Acessibilidade / Reportar erro

High-density lipoproteins: metabolic, clinical, epidemiological and therapeutic intervention aspects. An update for clinicians

REVIEW ARTICLE

High-density lipoproteins: metabolic, clinical, epidemiological and therapeutic intervention aspects. An update for clinicians

Neusa Forti; Jayme Diament

Instituto do Coração do Hospital das Clínicas – FMUSP, São Paulo, SP, Brazil

Mailing Address Mailing Address: Neusa Forti InCor Av. Dr. Enéas Carvalho de Aguiar, 44 05403-904 – São Paulo, SP, Brazil E-mail: nforti@incor.usp.br

Key words: High-density lipoproteins, reserve cholesterol transport, atherosclerosis, prevention.

High blood concentrations of low-density lipoproteins (LDL) and low blood concentrations of high-density lipoproteins (HDL) are primary factors for the development of atherosclerotic disease, the leading cause of death in industrialized countries. A decrease of approximately 30% in the morbidity-mortality rate of the disease is attributed to the reduction of LDL-C (cholesterol linked to LDL) by the statins. To improve this rate, the focus has shifted to possibilities of increasing HDL-C (cholesterol linked to HDL), the cholesterol fraction considered protective and anti-risk factor.

In this article, we address aspects of HDL metabolism and function, conditions that affect its blood levels, its role in atherosclerotic disease, as well as current and developing therapeutic approaches aimed to increasing its levels.

METABOLIC ASPECTS

Identification

Lipoproteins (LP) consist of lipids and proteins called apoproteins (apo). They are classified according to size, density, and lipid and apoprotein composition: chylomicrons (CHY), very low-density lipoprotein (VLDL), intermediate-density lipoproteins (IDL), low-density lipoproteins (LDL), and high-density lipoproteins (HDL).

HDL particles consist of 50% apoproteins (AI in larger quantity, AII, CI, CII, CIII, E, and J), 20% of free cholesterol (FC) and esterified (CE) cholesterol, 15% of phospholipids (PL), and 5% of triglycerides (TG) (Fig. 1). They are identified by lipoprotein separation using ultracentrifugation, electrophoresis, and nuclear magnetic resonance. In ultracentrifugation, HDL particles are separated at densities that range from 1.063 to 1.210, further separating into subfractions: HDL2 at densities 1.063 to 1.125, and HDL3 at densities 1.125 to 1.210. VHDL particles are found between densities 1.121 to 1.125. By electrophoresis, 90% to 95% of the HDL particles, i.e., virtually all those that contain more apoA-I, also known as LpA-I, present alpha mobility, and only 5% to 10% have beta mobility. By non-denaturating polyacrylamide gel electrophoresis, HDL particles, previously separated by ultracentrifugation, are classified into 5 subfractions by decreasing order of size: HDL2b, HDL2a, HDL3a, HDL3b and HDL3c. Nuclear magnetic resonance provides separation into 5 classes of subparticles: H5, H4 and H3, the largest, correspond to HDL2, and H2, H1 correspond to HDL3. In clinical laboratory, HDL particles are indirectly determined by the cholesterol associated with them (HDL-C)1-6.


According to contents of apoA-I and apoA-II, they are called LpA-I (most of them have the same density and HDL2 load) and LpA-I + AII (most of them have the same HDL3 density and load)5.

HDL Formation

HDL particles are formed in plasma and in the extravascular space7. In order to better understand this process, it is first of all necessary to be acquainted with the secretion of the main apolipoprotein components and cholesterol efflux from peripheral cells.

The liver secretes A-I and A-II apolipoproteins, whereas the intestine only secretes A-I. Free cholesterol present in peripheral cells generates oxysterols that are endogenous ligands for nuclear liver receptors (LXR) and retinoid receptors (RXR). These are also affected by the peroxisome proliferator-activated receptors (PPARs) alpha and gamma. The LXR/RXR receptors and the cholesterol-regulating protein (SREBP) control the transcription of the cholesterol-transporter carrier gene (ABCAI) existing on the surface of macrophages in the artery wall. The ABCAI transporter promotes the exit of free cholesterol from the cell, which will be captured by apoA-I by the influence of the plasma phospholipid transfer protein (PLTP). There are also the ABCG1 transporters, expressed in macrophages and in endothelial cells, and the ABCG4 which is expressed in the brain. These transporters also promote cholesterol efflux1,3,5,8-13.

Apolipoprotein A-I (apo A-I) participate in HDL formation through 3 pathways (Fig. 2)3: 1) capturing free cholesterol and phospholipids from peripheral cells, and forming pre-beta HDL. Influenced by lecithin-cholesterol acyltransferase (LCAT), free cholesterol is esterified, forming the round, mature, esterified-cholesterol-rich HDL particles. Still by influence of LCAT, the "mature" HDL particles are converted into HDL3 particles (denser, even richer in esterified cholesterol), and later, also by PLTP action, acquire phospholipids and are converted into HDL2 (less dense). It is worth mentioning that the interconversion between HDL2 and HDL3 takes place in arterial wall and in plasma (Fig. 3): PLTP and LCAT participate in converting HDL3 into HDL2; hepatic lipase (HL) and cholesteryl ester transfer protein (CETP, glycoprotein produced by the liver) participate in converting HDL2 into HDL311,13; 2) discoid particles containing apoA-I, phospholipids and free cholesterol are secreted by liver and intestine and are transformed, by the action of the LCAT enzyme, into mature HDL particles; 3) TG-rich lipoproteins (CHY, IDL, VLDL) undergo lipoprotein lipase (LLP) action, releasing fragments containing phospholipids, free cholesterol, and small apoproteins. These fragments go into the HDL pool.



Reverse Cholesterol Transport

After being formed, HDL particles are captured by liver via the SRB1 receptors (Scavenger receptor B, class 1) and apoE receptors8,14,15 (also present in adrenal glands and ovaries), that selectively remove esterified cholesterol from liver. Hepatic cholesterol is then excreted in bile, mediated by ABCG5/ABCG8 transporters7,11,14.

Additionally, there is exchange of cholesterol linked to HDL with TG of VLDL, IDL, and LDL, and this exchange is mediated by CETP whose expression is regulated by SREBP and LXR/RXR. Therefore, HDL becomes richer in TG, and cholesterol bound to LDL, IDL, and LDL is captured by B/E hepatic receptors1,5.

The reverse cholesterol transport, therefore, consist of two pathways: 1) 50% directly, by the uptake of HDL-free cholesterol involving SRB1; 2) 50% indirectly, involving CETP1.

Catabolism

Hepatic lipase (HL) acts on TG-rich HDL particles hydrolyzing phospholipids and TG, and facilitating interaction with SRB1 receptors. HDLs then return to the interstitium proportionally with more and smaller proteins, reinitiating the cholesterol removal cycle1. Some authors consider that apoA-I is broken down via SRB1, while others believe that apoA-1 and/or TG-rich HDLs are catabolized in the kidneys by endocytosis in proximal tube with participation of cubilin-megalin proteic complex1,5,7,16,17.

HDL Actions

Several different actions are attributed to HDLs, which taken all together, have an anti-atherogenic effect. The primary action is the reverse cholesterol transport, mentioned above. Other actions have been described in vitro and in animals, such as: antioxidant, anti-inflammatory, platelet antiaggregant, anticoagulant, profibrinolytic, and endothelial protection effects2,3,5,8,18.

HDL particles contain the paraoxonase enzyme that is capable of hydrolyzing lipid peroxides by catalyzing the reaction of oxidized phospholipids into LDL5,19. They eliminate the products of LDL oxidation (lipoperoxides and lysophosphatidylcholine)3,20. Anticoagulant and profibrinolytic effects are based on inhibition of X-factor and platelet activation, tPA secretion (tissue plasminogen activator), and PAI (plasminogen activator inhibitor)2,3,5,21. They stimulate C-reactive protein activity2.

Endothelial protection takes place by: 1) inhibition of oxidized LDL infiltration and consequent protection against acetylcholine-mediated vasodilation and antagonism of vasodilation inhibition caused by lysophosphatidylcholine20; 2) inhibition of adhesion of monocytes to the endothelium by inhibiting adhesion molecules to be expressed on the endothelial surface: VCAM1 (vascular cell adhesion molecule), ICAM1 (intracellular adhesion molecule), and E-selectin. Interleukins 1 (IL1) and 8 (IL8) are also reduced. The HDL2 particles, in particular, can completely eliminate the adhesion of monocytes to endothelial cells22; 3) smaller production of endothelin-1 by the endothelial cells23; 4) stimulation of prostacyclin synthesis and longer half-life, improving muscle relaxation24; 5) modulation of C-type natriuretic peptide production, which causes vasodilation25; 6) activation of eNOS (endothelial nitric oxide synthesis), releasing NO26,27; 7) stimulation of smooth muscle cell production, which may facilitate the recovery of cells lost after the rupture of the atherosclerotic lesion fibrous cap2,28; 8) prevention of cell injury and necrosis resulting from activation of the complement system29.

HDL Levels

Based on epidemiologic studies, HDL-C levels < 35 mg/dl are considered low for men, and < 45 mg/dl low for women30,31. Updated guidelines established that values < 40 mg/dl are undesirable and increase risk for coronary atherosclerotic disease (CAD), whereas values > 60 mg/dl are "protective"32-34. To characterize the metabolic syndrome, along with other abnormalities, HDL-C values < 40 mg/dl are established for men, < 50 mg/dl for women32,33. In recent publication, values < 50 mg/dl were established as risk for women.

Low or high HDL values result from rare genetic causes or are associated with other risk factors and/or medications.

Low HDL Levels

Genetic causes for low levels of HDL include: 1) total deficiency or mutations of apolipoprotein A-I. ApoA-I Milano and apoA-I Paris are mutations in which cysteine is replaced by arginine at locus 173 and 151, respectively. Despite low levels of HDL-c, patients with these mutations have no atherosclerotic involvement. In experiments, these variants were found to be more effective than the original apoA-I in preventing lipid oxidation, and they are also capable of mediating cholesterol efflux from macrophage cells by interacting with ABCAI36-38; 2) complete or partial LCAT deficiency. Fish-eye disease is one form of latter in which patients exhibit low HDL and apoA-I levels, accelerated apoA-I and apo-AII catabolism, remarkable lipid corneal arc, but rarely develop premature atherosclerosis39; 3) genetic disorders related to the ABCAI carrier, such as Tangier disease and familial hypoalphalipoproteinemia. In Tangier disease, HDL levels are extremely low, as are those of apoA-I and apoA-II; lipid deposits are formed in lymphoid organs, such as the tonsils, that become orange-colored, hepatosplenomegaly, and peripheral neuropathy40; 4) poorly explained disorders, such as cases of familial hypoalphalipoproteinemia, familial combined hyperlipidemia, and metabolic syndrome41.

Low HDL levels are clinical findings commonly associated with smoking (due to LCAT deficiency), visceral obesity (due to decreased LCAT and LLP), very low-fat diets, hypertriglyceridemia, and use of certain drugs (such as beta-adrenergic blocking agents, steroids, and androgenic progestogens)42-45.

Elevated HDL Levels

High HDL levels are rarely associated to genetic disorders, such as CETP or HL deficiency. In CEPT deficiency, HDL is very rich in esterified cholesterol, although there is no evidence that it has a protective effect against atherosclerosis13.

They are generally associated with the regular practice of aerobic exercises (by increasing LCAT and LLP, and reducing HL), diets rich in saturated fat and cholesterol (by delaying apoA-I clearance), regular alcohol consumption (by inhibiting HL and increasing apoA-I and apoA-II synthesis), and use of some medications such as estrogens (by increasing apoA-I production and inhibiting HL) and phenytoin42,46-50.

HDL Role in the Atherosclerotic Disease

Experimental, epidemiologic, clinical, and therapeutic intervention investigations have shown that there is an inverse relationship between HDL-C and atherosclerotic disease. To date, there are no reports of anatomical and pathological studies conducted on human subjects or animals showing the direct association of HDL and lesion progression. HDL atherogenicity may be influenced by non-measurable variables, including genetic and acquired factors, or by subclass concentration, metabolic kinetics, and not by absolute quantities of HDL5,51,52. Studies conducted with different populations showed that risk for CAD, if HDL-C is low, is accompanied by other lipid alterations, such as hypertriglyceridemia (specially if the TC/HDL-C is high), an increased concentration of remnant lipoproteins, and small and dense LDL particles. These alterations may be associated with the metabolic syndrome5,53-57.

Experimental Studies

ApoA-I administration to transgenic mice delays formation of atherosclerotic lesions7,58. In rabbits, injection of HDL-VHDL promoted the regression of fatty streak and lipid deposits caused by diets very rich in cholesterol. Injection of apoA-I in hyperlipidemic rabbits lead to reduction in intima thickness and in contents of macrophage cells after balloon injury 18,59-61. In rats with apo-E deficiency, administration of apoA-I was associated to increase of HDL-C (2 to 3x) and lower susceptibility (6x) to atherosclerosis, and also reduced neointimal formation after endothelial denudation18,62,63.

Epidemiological studies

Several epidemiological studies have shown that there is an inverse relationship between HDL-C blood levels and the risk for atherosclerotic disease in coronary arteries5,11,18. We call attention for:

1) World Health Organization study64, analysed data from 19 countries and showed an inverse correlation between HDL-C values and mortality due to CAD; 2) Framingham study65, in which an inverse correlation between HDL-C levels and CAD was observed in men and women of different age groups, and also that the risk is greater with HDL-C are < 40 mg/dl, regardless of LDL-C values. HDL-C > 65 mg/dL conferred protection against coronary events, even if LDL-c >160 mg/dl. The relationship was maintained after other risk factors had been controlled, such as diabetes mellitus, hyperglycemia, age, arterial hypertension, smoking, and obesity; 3) Prospective Cardiovascular Munster Study (PROCAM) 54, in which men with HDL-C < 35 mg/dL had a greater risk for CAD relative to those with HDL-C > 35 mg/dL; 4) control groups of the Multiple Risk Factor Intervention Trial (MRFIT)66 and of the Lipids Research Clinics Coronary Primary Prevention Trial (LRC-CPPT)67, in which the incidence of CAD was greater at HDL-C levels < 40 mg/dL; 5) Lipid Research Clinics Prevalence Mortality Follow-up Study68 shows an increase of 1 mg/dL in HDL-C associated to a 3.5% reduction in the risk for CAD, a mortality rate of 3.7% in men and 4.7% in women for HDL-C < 35 mg/dL, and the risk for cardiovascular disease (CVD) 6 times higher if HDL-C < 35 mg/dL.

Combined analysis of the Framingham, Lipid Research Clinics Prevalence Mortality Follow-up Study, and the LRC-CPPT and MRFIT control groups revealed that, after the adjustment for other risk factors, for each 1 mg/dl reduction in HDL-C, the risk for CAD increases 3% in women and 2% in men69.

It is worth mentioning that elevated HDL-C levels do not necessarily confer protection against atherosclerotic disease and, sometimes, are associated to an excessive cardiovascular risk. This situation was observed in PROCAM and Copenhagen City Heart studies in hypertriglyceridemic patients. In PROCAM and in a Belgium population sample, there was excessive mortality in the 5th quintile of HDL-C in comparison with intermediate levels4,54,70-72.

Clinical and therapeutic intervention studies

The inverse relationship between HDL-C blood concentrations and atherosclerotic lesions in coronary arteries has been demonstrated by several clinical and therapeutic intervention studies.

Atherosclerotic impairment and lipid deposits in different organs were observed in individuals with low levels of HDL-C of unknown etiology. In clinical setting, coronary artery injuries are linked to low levels of HDL-C associated to smoking, obesity, hypertriglyceridemia, and metabolic syndrome.

Drexel et al73 observed that the number of coronary atherosclerotic lesions was inversely proportional to HDL2 values, and this fraction is a better "predictor" of lesion extension. In a study conducted in 80 males with coronary disease, aged 27 to 55 years, smokers, sedentary, non-diabetic and non-obese, Giannini et al74 did not observe a significant relationship between mean HDL-C values and the intensity of damage to the coronary arteries, even though lower values had been found when the injury was more pronounced. Shah and Amin75, in a study conducted with patients who had undergone coronary angioplasty, observed that low HDL-C levels can be considered "predictive" of intimal proliferation, which leads to early restenosis.

Recently, Kuvin et al 76 showed that in coronary disease patients, the endothelium-mediated vasodilation was: 1) greater if LDL-C values were < 80 mg/dl, in comparison with HDL-C > 80 mg/dL and < 100 mg/dL, but this difference was significant only when HDL-C were < 40 mg/dL; 2) greater in individuals with LDL-C < 75 mg/dL and HDL-C < 36 mg/dL treated with niacin in comparison with no treated.

Studies of therapeutic intervention in patients with hyperlipidemia or in individuals with low blood concentrations of HDL-C showed, as explained below, that increase in HDL-C values is associated to a decrease in mortality and risks of new coronary events, as well possibility of lesion regression5,18,46,76-78.

Therapeutic measures to increase HDL-C

With regard to measures used to reduce morbidity-mortality due to CAD, over the past few years the focus has been shifted to those capable of increasing blood HDL-C concentrations, by changes in lifestyle or medication.

Lifestyle measures

Lifestyle measures include an adequate diet, physical exercise, smoking cessation, and lowering body weight for individuals who are overweight or obese.

Dietary habits

Specific diets intended to modify HDL-C levels require not only medical orientation, but also and attendance by a nutritionist8,46,79:

1) Saturated fatty acids raise HDL-C levels by delaying apoA-I clearance. Restriction of saturated fatty acids reduces TC and LDL-C, as well as HDL-C, and the latter is effective especially in women80-83; 2) omega 3 polyunsaturated fatty acids inhibit the hepatic secretion of VLDL and, thus, increase HDL-C. Omega 6 fatty acids reduce these levels by reducing apoA-I production and by increasing HDL catabolism; 3) monounsaturated fatty acids induce HDL-C elevation by reducing VLDL secretion; 4) unsaturated fatty acids, in trans- form, enhance apoA-I catabolism and, thus, reduce HDL-C84; 5) carbohydrates reduce HDL-C by increasing its catabolism as well as the secretion of VLDL. Recently, Aude et al85; showed that reduced ingestion of simple carbohydrates and an increased ingestion of complex carbohydrates, associated to an increase of mono-unsaturated fatty acids, result in a less marked reduction of HDL-C in comparison with the Phase 1 diet recommended by NCEP31. Westman et al86, in contrast, showed that increased HDL-C levels, induced by carbohydrate restriction, accompanied body weight loss; 6) alcohol consumption increasing HDL-C by inhibiting hepatic lipase and increasing apoA-I and A-II synthesis. This increase is dose-dependent: in normal individuals, the consumption of half a bottle of wine per day (39 g of alcohol) after six weeks increased HDL-C by 7 mg/dl, whereas one beer per day (13,5 g of alcohol) caused an increase of only 2 mg/dl87,88; 7) coffee reduces HDL-C by reducing LCAT and increasing CETP activities; 8) besides reducing lipid oxidation, cocoa polifenoles raise HDL-C by a mechanism not yet fully explained89.

Adoption of diet intended for lowering body weight is of greater importance. Caloric restriction increases HDL-C values. For each 3 kg of body weight lost, there is a corresponding 1 mg/dL elevation in HDL-C. Increase in LCAT and LLP activities is observed after a sustained six-week weight loss42,90,91. It should be emphasized that weight loss should be the main focus in metabolic syndrome, helping to control glycidic and lipidic disorders and arterial pressure92.

Smoking cessation

Smoking cessation increases LCAT activity and HDL-C concentrations return to previous levels within 30 to 60 days44,93.

Improve physical exercise

Physical exercise increases LCAT and LLP activity and reduces HL42,47 activity, resulting in increased HDL levels: 1) in both normal individuals and those with different types of dyslipidemia (hypercholesterolemia, isolated hypertriglyceridemia or associated with low levels of HDL-C, isolated low HDL-C)94,95; 2) in obese and non-obese individuals96; 3) particularly when associated to diet orientation. Compared to women, men have more significant HDL-C elevations when they adopt an adequate diet combined with physical exercises97; 4) success depends on quantity and not on intensity of the exercise98.

Over the period of one year, the association of physical exercise with fat and cholesterol restriction raised HDL-C values by 2% and reduced those of LDL-C (8%) and TG (21%). In comparison to control group, there was greater regression (29% x 6%) and lower progression (10% x 45%) of coronary lesions99.

Drug Therapy

Hypolipidemic agents (lipid lowering drugs – lld) in clinical practice

Fibrates, nicotinic acid, and statins increase HDL-C 10%, 20%, and 5 to 10% respectively. Fibrates activate PPARs alpha in the liver, increasing apoA-I production; they also reduce the regulation of SRB1, lowering HDL clearance. Nicotinic acid reduces apoA-I catabolism. Statins reduce CETP activity and increase apoA-I synthesis; reduction in CETP mass has been observed with the use of atorvastatin and sinvastatin5,11,77.

In a metanalysis of randomized studies conducted from 1966 to 2004 with fibrates and nicotinic acid, Birjmohum et al100 showed that, despite a more marked elevation of HDL-C with nicotinic acid (16% x 10%), the decline in events was similar (27% x 25%). But this reduction was significantly associated to elevations in HDL-C levels induced by gemfibrozil in primary and secondary prevention studies4,5,18. In the Helsinki Heart Study – HHS101, elevations in HDL-C (10%) and reductions in LDL-C (10%) and TG (43%) were observed. Increases of HDL-C and the HDL-C/TC ratio were related to risk of CAD. In Veterans Affairs High-Density Lipoprotein Intervention Trial - VAHIT102,103 (mean initial value of HDL-C was equal to 32 mg/dL), after one year the 6% increase in HDL-C levels was accompanied by a 22% reduction in the death rates due to CAD or non-fatal MI. For each 5 mg/dL increase in HDL-C, the risk of coronary events fell by 11%. This did not occur in the Bezafibrate Infarction Prevention study104,105 in which HDL-C were high and increased 18%; the conclusion was that an increase in low HDL-C translates into greater protection.

In primary prevention studies (Air Force/Texas Coronary Atherosclerosis Prevention Study – AFCAPS/Tex CAPS) 106 and West of Scotland Prevastatin Study – WOS) 107 or secondary prevention trials (Cholesterol and Recurrent Events Trial – CARE108, Scandinavian Sinvastatin Survival Study – 4S109, The Long–Term Intervention with Provastatin in Ischaemic Disease – LIPID110), statins also reduced cardiovascular events, even more significantly so when HDL-C values were lower. However, this correlation was not significant4. In the AFCAPS study106, reductions were 45%, 44%, and 15% respectively if HDL-C was < 34, 35 to 39, or > 40 mg/dL. The combined analysis of WOS, CARE, and LIPID (Prospective Pravastatin Pooling Project 111) studies showed a similar reduction in cardiac events (27% to 28%) in both placebo and treatment groups, considering the more elevated HDL-C percentile in relation to the lowest18. In the 4S study, a 1% increase in HDL-C was associated with a 0.80% reduction in risk, regardless of the decline in LDL-C levels112. More distinct angiographic and clinical benefits were also observed in the Lipoprotein and Coronary Atherosclerosis Study – LCAS5,18,113. Combination of simvastatin and niacin, administered for 3 years, reduced the risk of events by 90% and each 1% increase in HDL-C corresponded to a 1% reduction in cardiovascular risk (HDL – Atherosclerosis Treatment Study – HATS)5,18,114. In 167 cardiac patients (x = 67 years of age) treated with statins, addition of niacin after 12 months, increased HDL-C (21%), but no modifications were observed in carotid intima-media thickness (increased in the placebo group)115.

Whitney et al115 recently published the results of an angiographic placebo-controlled study designed for individuals with low levels of HDL-C. After 6 months of a Phase 2 diet of the American Heart Association and regular physical exercises, patients (HDL-C = 34 ± 6 mg/dL, LDL-C = 128 ± 27 mg/dL, and TC = 196 ± 31 mg/dL), were randomized to receive placebo combined with gemfibrozil (1200 mg/dL), nicotinic acid (250 to 3000 mg/day from the 3rd month) and cholestyramine (16 g/day from the 6th month). Reductions in TC (20%), LDL-C (26%), and TG (50%), as well as an increase in HDL-C levels (36%) were observed. After 3 years, treated group had a greater reduction in coronary obstructive processes (¯ 0.8% x ­ 1.4%) and in cardiovascular events (12.7% x 26.4%).

CETP inhibitors

Elevation of blood HDL-C levels is more pronounced due to the action of CETP inhibitors, drugs not yet available for clinical use5,11,18. Two of these are JTT-705 and torcetrapib.

Administration of JTT-705 to rabbits doubled HDL-C values, markedly reducing (70%) the size of the atherosclerotic lesion117. Healthy individuals, with HDL-C < 45 mg/dL, had HDL-C levels increased 37% after treated with JTT-705 for 4 weeks118.

In rabbits, torcetrapib significantly reduced atherosclerosis119. In human subjects with HDL-C < 40 mg/dL (x = 33 mg/dL) receiving 120 mg/day, the drug increased HDL-C levels 46% after 4 weeks and 106% after 8 weeks. When associated to 20 mg/day of atorvastatin, after 4 weeks the increase was 61%120.

Drugs in development

Currently, there are studies underway with drugs capable of raising HDL-C by different mechanisms of action: PPARs alpha and gamma agonists, apoA-I synthesis inducers, selective LXR activators or modulators, FXR activators (farnesoid X receptor / biliary bile acid receptor), endothelial and hepatic lipase inhibitors, SRB1 modulators, and –mimetics of sphingolipids11.

Other possibilities

In an effort to increase HLD-C levels, CETP inhibition was induced in rabbits by a "vaccine" (a synthetic peptide homologous to the sequence of peptides in catalytic CETP site), which reduced the aortic lesions by 39.6%121. In human subjects, HDL-C increase was small122.

Infusion of apoA-I increases the ABCAI-mediated cholesterol efflux. In rabbits, repeated infusions of apoA-I inhibited the progression of atherosclerosis and reduced lesion size123. Recently, Nissen et al124 showed that patients in acute phase of myocardial infarction had a reduction in atheroma volume due to endovenous administration of recombinant apoA-I Milano.

Summary

High density lipoproteins (HDL) constitute a class of heterogeneous lipoproteins with a complex, not fully understood metabolism, particularly concerning cholesteryl ester transfer protein (CETP) action and catabolism. HDLs are anti-atherogenic due to reverse cholesterol transport and to antioxidant, anti-inflammatory, anticoagulant, profibrinolytic and endothelial protection properties, (shown in vitro and in animals). Experimental, clinical, epidemiologic, and therapeutic intervention studies have shown that there is an inverse relationship between blood levels of these lipoproteins and the development of atherosclerotic disease in coronary arteries.

Measures to increase plasma HDL-C levels, besides changes in lifestyle habits (an adequate selection of diet components, smoking cessation, regular practice of aerobic exercises) include lipid lowering drugs (LLDs). Drugs capable of inhibiting CETP are currently in advanced stages of development. Research is also being conducted with other drugs that act on different points of lipid metabolism.

References

1. Kwiterovich PO. The metabolic pathways of high-density lipoprotein, low-density lipoprotein and triglycerides: a current review. Am J Cardiol. 2000;86(Suppl):5L-10L.

2. Nofer JR, Kehrel B, Fobker M, et al. HDL and arteriosclerosis: beyond reverse cholesterol transport. Atherosclerosis. 2002;161:1-16.

3. Chapman MJ. Are the effects of statins on HDL-cholesterol clinically relevant? Eur Heart J Suppl. 2004;6(Suppl C):C58-C63.

4. von Eckardstein A, Huang Y, Assmann G. Physiological role and clinical relevance of high-density lipoprotein subclasses. Curr Opin Lipidol. 1994;5:404-16.

5. Assmann G, Gotto AM. HDL-cholesterol and protective factors in atherosclerosis. Circulation. 2004;109:III8-14.

6. Warnick GR, Nauch M, Rifai N. Evolution of methods for measurement of HDL-cholesterol: from ultracentrifugation to homogenous assays. Clin Chem. 2001;47:579-96.

7. Passareli M, Quintão ER. Metabolismo das lipoproteínas de alta densidade. Rev Soc Cardiol Estado de São Paulo. 2000;6:734-43.

8. von Eckardstein A, Asmann G. Prevention of coronary heart disease by raising high-density lipoprotein cholesterol. Curr Opin Lipidol. 2000;11:627-37.

9. Orani JF, Vaughan AM. ABCA1mediated transport of cellular cholesterol and phospholipids to HDL apolipoproteins. Curr Opin Lipidol. 2000;11:253-60.

10. Huuskonen J, Ehnholm C. Phospholipid transfer protein in lipid metabolism. Curr Opin Lipidol. 2000;11:285-90.

11. Linsel-Nitschke, Tall A. HDL as a target treatment of atherosclerotic cardiovascular disease. Nat Rev Drug Discov. 2005;4:193-205.

12. Wang N, Lan D, Chen W, et al. ATP-binding cassette transporters G1 and G4 mediated cellular cholesterol efflux to high-density lipoproteins. Proc Natl Acad Sci USA. 2004;101:9774-9.

13. Forrester JS, Makkar R, Shah PK. Increasing high-density lipoprotein cholesterol in dyslipidemia by cholesteryl ester transfer protein inhibition. An update for clinicians. Circulation. 2005;111:1847-54.

14. Acton S, Rigotti A, Landschulz KT, et al. Identification of scavenger-receptor SRB1 as high-density lipoprotein receptor. Science. 1996;271:518-20.

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

16. Moestrup SK, Kozyraki R. Cubilin, a high-density lipoprotein receptor. Curr Opin Lipidol. 2000;11:133-40.

17. Hammad SM, Barth JL, Knaack C, et al. Megalin acts in concert with cubilin to mediate endocytosis of high-density lipoprotein. J Biol Chem. 2000;275:12003-8.

18. Asheikh AA, Kuvin JT, Karas RH. High-density lipoprotein cholesterol in the cardiovascular equation: Does the "good" still count? Atherosclerosis. 2005;180:217-23.

19. Matsuda Y, Hirata K, Inoue N, et al. High-density lipoprotein reverse inhibitory effect of oxidized low-density lipoprotein on endothelium dependent arterial relaxation. Circ Res. 1993;72:1103-9.

20. Mackness MI, Durrington PN, Mackness B. How high-density lipoprotein protects against the effects of lipid peroxidation. Curr Opin Lipidol. 2000;11:383-8.

21. Rosenson RS, Loewe GD. Effects of lipids and lipoproteins on thrombosis and rheology. Atherosclerosis. 1998;140:271-80.

22. Barter PJ. Inhibition of endothelial cell adhesion molecule expression by high-density lipoprotein. Clin Exp Pharmacol Physiol. 1997;24:286-7.

23. Horio T, Kohno M, Yasunari K, et al. Stimulation of endothelin-1 release by low-density and very-low-density lipoproteins in cultured human endothelial cells. Atherosclerosis. 1993;101:185-90.

24. Fleisher LN, Tall AR, Witte LD, et al. Stimulation of arterial endothelial cell prostacyclin synthesis by high-density lipoproteins. J Biol Chem. 1982;257:6653-5.

25. Sugiyama S, Kugiyama K, Matsumura T, et al. Lipoproteins regulated C-type natriuretic peptide secretion from cultured vascular endothelial cells. Arterioscler Thomb Vasc Biol. 1995;15:1968-74.

26. Ramet ME, Ramet M, Lu Q, et al. High-density lipoprotein increase the abundance of e NOS protein in human vascular endothelial cells by increasing its half-life. J Am Coll Cardiol. 2003;41:2288-97.

27. Kuvin JT, Patel AR, Sidhu M, et al. Relation between high-density lipoprotein cholesterol and peripheral vasomotor function. Am J Cardiol. 2003;92:275-9.

28. Resink TJ, Bochkov VN, Hahn AW, et al. Low- and high-density lipoproteins as mitogenic factors for vascular smooth muscle cells: individual, additive and synergistic effects. J Vasc Res 1995;32:328-38.

29. Packman CH, Rosenfeld SI, Leddy JP. High-density lipoprotein and its apolipoproteins inhibit cytolytic activity of complement. Studies on the nature of inhibitory moiety. Biochim Biophys Acta. 1985;812:107-15.

30. International Task Force for Prevention of Coronary Heart Disease. Coronary heart disease: reducing the risk. The scientific background to primary and secondary prevention of coronary heart disease. A worldwide view. Nutr Metab Cardiovasc Dis. 1998;8:205-71.

31. Expert Panel on Detection, Evaluation and Treatment of High Blood Cholesterol in Adults: The second report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation and Treatment of High-Blood Cholesterol in Adults (Adult Treatment Panel II). Circulation. 1994;89:1329-445.

32. 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;285:2486-97.

33. Grundy SM, Cleeman JI, Merz CN, et al. Implications of recent clinical trials for the National Education Program Adult Treatment Panel III guidelines. Circulation. 2004;110:227-39.

34. III Diretrizes Brasileiras sobre Dislipidemia e Diretriz de Prevenção da Aterosclerose do Departamento de Aterosclerose da Sociedade Brasileira de Cardiologia. Arq Bras Cardiol. 2001;77(Supp III):1-48.

35. Mosca L, Appel LJ, Benjamin EJ, et al. Evidence-based guidelines for cardiovascular prevention in women. Circulation. 2004;109:672-93.

36. Perez-Mendez O, Bruckert E, Franceschini G, et al. Metabolism of apolipoproteins AI e AII in subjects carrying similar apo AI mutations, apo AI Milano and apo AI Paris. Atherosclerosis. 2000;148:317-25.

37. Bielicki JK, Oda M. Apolipoprotein AI (Milano) and apolipoprotein AI (Paris) exhibit an anti-oxidant activity distinct from that of wild-type apolipoprotein AI. Biochemistry. 2002;41:2089-96.

38. Daum U, Langer C, Duverger N, et al. Apolipoprotein AI (R151C) Paris is defective in activation of lecithin-cholesterol acyl-transferase but not in initial lipid binding, formation of reconstituted lipoproteins, or promotion of cholesterol efflux. J Mol Med. 1999;77:614-22.

39. Kuivenhoven JA, Stalenhoef AF, Hill JS, et al. Two novel molecular defects in the LCAT gene are associated with fish eye disease. Arterioscler Thromb Vasc Biol. 1996;16:294-303.

40. Brooks-Wilson A, Marcil M, Clee SM, et al. Mutations in ABC1 in Tangier disease and familial high-density lipoprotein deficiency. Nat Genet. 1999;22:336-45.

41. Austin MA, King MC, Vranizan KM, et al. Atherogenic lipoprotein phenotype: a proposed genetic marker for coronary heart disease. Circulation. 1990;82:495-506.

42. Ashen MD, Blumenthal RS. Low HDL-cholesterol levels. N Engl J Med. 2005;352:1252-60.

43. Imamura H, Teshima K, Miyamoto N, et al. Cigarette smoking, high-density lipoprotein cholesterol subfractions, and lecitin-cholesterol acyltransferase in young women. Metabolism. 2002;51:1313-6.

44. Maeda K, Noguchi Y, Fukui T. The effect of cessation from cigarette smoking on the lipid and lipoprotein profiles: a meta-analysis. Prev Med. 2003;37:283-90.

45. Denke MA, Sempos CT, Grundy SM. Excess body weight: an under recognized contributor to high blood cholesterol levels in white American men. Arch Intern Med. 1993;153:1093-103.

46. Schaefer E. Lipoproteins, nutrition and heart disease. Am J Cardiol. 2002;75:191-212.

47. Després JP, Lamarche B. Low-intensity endurance exercise training, plasma lipoproteins and the risk of coronary heart disease. J Intern Med. 1994;236:7-22.

48. Rimm EB, Williams P, Fosher K, et al. Moderate alcohol intake and lower risk of coronary heart disease: meta-analysis of effects on lipids and haemostatic factors. Br Med J. 1999;319:1523-8.

49. Wash BW, Schiff I, Rosner B, et al. Effects of post menopausal estrogen replacement on lipids concentration and metabolism of plasma lipoproteins. N Engl J Med. 1991;325:1196-200.

50. Goerdt C, Keith M, Rubins HB. Effects of phenytoin on plasma high-density lipoprotein cholesterol in men with low levels of high-density lipoprotein cholesterol. J Clin Pharmacol. 1995;35:767-75.

51. von Eckardstein A, Nofer JR, Assmann G. High-density lipoproteins and atherosclerosis: role of cholesterol efflux and reverse cholesterol transport. Arterioscler Thromb Vasc Biol. 2001;21:13-27.

52. Gotto AM Jr. Low high-density lipoprotein as a risk factor in coronary heart disease: a working group report. Circulation. 2001;103:2213-8.

53. Expert Panel on Detection, Evaluation and Treatment of High Blood Cholesterol in Adults. 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). Final report. Circulation. 2002;106:3143-421.

54. Assmann G, Schulte H. Relation of high-density lipoprotein cholesterol and triglycerides to incidence of atherosclerotic coronary disease (the PROCAM experience). Am J Cardiol. 1992;70:733-7.

55. Despres JP, Lemieux I, Dagenais GR, et al. HDL-cholesterol as a marker of coronary heart disease: the Québec Cardiovascular Study. Atherosclerosis. 2000;153:263-72.

56. Schaefer EJ, Lamon-Fava S, Ordovas JM, et al. Factors associated with low and elevated plasma high-density lipoprotein cholesterol and apolipoprotein AI levels in the Framingham Offspring Study. J Lipid Res. 1994;35:871-82.

57. Austin MA, Rodriguez BL, McKnight B, et al. Low-density lipoprotein size, triglycerides and high-density lipoprotein as a risk factor for coronary heart disease in older Japanese-American men. Am J Cardiol. 2000;86:412-6.

58. Rubin EM, Krauss RM, Spangler EA, et al. Inhibition of early atherosclerosis in transgenic mice by human apolipoprotein AI. Nature. 1991;353:265-7.

59. Badimon JJ, Badimon L, Fuster V. Regression of atherosclerotic lesions by high-density lipoprotein plasma fraction in the cholesterol-fed rabbit. J Clin Invest. 1990;85:1234-41.

60. Miyasaki A, Sakuma S, Morikawa W, et al. Intravenous injection of rabbit apolipoprotein AI inhibits the progression af atherosclerosis in cholesterol-fed rabbits. Arterioscler Thromb Vasc Biol. 1995;15:1882-8.

61. Ameli S, Hultgardh-Nilsson A, Cercek B. Recombinant apolipoprotein AI Milano reduces intimal thickening after balloon injury in hypercholesterolemic rabbits. Circulation. 1994;90:1935-41.

62. Paszty C, Maeda N, Verstuyft J. Apolipoprotein AI transgene corrects apolipoprotein E deficiency-induced atherosclerosis in mice. J Clin Invest. 1994;94:899-903.

63. Plump AS, Scott CJ, Breslow JL. Human apolipoprotein AI gene expression increases high-density lipoprotein E deficient mouse. Proc Natl Acad Sci USA. 1994;91:9607-11.

64. Simons LD. Interelations of lipids-lipoproteins with coronary artery disease mortality in 19 countries. Am J Cardiol. 1986;27:50-106.

65. Castelli WP. Cholesterol and lipids in the risk of coronary heart disease: the Framingham Heart Study. Can J Cardiol. 1988;4(Suppl A):5A-10A.

66. Multiple Risk Factor Intervention Trial Research Group. Multiple Risk Factor Intervention Trial: Risk factor changes and mortality results. JAMA. 1982;248:1465-77.

67. Gordon DJ, Knoke J, Probstfield JL, et al. High-density lipoprotein cholesterol and coronary heart disease in hypercholesterolemic men. The Lipid Research Clinics Coronary Prevention Trial. Circulation. 1986;74:1217-25.

68. Gordon DJ, Ekelund LG, Karon JM, et al. Predictive value of exercise test for mortality in North American men: the Lipid Research Clinics Mortality Follow-up study. Circulation. 1986;74:252-61.

69. Gordon DJ, Probstfield JL, Garrisson RJ, et al. High-density lipoprotein cholesterol and cardiovascular disease. Four prospective American studies. Circulation. 1989;79:8-15.

70. Jeppesen J, Hein HO, Suadicani P, et al. Triglyceride concentration and ischemic heart disease. An eight-year follow-up in the Copenhagen Male Study. Circulation. 1998;97:1029-36.

71. Cullen P, Schulte H, Assmann G. The Munster Heart Study (PROCAM): total mortality in middle aged men is increased at low total and LDL cholesterol concentrations in smokers but not in non smokers. Circulation. 1997;96:2128-36.

72. De Backer G, De Bacquer D, Kornitzer M. Epidemiologic aspects of high-density lipoprotein cholesterol. Atherosclerosis. 1998;137(Suppl):S1-S6.

73. Drexel H, Ammann FW, Rentsch K, et al. Relation of the level of high-density lipoprotein subfraction to the presence and extent of coronay artery disease. Am J Cardiol. 1992;70:436-40.

74. Giannini SD, Forti N, Gois JM, et al. Relações entre níveis de HDL-colesterol, índices de risco coronário e o grau de aterosclerose avaliado por cinecoronariografia. Arq Bras Cardiol. 1985;44:305-10.

75. Shah P, Amin J. Low high-density lipoprotein level is associated with increased restenosis rate after coronary angioplasty. Circulation. 1992;85:1279-85.

76. Kuvin JT, Patel AR, Sliney KA, et al. Do ultra-low levels of low-density cholesterol confer additional benefit to endothelium? Am J Cardiol. 2005;95:93-5.

77. Nicholls SJ, Nissen SE. Strategies to promote HDL-c: an emerging therapeutic target. Eur Heart J. 2005;26:853-5.

78. Olsson AG, Schwartz GG, Szarek M, et al. High-density lipoprotein, but not low-density lipoprotein cholesterol levels influence prognosis after coronary syndrome: results from the MIRACL trial. Eur Heart J. 2005;26:890-6.

79. Betteridge DJ, Morrell JM. Clinicians' guide to Lipids and Coronary Heart Disease. London: Chapman Hall Medical; 1988. p.156.

80. Carrasco W, Lichtenstein AH, Welty FK, et al. Dietary restriction of saturated fat and cholesterol decreases HDL apo AI secretion. Arterioscler Thromb Vasc Biol. 1999;19:918-24.

81. Ginsberg H, Kris-Etherton P, Dennis B, et al. Effects of reducing dietary saturated fatty acids on plasma lipids and lipoproteins in healthy subjects: the Delta Study Protocol. Arterioscler Thromb Vasc Biol. 1998;18:441-9.

82. Walden CE, Retzlaff BM, Buck BL, et al. Differential effect of National Cholesterol Education Program (NCEP) Step II Diet on HDL cholesterol, its subfractions, and apoprotein A-I levels in hypercholesterolemic women and men after 1 year: the beFit Study. Arterioscler Thromb Vasc Biol. 2000;20:1580-7.

83. Li Z, Otvos J, Lamon-Fava S, et al. Men and women differ in lipoprotein response to dietary saturated fatty and cholesterol restriction. J Nutr. 2003;133:3428-33.

84. Mensink RP, Katan MB. Effect of dietary fatty acids on high-density and low-density lipoprotein cholesterol levels in healthy subjects. N Engl J Med. 1990;323:439-45.

85. Aude Y, Agatston AS, Lopez-Jimenez F, et al. The National Cholesterol Education Program Diet vs. a diet lower in carbohydrates and higher in protein and monounsatured fat: a randomized trial. Arch Intern Med. 2004;164:2141-6.

86. Westman EC. Is a low-carb, low-fat diet optimal? Arch Intern Med. 2005;165:1071-2.

87. Thornton J, Symes C, Heaton K. Moderate alcohol intake reduces bile cholesterol saturation and raises HDL cholesterol. Lancet. 1983;2:819-22.

88. McConnell MV, Vavouranakis I, Wu L, et al. Effects of a single, daily alcoholic beverage on lipid and hemostatic markers of cardiovascular risk. Am J Cardiol. 1997;80:1226-8.

89. Mursu J, Voutilainen B, Rissanen TH, et al. Dark chocolate consumption increases HDL cholesterol concentration and chocolate fatty acids may inhibit lipid peroxidation in healthy humans. Free Radic Biol Med. 2004;37:1351-9.

90. Dattilo AM, Kris-Etherton PM. Effects of weight reduction on blood lipids and lipoproteins: a meta-analysis. Am J Clin Nutr. 1992;56:320-8.

91. Weisweiler P. Plasma lipoproteins and lipase and lecithin-cholesterol acyltransferase activities in obese subjects before and after weight reduction. J Clin Endocrinol Metab. 1987;65:969-73.

92. Avila AL. Tratamento não farmacológico da síndrome metabólica: abordagem do nutricionista. Rev Soc Cardiol Estado de São Paulo. 2004;4:652-8.

93. Cullen P, Schulte H, Assmann G. Smoking, lipoproteins and coronary heart risk: data from the Munster Heart Study. Eur Heart J. 1998;19:1632-41.

94. Panagiotakos DB, Pitsavos G, Chrysohoou C, et al. Effect of leisure time physical activity on blood lipid levels: the ATTICA Study. Coronary Artery Disease. 2003;14:533-9.

95. Couillard C, Despres JP, Lamarche B, et al. Effects of endurance training on plasma HDL-cholesterol levels depend on levels of triglycerides: evidence from men of the Health Risk Factors, Exercise Training and Genetics (Heritage) Family Study. Arterioscler Thromb Vasc Biol. 2001;21:1226-32.

96. Wood PD, Stefanick ML, Williams PT, et al. The effects on plasma lipoprotein of a prudent weight-reducing diet, with or without exercise, in overweight men and women. N Engl J Med. 1991;325:461-6.

97. Stefanick M, Mackey S, Sheeman N, et al. Effects of diet and exercise in men and post menopausal women with low levels of HDL-cholesterol and high levels of LDL-cholesterol. N Engl J Med. 1988;339:12-20.

98. Krauss WE, Houmard JA, Duscha BD, et al. Effects of the amount and intensity of exercise on plasma lipoproteins. N Engl J Med. 2002;347:1483-92.

99. Schuler G, Hambrecht R, Schlier FG, et al. Regular physical exercise and low-fat diet. Effects on progression of coronary artery disease. Circulation. 1992;86:1-11.

100. Birjmohun RS, Hutten BA, Kastelein JJP, Stroes ES. Efficacy and safety of high-density lipoprotein cholesterol-increasing compounds: a metanalysis of randomized controlled trials. J Am Coll Cardiol. 2005;45:185-97.

101. 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;260:641-51.

102. Rubins HB, Robins SJ, Collins D, et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. N Engl J Med. 1999;341:410-8.

103. Robins SJ, Collins D, Wittes JT, et al. Relation of gemfibrozil treatment and lipid levels with major coronary events: VAHIT, a randomized controlled trial. JAMA. 2001;285:1585-91.

104. BIP Study Group. Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease: the Bezafibrate Infarction Prevention (BIP) Study. Circulation. 2000;102:21-7.

105. Rizos E, Mikhailidis DP. Are high-density lipoprotein and triglyceride levels important in secondary prevention? Impressions from the BIP and VA-HIT trials. Int J Cardiol. 2002;82:199-207.

106. Downs JR, Clearfield 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: Air Force/Texas Coronary Atherosclerosis Prevention. JAMA. 1998;279:1615-22.

107. Shepherd J, Cobbe SM, Ford I, et al., for the West of Scotland Coronary Prevention Study Group. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. N Engl J Med. 1995;333:1301-7.

108. Sacks FM, Pfeffer MA, Moye LA, et al., for the Cholesterol and Recurrent Events Trial Investigators. The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. N Engl J Med. 1996;335:1001-9.

109. Scandinavian Simvastatin Survival Study Group. Ramdomized trial of cholesterol lowering in 4,444 patients with coronary heart disease: The Scandinavian Simvastatin Survival Study (4S). Lancet. 1994;344:1383-9.

110. The 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;339:1349-57.

111. Sacks FM, Tonkin AM, Shepherd J, et al. Effects of pravastatin on coronary disease events in subgroups defined by coronary risk factors: the Prospective Pooling Project. Circulation. 2000;102:1893-900.

112. Pedersen TR, Olsson AG, Faergeman O, et al. Lipoprotein changes and reduction in incidence of major coronary heart disease events in the Scandinavian Simvastatin Survival Study (4s). Circulation. 1998;97:1453-60.

113. Ballantyne CM, Herd JA, Ferlic LL, et al. Influence of low HDL on progression of coronary artery disease and response to fluvastatin therapy. Circulation. 1999;99:736-43.

114. 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;345:1583-92.

115. 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;110:3512-7.

116. Whitney E, Krasushi RA, Personius BE, et al. A randomized trial of a strategy for increasing high-density lipoprotein levels; effects on progression of coronary heart disease and clinical events. Ann Intern Med. 2005;142:95-104.

117. Okamoto H, Yonemori F, Wakitami K, et al. A cholesteryl ester transfer protein inhibitor attenuates atherosclerosis in rabbits. Nature. 2000;406:203-7.

118. Grooth GJ, Kuivenhoven JA, Stalenhoef AF, et al. Efficacy and safety of a novel cholesteryl ester transfer protein inhibitor, JTT-705, in humans: a randomized phase II dose-response. Circulation. 2002;105:2159-65.

119. Morehouse LA, Sugarman ED, Bourassa PA, et al. HDL elevation by the CETP-inhibitor torcetrapib prevents aortic atherosclerosis in rabbits. Circulation. 2004;110(Suppl):III243.

120. Brousseau ME, Schaefer EJ, Wolfe ML, et al. Effects of an inibitor of cholesteryl ester transfer protein on HDL-cholesterol. N Engl J Med. 2004;350:1505-15.

121. Rittershaus CV, 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;20:2106-12.

122. Davidson MH, Maki K, Umporowicz D, et al. The safety and immunogenicity of a CETP vaccine in healthy adults. Atherosclerosis. 2003;169:113-20.

123. Miyasaki A, Sakuma S, Morikawa W, et al. Intravenous injection of a rabbit apolipoprotein inhibits the progression of atherosclerosis in cholesterol-fed rabbits. Arterioscler Thromb Vasc Biol. 1995;15:1882-8.

124. Nissen SE, Tsunoda T, Tuzcu EM, et al. Effect of recombinant apo AI Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. JAMA. 2003;290:2292-300.

A relação inversa entre concentrações sanguíneas de HDL-c e lesões ateroscleróticas das artérias coronárias está demonstrada por vários estudos clínicos e de intervenção terapêutica.

Manuscript received February 13, 2006; revised manuscript received May 10, 2006; accepted June 22, 2006

  • Mailing Address:
    Neusa Forti
    InCor
    Av. Dr. Enéas Carvalho de Aguiar, 44
    05403-904 – São Paulo, SP, Brazil
    E-mail:
  • Publication Dates

    • Publication in this collection
      06 Mar 2007
    • Date of issue
      Nov 2006
    Sociedade Brasileira de Cardiologia - SBC Avenida Marechal Câmara, 160, sala: 330, Centro, CEP: 20020-907, (21) 3478-2700 - Rio de Janeiro - RJ - Brazil, Fax: +55 21 3478-2770 - São Paulo - SP - Brazil
    E-mail: revista@cardiol.br