of bihelical amphipathic peptides. A structural determinant for the
specicity of ABCA1-dependent cholesterol efux by peptides. J Biol
Chem 2008;283:3227382.
287. Krimbou L, Jahagirdar R, Ruel I, et al. Abstract 679: oral adminis-
tration of compound RVX-208 increases serum levels of ApoA-I and
improves high-density lipoprotein-mediated cholesterol efux in
African green monkeys. Abstract presented at: Scientic Sessions
Meeting of the American Heart Association; October 16, 2007;
Orlando, FL.
288. Krimbou L, Jahagirdar R, Bailey D, et al. Abstract 1696: compound
RVX-208 modulates HDL-C and function in non-human primates
and in early (phase I) human trials. Paper presented at: Scientic
Sessions Meeting of the American Heart Association; October 28,
2008; New Orleans, LA.
289. Johansson J, Jahagirdar R, Genest J. Use of RVX-208 to increase
apolipoprotein A-I and HDL in animals and phase I clinical trials.
Paper presented at: Annual Conference of Arteriosclerosis, Throm-
bosis, and Vascular Biology; April 16, 2008; Atlanta, GA.
Key Words: high-density lipoprotein y coronary artery disease y
prognosis y prevention.
1299 JACC Vol. 55, No. 13, 2010 Natarajan et al.
March 30, 2010:128399 HDL Therapies
Downloaded From: http://content.onlinejacc.org/ on 06/01/2014

S-ar putea să vă placă și