atherosclerotic biology and epidemiology of disease

Upload: mendedheart

Post on 30-May-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    1/26

    Editors: Topol, Eric J.

    Title: Textbook of Cardiovascular Medicine, 3rd Edition

    Copyright Lippincott Williams & Wilkins

    > Table of Contents > Section One - Preventive Cardiology > Chapter 1 - Atherosclerotic

    Biology and Epidemiology of Disease

    Chapter 1

    Atherosclerotic Biology and Epidemiologyof Disease

    James H.F. Rudd

    John R. Davies

    Peter L. Weissberg

    Epidemiology of Cardiovascular DiseaseAtherosclerosis, with its complications, is the leading cause of mortality and

    morbidity in the developed world. In the United States, a snapshot of the

    population reveals that 60 million adults currently suffer from atherosclerotic

    cardiovascular disease, which accounts for 42% of all deaths annually, at a cost

    to the nation of $128 billion. Fortunately, despite this catastrophic burden of

    disease, much evidence has emerged over the last decade suggesting that the

    progression of atherosclerosis can be slowed or even reversed in many people

    with appropriate lifestyle and drug interventions.

    The origin of the current ep idemic of cardiovascular disease can be traced back

    to the time of industrialization in the 1700s. The three factors largely

    responsible for this were an increase in the use of tobacco products, reduced

    physical activity, and the adoption of a diet high in fat, calories, and cholesterol.

    This rising tide of cardiovascular d isease continued into the twentieth century,

    but began to recede when data from the Framingham study identified a number

    of modifiable risk factors for cardiovascular disease, including cigarette smoking,

    hypertension, and hypercholesterolemia (1).

    The number of deaths per 100,000 attributable to cardiovascular disease peaked

    in the Western world in 1964 to 1965, since which time there has been a gradual

    decline in death rates (Fig. 1.1) (2). The age-adjusted coronary heart disease

    (CHD) mortality in the United States dropped by more than 40% and

    cerebrovascular disease mortality by more than 50%, with the greatest

    reductions being seen among whites and men. This reduction has occurred

    despite a quadrupling of the proportion of the population older than 65 years of

    age and has been due to a number of factors, particularly major health

    promotion campaigns aimed at reducing the prevalence of Framingham risk

    factors. Indeed, there has been a substantial change in the prevalence of

    population cardiovascular risk factors over the last 30 years (Table 1.1). The war

    is not won, however, and the decline in the death rate from card iovascular

    disease slowed in the 1990s (Fig. 1.2). This is likely owing to a large increase in

    the prevalence of both obesity and type 2 diabetes mellitus, as well as a

    resurgence of cigarette smoking in some sectors o f society (3). Female death

    rates from cardiovascular disease overtook male death rates in 1984 and have

    shown a smaller decline over the last 30 years (4). The consequences of

    Page 1 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    2/26

    atherosclerosis are also beginning to be felt in less well-developed regions of the

    globe (5), with death from atherosclerotic cardiovascular disease set to replace

    infection as the leading cause of death in the Third World in the near future. This

    phenomenon is further illustrated by the increase in CHD mortality in countries

    of Eastern and Central Europe (most notably countries of the former Soviet

    Union). For example, in the Ukraine the age standardized death rate in the year

    2000 was just over 800 per 100,000 people representing an increase of over60% when compared to 1990 (6).

    A further note of caution should also be struck. Western countries are

    experiencing a dramatic increase in the prevalence of heart failure. In the United

    States, almost 5 million people carry a diagnosis of heart failure (7), thus

    singling it out as an emerging epidemic (8). However, the determinants of this

    epidemic have yet to be fully elucidated, with some epidemiologic studies

    pointing toward hypertension as the driving factor (9) and others suggesting

    CHD as the predominant cause (10).

    Biology of AtherosclerosisTraditionally, atherosclerosis has been viewed as a degenerative d isease,affecting predominantly older people, slowly progressing over many years, and

    eventually leading to symptoms through mechanical effects of blood flow. The

    perceived insidious and relentless nature of its development has meant that

    a somewhat pessimistic view of the potential to modify its progression by

    medical therapy has held sway. There has been little emphasis on the diagnosis

    and treatment of high-risk asymptomatic patients. Disease management has

    instead been dominated by interventional r evascularization approaches,

    targeting the largest and most visible or symptomatic lesions with coronary

    angioplasty or bypass surgery.

    Recently, for several reasons, this defeatist view of the pathogenesis and

    progression of atherosclerosis has begun to change. First, careful descriptive

    P.3

    FIGURE 1.1. Trends in death rates for heart diseases: United States,

    19001991. (Source: Feinleib M. Trends in heart d isease in the United

    States [review]. Am J Med Sci1995;310[Suppl 1]:S8S14, with

    permission.)

    Page 2 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    3/26

    studies of the underlying pathology of atherosclerosis have revealed that

    atherosclerotic plaques differ in their cellular composition and that the cell types

    predominating in the plaque can determine the risk of fatal clinical events. A

    high degree of plaque inflammation is particularly dangerous. Second, recent

    epidemiologic work has identified many new potentially modifiable risk factors

    for atherosclerosis, above and beyond those highlighted as a result of the

    Framingham study (11). The third and most important reason is because severallarge-scale clinical trials have reported that drugs in particular, the HMG-CoA

    reductase inhibitors (statins)are able to reduce the number of clinical events

    in patients with established atherosclerosis and do so without necessarily

    affecting the size of atherosclerotic plaques. These three strands of evidence

    have shown that, rather than being an irreversibly progressive disease,

    atherosclerosis is a dynamic, inflammatory process that may be amenable to

    medical therapy. Understanding the cellular and mo lecular interactions that

    determine the development and progression of atherosclerosis brings with it

    opportunities to develop novel therapeutic agents targeting key molecular and

    cellular interactions in its etiology. In addition, the recognition that the clinical

    consequences of atherosclerosis depend almost entirely on plaque compositionargues for a new approach to diagnosis, with less emphasis placed on the degree

    of lumen narrowing and more interest in the cellular composition of the plaque.

    TABLE 1.1 Temporal Changes in Coronary RiskFactors

    Cigarette smoking 1960 Men, 55%; women, 33%

    1990 Men, 30%; women, 27%

    Undiagnosed hypertension 1960

    1980

    52%

    29%

    Mean serum cholesterol 1960

    1990

    225 mg/dL

    208 mg/dL

    Diabetes mellitus 1970 2.6%

    1990 9.1%

    Sedentary lifestyle 1970 41%

    1985 27%

    Obesity 1960 25%

    Page 3 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    4/26

    Normal Artery

    The healthy artery consists of three histologically d istinct layers. Innermost andsurrounding the lumen is the tunica intima, which comprises a single layer of

    endothelial cells in close proximity to the internal elastic lamina. The tunica

    media surrounds the internal elastic lamina, and its composition varies

    depending on the type of artery. The tunica media of the smallest arterial

    vessels, arterioles, comprises a single layer of vascular smooth muscle cells

    (VSMCs). Small arteries have a similar structure but with a thicker layer of

    medial VSMCs. Arterioles and small arteries are termed resistance vessels

    because they contribute vascular resistance and, hence, directly affect blood

    pressure. At the opposite end of the spectrum are large elastic or conduit

    arteries, named for the high proportion of elastin in the tunica media. The tunica

    media of all arteries is contained within a connective tissue layer that containsblood vessels and nerves and that is known as the tunica adventitia. In normal

    arteries, the vessel lumen diameter can be altered by contraction and relaxation

    of the medial VSMCs in response to a variety of systemic and locally released

    signals.

    Atherosclerotic VesselAtherosclerosis is primarily a disease affecting the intimal layer of elastic

    arteries. For reasons that remain largely unknown, some arterial beds appear

    more prone than others. Coronary, carotid, cerebral, and renal arteries and the

    aorta are most often involved. The arteries supplying the lower limbs are also

    vulnerable to disease. Interestingly, the internal mammary artery is almost

    always spared, making it an invaluable vessel for coronary bypass surgery.

    Atherosclerotic lesions develop over many years and pass through several

    overlapping stages. Histologically, the earliest lesion is a subendo thelial

    accumulation of lipid-laden macrophage foam cells and associated T lymphocytes

    known as a fatty streak. Fatty streaks are asymptomatic and nonstenotic.

    Postmortem examinations have shown that they are present in the aorta at the

    end of the first decade of life, are present in the coronary arteries by the

    second, and begin to appear in the cerebral circulation by the third decade. With

    time, the lesion progresses and the core of the early plaque becomes necrotic,

    containing cellular debris, crystalline cholesterol, and inflammatory cells,particularly macrophage foam cells. This necrotic core becomes bounded on its

    luminal aspect by an endothelialized fibrous cap, consisting of VSMCs embedded

    1990 38%

    From Miller M, Vogel RA. The practice of coronary disease prevention.

    Baltimore: Williams & Wilkins, 1996.

    Page 4 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    5/26

    in an extensive collagenous extracellular matrix. Inflammatory cells are also

    present in the fibrous cap, concentrated particularly in the shoulder

    regions, where T cells, mast cells, and especially macrophages have a tendency

    to accumulate. Advanced lesions may become increasingly complex, showing

    evidence of calcification, ulceration, new microvessel formation, and rupture or

    erosion (12). Microvessels within the plaque may play important roles in the

    formation of macrophage-rich vulnerable atheroma by providing an extendedsurface area of activated endothelial cells to hasten recruitment o f further

    inflammatory cells as well as by promotion of intraplaque hemorrhage (13).

    Page 5 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    6/26

    Thus, the composition of atherosclerotic plaques is variable, dynamic and

    complex, and it is the interaction between the various cell types within a plaque

    that determines the progression, complications, and outcome of the disease.

    Cellular Roles in Atherogenesis

    Endothelial CellsThe endothelium plays a central role in maintaining vascular health by virtue of

    its vital anti-inflammatory and anticoagulant properties. Many of these

    characteristics are mediated by the nitric oxide (NO) mo lecule. This molecule

    was discovered in the 1980s, having been isolated from lipopolysaccharide-

    primed macrophages (14). NO is synthesized by endothelial cells under the

    control of the enzyme endothelial NO synthase (NOS) and has a number of anti-

    atherogenic properties. First, it acts as a powerful inhibitor of platelet

    aggregation on endothelial cells. Second, it can reduce inflammatory cell

    recruitment into the intima by abrogating the expression of genes involved in

    this process, such as those encoding intercellular adhesion molecule-1 (ICAM-1),

    vascular cell adhesion molecule-1 (VCAM-1), P-selectin, and monocyte

    chemoattractant protein-1 (MCP-1) (15,16,17). There is some evidence that NO

    may also reduce lipid entry into the arterial intima (18). NO is also a potentanti-inflammatory molecule and, depending on concentration, may be a

    scavenger or a producer of potentially destructive oxygen free radicals, such as

    peroxynitrite (19,20,21). The earliest detectable manifestation of atherosclerosis

    is a decrease in the bioavailability of NO in response to pharmacologic or

    hemodynamic stimuli (22). This may occur for two reasons. Either there may be

    decreased manufacture of NO because of endothelial cell dysfunction, or

    increased NO breakdown may take place. There is evidence that both

    mechanisms may be important in differ ent situations (23). Many atherosclerosis

    risk factors can lead to impaired endothelial function and reduced NO

    bioavailability. For example, hyperlipidemic patients have reduced NO-dependent

    vasodilatation, which is reversed when patients are treated with lipid-loweringmedication (24). Diabetics also have impaired endo thelial function, occurring

    primarily as a result of impaired NO production. There is, however, some

    evidence to suggest that increased oxidative stress leading to enhanced NO

    breakdown may also be a factor in early endothelial dysfunction (25). Similarly,

    other risk factors for atherosclerosis, such as hypertension and cigarette

    smoking, are associated with reduced NO bioavailability (26,27). In cigarette

    smokers, endothelial impairment is thought to be caused by enhanced NO

    degradation by oxygen-derived free-radical agents such as the superoxide ion.

    There are also other consequences of an increased reactivity between NO and

    superoxide species. The product of t heir interaction, ONOO (peroxynitrite), is

    a powerful oxidizing agent and can reach high concentrations in atheroscleroticlesions. This may result in cellular oxidative injury.

    FIGURE 1.2. A. Death rates from CHD, men and women aged 3574,

    2000, selected countries. B. Changes in death rates from CHD, men and

    women aged 3574, between 1990 and 2000, selected countries.

    P.4

    P.5

    Page 6 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    7/26

    Another consequence of endothelial cell dysfunction that occurs in early

    atherosclerosis is the expression of surface-bound selectins and adhesion

    molecules, including P-selectin, ICAM-1, and VCAM-1. These molecules attract

    and capture circulating inflammatory cells and facilitate their migration into the

    subendothelial space (22). Normal endothelial cells do not express these

    molecules, but their appearance may be induced by abnormal arterial shear

    stress, subendothelial oxidized lipid, and, in diabetic patients, the presence ofadvanced glycosylation products in the arterial wall. The importance of selectins

    and adhesion molecules in the development of atherosclerosis is demonstrated in

    experiments using mice, which lack their expression. These animals develop

    smaller lesions with a lower lipid content and fewer inflammatory cells than

    control mice when fed a lipid-rich diet (28). Animal models have reinforced the

    importance of inflammatory cell recruitment to the pathogenesis of

    atherosclerosis, but because inflammatory cells are never seen in the intima in

    the absence of lipid, the results suggest that subendothelial lipid accumulation is

    also necessary for the development of atherosclerosis.

    The tendency for atherosclerosis to occur preferentially in particular sites may

    be explained by subtle variations in endothelial function. This is probably caused

    by variations in local blood flow patterns, especially conditions of low flow, which

    can influence expression of a number of endothelial cell genes, including those

    encoding ICAM-1 and endothelial NOS (29,30). In addition to flow speed, flow

    type can have a direct effect on cell morphology. In areas of laminar flow

    (atheroprotective flow), endothelial cells tend to have an ellipsoid shape,

    contrasting with the situation found at vessel branch points and curves, where

    turbulent flow (atherogenic flow) induces a conformational change toward

    polygonal-shaped cells (31). Such cells have an increased permeability to low-

    density lipoprotein (LDL) cholesterol and may promote lesion formation (32).

    These data are consistent with the idea that the primary event in atherogenesisis endothelial dysfunction. The endothelium can be damaged by a variety of

    means, leading to dysfunction and, by unknown mechanisms, subsequent

    subendothelial lipid accumulation. In this situation, the normal homeostatic

    features of the endothelium break down; it becomes more adhesive to

    inflammatory cells and platelets, it loses its anticoagulant properties, and there

    is reduced bioavailability of NO. Importantly, endothelial function is improved by

    drugs that have been shown to substantially reduce death from vascular disease,

    including statins and angiotensin-converting enzyme inhibitors (33,34).

    Inflammatory Cells

    LDL from the circulation is able to diffuse passively through the tight junctionsthat bind neighboring endothelial cells. The rate of passive diffusion is increased

    when circulating levels of LDL are elevated. In add ition, other lipid fractions may

    be important in atherosclerosis. Lipoprotein(a) has the same basic molecular

    structure as LDL, with an additional apolipoprotein(a) element attached by a

    disulfide bridge. It has been shown to be highly atherogenic (35), accumulate in

    the arterial wall in a manner similar t o LDL (36), impair vessel fibrinoly sis (37),

    and stimulate smooth muscle cell proliferation (38). The accumulation of

    subendothelial lipids, particularly when at least partly oxidized, is thought to

    stimulate the local inflammatory reaction that initiates and maintains activation

    of overlying endothelial cells. The activated cells express a variety of selectins

    and adhesion molecules and also produce a number of chemokinesinparticular, MCP-1, whose expression is upregulated by the presence of oxidized

    Page 7 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    8/26

    LDL in the subendothelial space (39). Interestingly, the protective effect of high-

    density lipoprotein (HDL) against atherosclerotic vascular disease may be partly

    explained by its ability to block endothelial cell expression of adhesion molecules

    (40,41). Chemokines are proinflammatory cytokines responsible for

    chemoattraction, migration, and subsequent activation of leukocytes. Mice

    lacking the MCP-1 gene develop smaller atherosclerotic lesions than normal

    animals (42). The first stage of inflammatory cell recruitment to the intima isthe initiation of rolling of monocytes and T cells along the endothelial cell

    layer. This phenomenon is mediated

    by the selectin molecules, which selectively bind ligands found on these

    inflammatory cells. The subsequent firm adhesion to and migration of leukocytes

    through the endothelial cell layer depends on the endothelial expression of

    adhesion molecules such as ICAM-1 and VCAM-1 and their binding to appropriate

    receptors on inflammatory cells. Once present in the intima, monocytes

    differentiate into macrophages under the influence of chemokines such as

    macrophage colony-stimulating factor. Such molecules also stimulate the

    expression of the scavenger receptors that allow macrophages to ingest oxidizedlipids and to develop into macrophage foam cells, the predominant cell in an

    early atherosclerotic lesion. The formation of scavenger receptors is also

    regulated by peroxisome proliferator-activated receptor- (PPAR- a nuclear

    transcription factor expressed at high levels in foam cells) (43). PPAR- agonists

    (glitazones), which are used to treat patients with type 2 diabetes, have been

    shown to have many anti-atherogenic effects, including increasing production of

    NO (44), decreased endothelial inflammatory cell recruitment and reduced

    vascular endothelial growth factor (VEGF) expression (45). Also, PPAR-

    agonists can reduce the lipid content of plaques by enhancing reverse cholesterol

    transport from plaque to liver. Positive results with these drugs in patients with

    type 2 diabetes are emerging. As well as reducing matrix metalloproteinase(MMP) 9 levels, glitazones also significantly ameliorated C-reactive protein (CRP)

    and CD40 ligand levels, as well as causing d irect plaque regression in a rabbit

    atheroma model (46). Clearly their u se in large clinical trials in patients without

    diabetes as anti-atheroma drugs is awaited with interest.

    In early atherosclerosis at least, the macrophage can be thought of as

    performing a predominantly beneficial role as a neutralizer of potentially

    harmful oxidized lipid components in the vessel wall. However, macrophage foam

    cells also synthesize a variety of proinflammatory cytokines and growth factors

    that contribute both beneficially and detrimentally to the evolution of the plaque.

    Some of these factors are chemoattractant (osteopontin) and growth-enhancing

    (platelet-derived growth factor) for VSMCs (12,47). Under the influence of thesecytokines, VSMCs migrate from the media to the intima, where they adopt a

    synthetic phenotype, well-suited to matrix production and protective fibrous cap

    formation.

    However, activated macrophages have a high rate of apoptosis. Once dead, they

    release their lipid content, which becomes part of the core of the plaque, thereby

    contributing to its enlargement. The apoptotic cells also contain high

    concentrations of tissue factor, which may invoke thrombosis if exposed to

    circulating platelets (48). Interestingly, the selective glycoprotein 2b3a receptor

    antagonist abciximab has been shown to have an effect on the levels of tissue

    factor found in monocytes. In an in vitro study by Steiner (49), the drug

    attenuated both the amount of tissue factor and its RNA levels. As tissue factor

    is a potent instigator of the clotting cascade, this role may explain part of the

    P.6

    Page 8 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    9/26

    protective effect of abciximab on the microcirculation of patients with acute

    coronary syndromes (50).

    It is now generally recognized that the pathologic progression and consequences

    of atherosclerotic lesions are determined by dynamic interactions between

    inflammatory cells recruited in response to subendothelial lipid accumulation,

    and the local reparative wound healing response of surrounding VSMCs.

    Vascular Smooth Muscle CellsVSMCs reside mostly in the media of healthy adult arteries, where their role is to

    regulate vascular tone. Thus, medial VSMCs contain large amounts of contractile

    proteins, including myosin, -actin, and tropomyosin. Continued expression of

    this contractile phenotype is maintained by the influence of extracellular

    proteins in the media, which act via integrins in the VSMC membrane. In

    atherosclerosis, however, the cells become influenced by cytokines produced by

    activated macrophages and endothelial cells. Under these influences, VSMCs

    migrate to the intima and undergo a phenotypic change characterized by a

    reduction in content of contractile proteins and a large increase in the number ofsynthetic organelles. This migration of VSMCs from the media to the intima, and

    the consequent change from a contractile to a synthetic phenotype, was

    previously thought to be a crucial step in the development of atherosclerosis in

    the modified response to injury hypothesis discussed previously. More recently,

    it has been recognized that intimal VSMCs in atherosclerotic plaques bear a

    remarkable similarity to VSMCs found in the early developing blood vessels (51),

    suggesting that intimal VSMCs may be performing a beneficial, reparative role

    rather than a destructive one in atherosclerosis. VSMCs are well-equipped for

    this action. First, they can express the prot einases that they require to break

    free from the medial basement membrane and allow them to migrate to the site

    of inflammation or injury in response to chemokines. Second, they can producevarious growth factors, including VEGF and platelet-derived growth factor, that

    act in an autocrine loop to facilitate their proliferation at the site of injury.

    Finally, and most important, they produce large quantities of matrix proteins, in

    particular glycosaminoglycans, elastin, and collagen isoforms 1 and 3, necessary

    to repair the vessel and form a fibrous cap over the lipid-rich core of the lesion.

    This fibrous cap separates the highly thrombogenic lipid-rich plaque core from

    circulating platelets and the proteins of the coagulation cascade and also confers

    structural stability to the atherosclerotic lesion. And because the VSMC is t he

    only cell capable of synthesizing this cap, it follows that VSMCs play a pivo tal

    role in maintaining plaque stability and protecting against the potentially fatal

    thrombotic consequences of atherosclerosis (52).

    Cellular Interactions and Lesion StabilityGenerally, early atherosclerosis progresses without symptoms until a lesion

    declares itself in one of two ways. As discussed, macrophage foam cells may

    undergo apoptosis, especially in the p resence of high concentrations of oxidized

    LDL. Their cellular remnants then become part of an enlarging lipid-rich core.

    Plaque size thus increases, and there may be a consequent reduction in vessel

    lumen area. At times of increased demand, such as exercise, this may be

    sufficient to cause ischemic symptoms such as angina. More hazardous is if the

    plaque presents with disruption of the fibrous cap, leading to exposure of the

    thrombogenic lipid core. This is likely to result in subsequent plateletaccumulation and activation, fibrin deposition, and intravascular thrombosis.

    Page 9 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    10/26

    Depending on factors such as collateral blood supply, extent of arterial

    thrombus, and local fibrinolytic activity, the end result may be arterial occlusion

    and downstream necrosis.

    By studying the pathology of ruptured plaques, several characteristics have been

    identified that seem to be predictive of the risk of rupture in individual lesions

    (53). Plaques that are vulnerable to rupture tend to have thin fibrous caps (

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    11/26

    range, and a correlation between CRP level and coronary events was

    demonstrated only after development of a highly sensitive assay for CRP that

    was capable of measuring levels below the lower limit of detection of

    conventional assays. The risk of clinical events associated with an elevated CRP

    seems to be independent of the presence of other Framingham risk factors for

    atherosclerosis. Elevated CRP also predicts near-term plaque rupture events as

    well those up to 20 years in the future, suggesting that inflammation isimportant in both early and late atherosclerosis (71). Additionally, CRP level

    accurately indicates the likelihood of sudden cardiac death, a condition usually

    associated with multiple atherosclerotic plaque ruptures (72). However, despite

    initial enthusiasm, large meta-analyses have suggested that the relative risk of a

    cardiovascular event is increased by only approximately 1.5 times in those

    people with a baseline elevated CRP (above 3 mg/dL) (73,74). With such a

    modest predictive value, it may be that the routine measurement of CRP alone in

    asymptomatic patients is not yet justified for accurate disease prediction.

    Similar, although less compelling, correlations with clinical events have also

    been published for other markers of inflammation, including soluble ICAM-1,

    VCAM-1, P-selectin, and interleukin-6 (the primary driver of CRP production)

    (75,76,77,78). Results of these studies have been interpreted by some as

    indicating that atherosclerosis arises as a consequence of a systemic

    inflammatory process (e.g., chronic infection) and by others that it reflects the

    inflammatory processes of atherosclerosis itself. However, there is accumulating

    evidence in favor of the latter interpretation.

    Two Forms of Plaque Disruption: Fibrous CapRupture and Endothelial ErosionAtherosclerotic plaques become life threatening when they initiate clot formation

    in the vessel lumen and disturb b lood flow. This can occur in two different ways.Either there can be fibrous cap rupture, with consequent exposure of the

    thrombogenic extracellular matrix of the cap and the tissue factorrich lipid

    core to circulating blood, o r less commonly, there is erosion of the endothelial

    cells covering the fibrous cap, also potentially leading to the formation of a

    platelet-rich thrombus. Endothelial erosion probably accounts for approximately

    30% of acute coronary syndromes overall and seems particularly common in

    women (79). Both forms of plaque disruption invariably lead to local platelet

    accumulation and activation. This may result in triggering of the clotting

    cascade, thrombus formation, and, if extensive, complete vessel occlusion.

    Platelet-rich thrombus contains chemokines and mitogens, in particular platelet-

    derived growth factor and thrombin that induce migration and proliferation of

    VSMCs from the arterial media to the plaque and transforming growth factor-

    that contributes to healing of the disrupted lesion (80). Platelets also express

    CD40 on their cell membrane, which causes local endothelial cell activation,

    resulting in the recruitment of more inflammatory cells to the lesion and

    perpetuating the cycle of inflammation, rupture, and thrombosis. However,

    fibrous cap rupture or erosion does not invariably lead to vessel occlusion. Up to

    70% of plaques causing high-grade stenosis contain histologic evidence of

    previous subclinical plaque rupture with subsequent repair (81). This is

    particularly likely to occur if high blood flow through the vessel prevents the

    accumulation of a large occlusive thrombus. Thus, nonocclusive plaque rupture

    induces formation of a new fibrous cap over the organizing thrombus, which

    restabilizes the lesion but at the expense of increasing its size. Because this

    occurs suddenly, there is little opportunity for adaptive remodeling of the artery,

    Page 11 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    12/26

    and the healed lesion may now impede flow sufficiently to produce ischemic

    symptoms. This explains why patients who have previously had normal exercise

    tolerance may suddenly develop symptoms of stable angina pectoris. It also

    follows that if lesions can grow as a consequence of repeated episodes of silent

    rupture and repair, an inhibition of plaque rupture rate will reduce progression of

    atherosclerosis. Therefore, atheromatous plaques may become larger by two

    methods. The first is a gradual increase in size as a consequence of macrophagefoam cell accumulation and incorporation of apoptotic cells into an enlarging

    necrotic lipid-laden plaque core. The second is a stepwise increase in size

    because of repeated, often silent episodes

    of plaque rupture or erosion with subsequent VSMC-driven repair.

    Balance of Atherosclerosis: TherapeuticImplicationsAtherosclerosis is a dynamic process in which the balance between the

    destructive influence of inflammatory cells and the r eactive, stabilizing effects of

    VSMCs determines outcome (Fig. 1.3). This balance can be tipped toward plaque

    rupture by factors such as an atherogenic lipoprotein profile, high levels of lipid

    oxidation, local free radical generation, and genetic variability in expression and

    activity of certain central inflammatory molecules. For example, an association

    between plaque progression and a polymorphism in the stromelysin-1 gene

    promoter has been described (82). Until recently, it was also thought that

    infectious organisms might be involved in atherosclerosis, either as plaque

    initiators or as having some role in initiating plaque rupture. Chlamydia

    pneumoniae is found in plaques, localizing at high concentrations within

    macrophages, but is rarely found in normal arteries (83). Although these data

    imply a pathologic association between the

    presence of chlamydia infection and atherosclerosis, neither a causative role nor

    an association between serum markers of infection and ischemic heart disease

    has been established. Although animal work has shown that healthy rabbits

    nasally inoculated with chlamydia develop extensive atherosclerosis (84), the

    situation appears to be somewhat different in humans. Two large prospective

    studies and an extensive meta-analysis of previous data failed to show any

    association between serum markers of infection with chlamydia and incidence of

    or mortality from ischemic heart disease (85,86). These results effectively

    excluded a strong association but allowed the possibility of a weaker link. This

    hypothesis has now been effectively rejected after several negative trials of

    antibiotics in coronary artery disease (87,88,89).

    P.8

    P.9

    Page 12 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    13/26

    The balance can be tipped toward plaque stability by a reduction in plaque

    inflammation or an increase in VSMC-driven repair. Lipid reduction, by whatever

    means, reduces clinical events. Evidence that this may be due t o a plaque-

    stabilizing effect comes from animal studies that showed that statins reduced

    inflammatory cell and increased VSMC content of p laques (90,91), changes that

    would be expected to enhance stability. Dietary lip id lowering in rabbits also

    reduced the number of microvessels in the aortic intima, suggesting another

    mechanism of favorably altering the biology of plaques (92).

    More important, however, evidence from human clinical studies also points to a

    plaque-stabilizing effect of statins. Despite angiographic studies showing t hat

    statins produce only a small, hemodynamically insignificant reduction in lumen

    stenosis (93,94), more sensitive intravascular ultrasound studies have shown

    beyond doubt that statins can halt lesion enlargement in the coronary arteries,

    with the most benefit being seen with higher doses of the most potent drugs

    (95). Statins can also reduce new lesion formation, and, importantly, the number

    of new vessel occlusions. These arise after a plaque ruptu res, leading to an

    occlusive thrombus in the context of a well-collateralized myocardial circulation.

    This seems to imply that statins stabilize plaques by reducing rupture rate. Thisconclusion is supported by the results of all the large primary and secondary

    prevention studies, which have demonstrated that statins (p ravastatin,

    simvastatin, and lovastatin) produce major reductions in events owing to plaque

    rupture, such as myocardial infarction and stroke (34,96,97,98,99). Because

    statins have only a modest effect on plaque size but cause profound reductions

    in the number of clinical events, these studies highlight the inadequacy of

    angiography for the prediction of clinical events and suggest that statins have

    beneficial effects on plaque inflammation in addition to, or as a result o f, their

    lipid-lowering effects. Importantly, this notion is supported by the observation

    that the reduction in clinical events due to statin therapy is accompanied by a

    parallel reduction in CRP levels that is unlikely to be caused by effects of statinson nonatherosclerotic inflammation (100,101). Also, in the first study of its kind,

    it has been shown that statins reduce inflammation and increase plaque collagen

    FIGURE 1.3. Cellular interactions in the development and progression of

    atherosclerosis. (Source: Weissberg PL. Atherogenesis: current

    understanding of the causes of atheroma. Heart2000;83:247 252, with

    permission.)

    Page 13 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    14/26

    content in human carotid artery atherosclerosis (102). However, the various

    statins do differ in their anti-inflammatory effect; in the REVERSAL study,

    atorvastatin achieved a far greater reduction in CRP than pravastatin (95);

    whether this has important clinical relevance is not yet known.

    Statin drugs may help to stabilize plaques in a number of different ways. It is

    known that they can exert d irect effects on endothelial cell function,

    inflammatory cell number and activity, VSMC proliferation, platelet aggregation,

    and thrombus formation (103,104,105,106,107). Evidence that non lipid-

    lowering effects may be important in vivo comes from animal studies in which

    pravastatin caused beneficial changes in plaque composition (but not size), even

    when lipid levels were maintained at pretreatment levels (91). Additionally, in

    mice, simvastatin has direct anti-inflammatory effects comparable to those of

    indomethacin (108). Recently, a newly recognized effect o f statins as immune

    modulators has been described, whereby major histocompatibility complex class

    IImediated T-cell activation is reduced by a variety of statins (109). However,

    the matter of nonlipid-lowering effects of statins is not yet proven beyond

    doubt: several of the pleiotrop ic anti-inflammatory effects of statins (decreased

    expression of MMPs, and tissue factor) o ccur in animals on a lipid-lowering d iet

    alone, without exposure to drugs of any kind (110). In addition, the

    administration of other forms of anti-inflammatory drugs to patients with

    atherosclerosis does not seem to confer any clinical benefit and may do harm;

    the cyclooxygenase-2 (COX-2) class of drugs are a case in point, causing a

    doubling of the rate of myocardial infarction in one study (111).

    RestenosisRestenosis describes the late loss of gain in lumen diameter achieved

    immediately after balloon dilatation of an atherosclerotic plaque. For many

    years, it has been thought of as an undesirable response to vascular injury.However, in effect, it represents an extreme form of plaque stabilization.

    Whether performed on a stable or unstable plaque, balloon angioplasty causes

    endothelial disruption and often substantial damage to the full thickness of the

    vessel wall. The initial thrombotic response that would otherwise lead to early

    vessel occlusion is prevented by antiplatelet and antithrombotic therapy. There

    then follows a reparative response driven by medial VSMCs and adventitial

    myofibroblasts. The former form a matrix-rich neointima over the exposed

    plaque, whereas the latter produce a collagenous matrix in the adventitia. The

    net result is that the adventitial reaction splints the vessel and prevents

    the positive remodeling that would normally allow expansion of the vessel to

    accommodate the neointima. However, although this phenomenon may lead to

    angiographic or clinical restenosis, much more important, it renders the lesion

    stable, making the likelihood of a further plaque rupture at that site extremely

    remote. In effect, by stimulating a vigorous VSMC repair response, balloon

    angioplasty tips the balance of atherosclerosis in favor of plaque stability.

    This phenomenon undoubtedly underlies the success of angioplasty in the

    treatment of acute myocardial infarction. Most o f the adverse effects of the

    response to balloon angioplasty on remodeling can be countered by deployment

    of a stent, particularly the drug-eluting variety, where significant restenosis is

    rarely encountered. The drugs used to coat the stents are antiproliferative

    agents, and are highly effective at eliminating restenosis (112). However, by

    impairing the synthetic ability of the VSMCs of the cap, there have been r eports

    of early thrombotic occlusions of treated arteries, although longer term analysis

    of the data suggest that t his is not frequent (113). Nevertheless, drug-eluting

    Page 14 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    15/26

    stents are likely to become universally used in the catheter laboratory in the

    near future.

    Controversies and Personal PerspectivesMany issues concerning the initiation and progression of atherosclerosis remain

    to be resolved. In particular, controversy persists over the extent to which

    endothelial dysfunction precedes or is the consequence of intimal lipid

    accumulation; the relative contributions of endothelial erosion and plaque

    rupture to clinical events; and the ext ent to which statins achieve their plaque-

    stabilizing effects directly via lipid lowering or by their so-called pleiotropic

    effects on the intercellular interactions that lead to plaque rupture. Integral to

    this

    last issue is the outstanding question of what is the optimal level of lipid

    reduction. In other words, is lower LDL always better?

    Despite these controversies, it is certain that drug treatment will become

    increasingly prominent in the management of patients with, and at high risk ofdeveloping, atherosclerosis. Improvements in drug design will come from a

    number of complementary approaches. First, improvement will come by

    modifications of existing molecules, based on understanding how currently

    available drugs such as statins and angiotensin-converting enzyme inhibitors

    influence plaque progression. This will include evaluation of how other lipid-

    modifying strategies, such as inhibiting cholesterol absorption in the gut and

    modifying the balance between pro- and anti-atherogenic lipoproteins and

    triglycerides, might influence the atherosclerotic process. Second, improvements

    will come by targeting molecular interactions known to be involved in

    atherogenesis. Likely candidates include endothelial adhesion molecules, MMPs,

    inflammatory cytokines and their signaling molecules, in particular, nuclearfactor- B and its downstream transcriptional activators. Here the challenge lies

    in identifying pathways or molecular species that are specific for atherosclerosis

    whose modification will not compromise the normal inflammatory response to

    pathogens. This approach will include developing regulators of VSMC behavior,

    such as modulators of transforming growth factor- driven matrix production,

    that may lead to enhanced maintenance of the fibrous cap. Another important

    example includes establishing the role of drugs targeting peroxisome

    proliferatoractivated receptors in modifying inflammation and the vascular

    consequences of the metabolic syndrome that links insulin resistance, diabetes,

    hypertension, and dyslipidemia with premature atherosclerosis. The third

    approach is to use new technologies such as p roteomics to design new

    therapeutic molecules and gene array technologies to identify new molecular

    targets in vascular disease. In addition, as a consequence of sequencing the

    human genome, a number of orphan receptors have already been

    identified that might provide vascular-specific targets for novel therapies.

    Finally, local drug delivery to high-risk plaques with drug-eluting stents has been

    proposed as a means of reducing risk of rupture (plaque passivation) (115,116).

    This approach will need better methods of identifying high-risk plaques, which

    will probably include invasive imaging data derived from IVUS and thermography

    coupled with noninvasive methods such as high-resolution molecular magnetic

    resonance imaging and possibly Fluorodeoxyglucose positron emission

    tomography (FDG-PET) (117,118).

    P.10

    Page 15 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    16/26

    The FutureIt is almost inconceivable that advances in our understanding of the

    atherosclerotic disease process will not lead to the development of new anti-

    atheroma drugs that will act synergistically with statins and angiotensin-

    converting enzyme inhibitors. For example, a novel HDL-like molecule has

    recently been shown to reduce atheroma burden when given by intravenousinfusion over 5 weeks to a high-risk group of patients (114). Furthermore, we

    predict that advances in genetics and diagnostics will combine with therapeutic

    advances to produce substantial reductions in p remature cardiovascular deaths.

    Thus, new gene polymorphisms and mutations will be identified that confer

    increased likelihood either of developing atheroma or of experiencing its

    consequences. This will lead, in turn, to better prescription of lifestyle

    modifications and better targeting of current and new therapies for primary

    prevention of cardiovascular events. This approach will be led by new diagnostic

    testsbased on specific circulating markers of vascular inflammation and

    imaging of the inflammatory process underlying plaque rupture that will allow

    better preclinical diagnosis of patients at gr eatest risk of cardiovascular eventsand subsequent monitoring of plaque-modifying therapies.

    References

    1. Wong ND, Wilson PW, Kannel WB. Serum cholesterol as a prognostic factor

    after myocardial infarction: the Framingham Study. Ann Intern Med

    1991;115:687 693.

    2. National Heart, Lung, and Blood Institute (NHLBI). NHLBI fact book fiscal

    year 1997. Bethesda, MD: NHBLI, 1998.

    3. Cooper R, Cutler J, Desvigne-Nickens P, et al. Trends and disparities in

    coronary heart disease, stroke, and other cardiovascular diseases in the

    United States: findings of the national conference on cardiovascular disease

    prevention. Circulation 2000;102:3137 3147.

    4. McGovern PG, Pankow JS, Shahar E, et al. Recent trends in acute coronary

    heart diseasemortality, morbidity, medical care, and risk factors. The

    Minnesota Heart Survey Investigators. N Engl J Med1996;334:884 890.

    5. Reddy KS, Yusuf S. Emerging epidemic of cardiovascular disease in

    developing countries. Circulation 1998;97:596 601.

    6. British Heart Foundation Health Promotion Research Group. Coronary

    heart disease statistics. London: British Heart Foundation, 2005:1 200.

    7. Hunt SA, Baker DW, Chin MH, et al. ACC/AHA guidelines for the evaluation

    and management of chronic heart failure in the adult: executive summary. A

    report of the American College of Cardiology/American Heart Association

    Task Force on Practice Guidelines (Committee to revise the 1995 Guidelinesfor the Evaluation and Management of Heart Failure). J Am Coll Cardiol

    Page 16 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    17/26

    2001;38:2101 2113.

    8. Braunwald E. Shattuck lecture cardiovascular medicine at the turn of

    the millennium: triumphs, concerns, and opportunities. N Engl J Med1997;

    337:1360 1369.

    9. Ho KK, Pinsky JL, Kannel WB, et al. The epidemiology of heart failure: the

    Framingham Study. J Am Coll Cardiol1993;22:6A 13A.

    10. Gheorghiade M, Bonow RO. Chronic heart failure in the United States: a

    manifestation of coronary artery disease. Circulation 1998;97:282 289.

    11. Fruchart JC, Nierman MC, Stroes ES, et al. New risk factors for

    atherosclerosis and patient risk assessment. Circulation 2004;109:III15-

    III19.

    12. Shanahan CM, Cary NR, Metcalfe JC, et al. High expression of genes fo r

    calcification-regulating proteins in human atherosclerotic plaques. J Clin

    Invest1994;93:2393 2402.

    13. Aikawa M, Sugiyama S, Hill CC, et al. Lipid lowering reduces oxidative

    stress and endothelial cell activation in rabbit atheroma. Circulation

    2002;106:1390 1396.

    14. Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the

    biological activity of endothelium-derived relaxing factor. Nature 1987;

    327:524 526.

    15. Gauthier TW, Scalia R, Murohara T, et al. Nitric oxide prot ects against

    leukocyte-endothelium interactions in the early stages of

    hypercholesterolemia. Arterioscler Thromb Vasc Biol1995;15:1652 1659.

    16. Tsao PS, Buitrago R, Chan JR, et al. Fluid flow inhibits endothelial

    adhesiveness. Nitric oxide and transcriptional regulation of VCAM-1.

    Circulation 1996;94:1682 1689.

    17. Tsao PS, Wang B, Buitrago R, et al. Nitric oxide regulates monocyte

    chemotactic protein-1. Circulation 1997;96:934 940.

    18. Cardona-Sanclemente LE, Born GV. Effect of inhibition of nitric oxide

    synthesis on the uptake of LDL and fibrinogen by arterial walls and other

    organs of the rat. Br J Pharmacol1995;114:1490 1494.

    19. Anggard E. Nitric oxide: mediator, murderer, and medicine [see

    comments]. Lancet1994;343:1199 1206.

    Page 17 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    18/26

    20. Bhagat K, Vallance P. Nitric oxide 9 years on. J R Soc Med

    1996;89:667 673.

    21. Hobbs AJ, Higgs A, Moncada S. Inhibition of nitric oxide synthase as a

    potential therapeutic target. Annu Rev Pharmacol Toxicol

    1999;39:191 220.

    22. Ross R. Atherosclerosis-an inflammatory disease. N Engl J Med

    1999;340:115 126.

    23. Li H, Forstermann U. Nitric oxide in the pathogenesis of vascular disease.

    J Pathol2000;190:244 254.

    24. Stroes ES, Koomans HA, de Bruin TW, et al. Vascular function in the

    forearm of hypercholesterolaemic patients off and on lipid-loweringmedication. Lancet1995;346:467 471.

    25. Williams SB, Cusco JA, Roddy MA, et al. Impaired nitric oxide-mediated

    vasodilation in patients with non-insulin-dependent diabetes mellitus. J Am

    Coll Cardiol1996;27:567 574.

    26. Heitzer T, Just H, Munzel T. Antioxidant vitamin C improves endothelial

    dysfunction in chronic smokers [see comments]. Circulation 1996;94:6 9.

    27. Panza JA, Garcia CE, Kilcoyne CM, et al. Impaired endothelium-

    dependent vasodilation in patients with essential hypertension. Evidence that

    nitric oxide abnormality is not localized to a single signal transduction

    pathway. Circulation 1995;91:1732 1738.

    28. Nakashima Y, Plump AS, Raines EW, et al. ApoE-deficient mice develop

    lesions of all phases of atherosclerosis throughout t he arterial tree.

    Arterioscler Thromb 1994;14:133 140.

    29. Resnick N, Yahav H, Khachigian LM, et al. Endothelial gene regulation by

    laminar shear stress. Adv Exp Med Biol1997;430:155 164.

    30. Topper JN, Cai J, Falb D, et al. Identification of vascular endothelial

    genes differentially responsive to fluid mechanical stimuli: cyclooxygenase-2,

    manganese superoxide dismutase, and endothelial cell nitric oxide synthase

    are selectively up-regulated by steady laminar shear stress. Proc Natl Acad

    Sci U S A 1996;93:10417 10422.

    31. Cunningham KS, Gotlieb AI. The role of shear stress in the pathogenesis

    of atherosclerosis. Lab Invest2005;85:9 23 .

    P.11

    Page 18 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    19/26

    32. Gimbrone MA, Jr. Vascular endothelium, hemodynamic forces, and

    atherogenesis. Am J Pathol1999;155:1 5.

    33. Yusuf S, Sleight P, Pogue J, et al. Effects of an angiotensin-converting-

    enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients.

    The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med

    2000;342:145 153.

    34. 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. The Long-Term Intervention with Pravastatin in

    Ischaemic Disease (LIPID) Study Group. N Engl J Med

    1998;339:1349 1357.

    35. Poon M, Zhang X, Dunsky KG, et al. Apolipoprotein(a) induces monocyte

    chemotactic activity in human vascular endothelial cells. Circulation1997;96:2514 2519.

    36. Rath M, Niendorf A, Reblin T, et al. Detection and quantification of

    lipoprotein(a) in the arterial wall of 107 coronary bypass patients.

    Arteriosclerosis 1989;9:579 592.

    37. Loscalzo J, Weinfeld M, Fless GM, et al. Lipoprotein(a), fibrin binding,

    and plasminogen activation. Arteriosclerosis 1990;10:240 245.

    38. Grainger DJ, Kirschenlohr HL, Metcalfe JC, et al. Proliferation of human

    smooth muscle cells promoted by lipoprotein(a). Science

    1993;260:1655 1658.

    39. Boring L, Gosling J, Cleary M, et al. Decreased lesion formation in CCR2-

    /mice reveals a role for chemokines in the initiation of atherosclerosis.

    Nature 1998;394:894 897.

    40. Xia P, Vadas MA, Rye KA, et al. High density lipoproteins (HDL) interrupt

    the sphingosine kinase signaling pathway. A possible mechanism forprotection against atherosclerosis by HDL. J Biol Chem

    1999;274:33143 33147.

    41. Calabresi L, Franceschini G, Sirtori CR, et al. Inhibition of VCAM-1

    expression in endothelial cells by reconstituted high density lipoproteins.

    Biochem Biophys Res Commun 1997;238:61 65.

    42. Gosling J, Slaymaker S, Gu L, et al. MCP-1 deficiency reduces

    susceptibility to atherosclerosis in mice that overexpress human

    apolipoprotein B.J Clin Invest

    1999;103:773

    778.

    Page 19 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    20/26

    43. Tontonoz P, Nagy L, Alvarez JG, et al. PPARgamma promotes

    monocyte/macrophage differentiation and uptake of oxidized LDL. Cell

    1998;93:241 252.

    44. Calnek DS, Mazzella L, Roser S, et al. Peroxisome proliferator-activated

    receptor gamma ligands increase release of nitric oxide from endothelial

    cells. Arterioscler Thromb Vasc Biol2003;23:52 57.

    45. Panigrahy D, Singer S, Shen LQ, et al. PPARgamma ligands inhibit

    primary tumor growth and metastasis by inhibiting angiogenesis. J Clin

    Invest2002;110:923 932.

    46. Corti R, Osende JI, Fallon JT, et al. The selective peroxisomal

    proliferator-activated receptor-gamma agonist has an additive effect on

    plaque regression in combination with simvastatin in experimental

    atherosclerosis: in vivo study by high- resolution magnetic resonanceimaging. J Am Coll Cardiol2004;43:464 473.

    47. Liaw L, Almeida M, Hart CE, et al. Osteopontin promotes vascular cell

    adhesion and spreading and is chemotactic for smooth muscle cells in vitro.

    Circ Res 1994;74:214 224.

    48. Zaman AG, Helft G, Worthley SG, et al. The role of plaque rupture and

    thrombosis in coronary artery disease. Atherosclerosis 2000;149:251 266.

    49. Steiner S, Seidinger D, Huber K, et al. Effect of glycoprotein IIb /IIIa

    antagonist abciximab on monocyte-platelet aggregates and tissue factor

    expression. Arterioscler Thromb Vasc Biol2003;23:1697 1702.

    50. CAPTURE investigators. Randomised placebo-controlled trial of abciximab

    before and during coronary intervention in refractory unstable angina: the

    CAPTURE Study. Lancet1997;349:1429 1435.

    51. Shanahan CM, Weissberg PL. Smooth muscle cell heterogeneity: patterns

    of gene expression in vascular smooth muscle cells in vitro and in vivo.Arterioscler Thromb Vasc Biol1998;18:333 338.

    52. Libby P. Molecular bases of the acute coronary syndromes. Circulation

    1995;91:2844 2850.

    53. Galis ZS, Sukhova GK, Lark MW, et al. Increased expression of matrix

    metalloproteinases and matrix degrading activity in vulnerable regions of

    human atherosclerotic plaques. J Clin Invest1994;94:2493 2503.

    54. Davies MJ. Stability and instability: two faces of coronary

    atherosclerosis. The Paul Dudley White Lecture 1995. Circulation

    Page 20 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    21/26

    1996;94:2013 2020.

    55. Kolodgie FD, Virmani R, Burke AP, et al. Pathologic assessment of the

    vulnerable human coronary plaque. Heart2004;90:1385 1391.

    56. Kockx MM, Cromheeke KM, Knaapen MW, et al. Phagocytosis andmacrophage activation associated with hemorrhagic microvessels in human

    atherosclerosis. Arterioscler Thromb Vasc Biol2003;23:440 446.

    57. Warner SJ, Friedman GB, Libby P. Immune interferon inhibits

    proliferation and induces 2-5-oligoadenylate synthetase gene

    expression in human vascular smooth muscle cells. J Clin Invest

    1989;83:1174 1182.

    58. Amento EP, Ehsani N, Palmer H, et al. Cytokines and growth factors

    positively and negatively regulate interstitial collagen gene expression inhuman vascular smooth muscle cells. Arterioscler Thromb

    1991;11:1223 1230.

    59. Libby P, Sukhova G, Lee RT, et al. Cytokines regulate vascular f unctions

    related to stability of the atherosclerotic plaque. J Cardiovasc Pharmacol

    1995;25[Suppl 2]:S9S12.

    60. Geng YJ, Wu Q, Muszynski M, et al. Apoptosis of vascular smooth muscle

    cells induced by in vitro stimulation with interferon-gamma, tumor necrosis

    factor-alpha, and interleukin-1 beta. Arterioscler Thromb Vasc Biol1996;16:19 27.

    61. Sukhova GK, Schonbeck U, Rabkin E, et al. Evidence for increased

    collagenolysis by interstitial collagenases-1 and -3 in vulnerable human

    atheromatous plaques. Circulation 1999;99:2503 2509.

    62. Boyle JJ, Bowyer DE, Weissberg PL, et al. Human blood-derived

    macrophages induce apoptosis in human plaque-derived vascular smooth

    muscle cells by Fas-ligand/Fas interactions. Arterioscler Thromb Vasc Biol

    2001;21:1402 1407.

    63. Bennett MR, Macdonald K, Chan SW, et al. Cooperative interactions

    between RB and p53 regulate cell pro liferation, cell senescence, and

    apoptosis in human vascular smooth muscle cells from atherosclerotic

    plaques. Circ Res 1998;82:704 712.

    64. Ross R, Wight TN, Strandness E, et al. Human atherosclerosis. I. Cell

    constitution and characteristics of advanced lesions of the superficial femoral

    artery. Am J Pathol1984;114:79 93.

    65. Bennett MR, Littlewood TD, Schwartz SM, et al. Increased sensitivity of

    Page 21 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    22/26

    human vascular smooth muscle cells from atherosclerotic plaques to p53-

    mediated apoptosis. Circ Res 1997;81:591 599.

    66. Ridker PM, Cushman M, Stampfer MJ, et al. Inflammation, aspirin, and

    the risk of cardiovascular disease in apparently healthy men. N Engl J Med

    1997;336:973 979.

    67. Ridker PM, Hennekens CH, Buring JE, et al. C-reactive protein and other

    markers of inflammation in the prediction of cardiovascular disease in

    women. N Engl J Med2000;342:836 843.

    68. Ridker PM. High-sensitivity C-reactive protein: potential adjunct for

    global risk assessment in the primary p revention of cardiovascular disease.

    Circulation 2001;103:1813 1818.

    69. Sacks FM, Ridker PM. Lipid lowering and beyond: results from the CAREstudy on lipoproteins and inflammation. Cholesterol and Recurrent Events.

    Herz1999;24:51 56.

    70. Ridker PM, Cook N. Clinical usefulness of very high and very low levels of

    C-reactive protein across the full range of Framingham Risk Scores.

    Circulation 2004;109:1955 1959.

    71. Sakkinen P, Abbott RD, Curb JD, et al. C-reactive protein and myocardial

    infarction. J Clin Epidemiol2002;55:445 451.

    72. Albert CM, Ma J, Rifai N, et al. Prospective study of C-reactive protein,

    homocysteine, and plasma lipid levels as predictors of sudden cardiac death.

    Circulation 2002;105:2595 2599.

    73. Pepys MB. CRP or not CRP? That is the question. Arterioscler Thromb

    Vasc Biol2005;25:1091 1094.

    74. Danesh J, Wheeler JG, Hirschfield GM, et al. C-reactive protein and other

    circulating markers of inflammation in the prediction of coronary heart

    disease. N Engl J Med2004;350:1387 1397.

    75. Ridker PM, Hennekens CH, Roitman-Johnson B, et al. Plasma

    concentration of soluble intercellular adhesion molecule 1 and risks of future

    myocardial infarction in apparently healthy men [see comments]. Lancet

    1998;351:88 92.

    76. Ridker PM, Buring JE, Rifai N. Soluble P-selectin and the risk of future

    cardiovascular events. Circulation 2001;103:491 495.

    77. Peter K, Weirich U, Nordt TK, et al. Soluble vascular cell adhesion

    Page 22 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    23/26

    molecule-1 (VCAM-1) as potential marker of atherosclerosis. Thromb

    Haemost1999;82[Suppl 1]:3843.

    78. Ridker PM, Rifai N, Stampfer MJ, et al. Plasma concentration of

    interleukin-6 and the risk of future myocardial infarction among apparently

    healthy men. Circulation 2000;101:1767 1772.

    79. Farb A, Burke AP, Tang AL, et al. Coronary plaque erosion without

    rupture into a lipid core. A frequent cause of coronary thrombosis in sudden

    coronary death. Circulation 1996;93:1354 1363.

    80. McNamara CA, Sarembock IJ, Bachhuber BG, et al. Thrombin and

    vascular smooth muscle cell proliferation: implications for atherosclerosis

    and restenosis. Semin Thromb Hemost1996;22:139 144.

    81. Davies MJ. Acute coronary thrombosisthe role of plaque disruptionand its initiation and prevention. Eur Heart J1995;16[Suppl L]:37.

    82. Ye S, Eriksson P, Hamsten A, et al. Progression of coronary

    atherosclerosis is associated with a common genetic variant of the human

    stromelysin-1 promoter which results in reduced gene exp ression. J Biol

    Chem 1996;271:13055 13060.

    83. Kol A, Sukhova GK, Lichtman AH, et al. Chlamydial heat shock protein 60

    localizes in human atheroma and regulates macrophage tumor necrosisfactor-alpha and matrix metalloproteinase expression. Circulation

    1998;98:300 307.

    84. Muhlestein JB, Anderson JL, Hammond EH, et al. Infection with

    Chlamydia pneumoniae accelerates the development of atherosclerosis and

    treatment with azithromycin prevents it in a rabbit model. Circulation

    1998;97:633 636.

    85. Danesh J, Whincup P, Walker M, et al. Chlamydia pneumoniae IgG titres

    and coronary heart disease: prospective study and meta-analysis [seecomments]. BMJ2000;321:208 213.

    86. Wald NJ, Law MR, Morris JK, et al. Chlamydia pneumoniae infection and

    mortality from ischaemic heart disease: large prospective study [see

    comments]. BMJ2000;321:204 207.

    87. O'Connor CM, Dunne MW, Pfeffer MA, et al. Azithromycin for the

    secondary prevention of coronary heart disease events: the WIZARD study: a

    randomized controlled trial. JAMA 2003;290:1459 1466.

    88. Cannon CP, Braunwald E, McCabe CH, et al. Intensive versus moderate

    P.12

    Page 23 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    24/26

    lipid lowering with statins after acute coronary syndromes. N Engl J Med

    2004;350:1495 1504.

    89. Zahn R, Schneider S, Frilling B, et al. Antibiotic therapy after acute

    myocardial infarction: a prospective randomized study. Circulation

    2003;107:1253 1259.

    90. Shiomi M, Ito T, Tsukada T, et al. Reduction of serum cholesterol levels

    alters lesional composition of atherosclerotic plaques. Effect of pravastatin

    sodium on atherosclerosis in mature WHHL rabb its. Arterioscler Thromb Vasc

    Biol1995;15:1938 1944.

    91. Williams JK, Sukhova GK, Herrington DM, et al. Pravastatin has

    cholesterol-lowering independent effects on the artery wall of atherosclerotic

    monkeys. J Am Coll Cardiol1998;31:684 691.

    92. Aikawa M, Rabkin E, Okada Y, et al. Lipid lowering by diet reduces matrix

    metalloproteinase activity and increases collagen content of rabbit atheroma:

    a potential mechanism of lesion stabilization [see comments]. Circulation

    1998;97:2433 2444.

    93. Pitt B, Mancini GB, Ellis SG, et al. Pravastatin Limitation of

    Atherosclerosis in the Coronary arteries (PLAC I): reduction in

    atherosclerosis progression and clinical events. PLAC I investigation. J Am

    Coll Cardiol1995;26:1133 1139.

    94. MAAS Investigators. Effect of simvastatin on coronary atheroma: the

    Multicentre Anti- Atheroma Study (MAAS). Lancet1994;344:633 638.

    95. Nissen SE, Tuzcu EM, Schoenhagen P, et al. Statin therapy, LDL

    cholesterol, C-reactive protein, and coronary artery disease. N Engl J Med

    2005;352:29 38.

    96. Sacks FM, Pfeffer MA, Moye LA, et al. The effect of pravastatin on

    coronary events after myocardial infarction in patients with average

    cholesterol levels. Cholesterol and Recurrent Events trial investigators. NEngl J Med1996;335:1001 1009.

    97. Shepherd J, Cobbe SM, Ford I, et al. Prevention of coronary heart

    disease with pravastatin in men with hypercholesterolemia. West of Scotland

    Coronary Prevention Study Group. N Engl J Med1995;333:1301 1307.

    98. 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 Study. JAMA 1998;279:1615 1622.

    Page 24 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    25/26

    99. 4S Study. Randomised trial of cholesterol lowering in 4444 patients with

    coronary heart disease: the Scandinavian Simvastatin Survival Study (4S).

    Lancet1994;344:1383 1389.

    100. Jialal I, Stein D, Balis D, et al. Effect of hydroxymethyl glutaryl

    coenzyme a reductase inhibitor therapy on high sensitive C-reactive protein

    levels. Circulation 2001;103:1933 1935.

    101. Ridker PM, Rifai N, Pfeffer MA, et al. Inflammation, pravastatin, and the

    risk of coronary events after myocardial infarction in patients with average

    cholesterol levels. Cholesterol and Recurrent Events (CARE) investigators.

    Circulation 1998;98:839 844.

    102. Crisby M, Nordin-Fredriksson G, Shah PK, et al. Pravastatin treatment

    increases collagen content and decreases lipid content, inflammation,

    metalloproteinases, and cell death in human carotid plaques: implications forplaque stabilization. Circulation 2001;103:926 933.

    103. Katznelson S, Wang XM, Chia D, et al. The inhibitory effects of

    pravastatin on natural killer cell activity in vivo and on cytotoxic T

    lymphocyte activity in vitro. J Heart Lung Transplant1998;17:335 340.

    104. Lacoste L, Lam JY, Hung J, et al. Hyperlipidemia and coronary disease.

    Correction of the increased thrombogenic potential with cholesterol reduction

    [see comments]. Circulation 1995;92:3172 3177.

    105. Negre-Aminou P, van Vliet AK, van Erck M, et al. Inhibition of

    proliferation of human smooth muscle cells by various HMG-CoA reductase

    inhibitors; comparison with other human cell types. Biochim Biophys Acta

    1997;1345:259 268.

    106. Rosenson RS, Tangney CC. Antiatherothrombotic properties of statins:

    implications for cardiovascular event reduction [see comments]. JAMA

    1998;279:1643 1650.

    107. Treasure CB, Klein JL, Weintraub WS, et al. Beneficial effects of

    cholesterol-lowering therapy on the coronary endothelium in patients with

    coronary artery disease. N Engl J Med1995;332:481 487.

    108. Sparrow CP, Burton CA, Hernandez M , et al. Simvastatin has anti-

    inflammatory and antiatherosclerotic activities independent of plasma

    cholesterol lowering. Arterioscler Thromb Vasc Biol2001;21:115 121.

    109. Kwak B, Mulhaupt F, Myit S, et al. Statins as a newly recognized type of

    immunomodulator.Nat Med

    2000;6:1399

    1402.

    Page 25 of 26Ovid: Textbook of Cardiovascular Medicine

    12/05/2009mk:@MSITStore:M:\Topol%20Textbook%20of%20CardioVascular%20medicine.C...

  • 8/14/2019 AtheroSclerotic Biology and Epidemiology of Disease

    26/26

    110. Schonbeck U, Libby P. Inflammation, immunity, and HMG-CoA reductase

    inhibitors: statins as antiinflammatory agents? Circulation 2004;109:II18-

    II26.

    111. Mukherjee D, Nissen SE, Topol EJ. Risk of cardiovascular events

    associated with selective COX-2 inhibitors. JAMA 2001;286:954 959.

    112. Sousa JE, Costa MA, Abizaid A, et al. Four-year angiographic and

    intravascular ultrasound follow-up of pat ients treated with sirolimus-eluting

    stents. Circulation 2005;111:2326 2329.

    113. Babapulle MN, Joseph L, Belisle P, et al. A hierarchical Bayesian meta-

    analysis of randomised clinical trials of drug-eluting stents. Lancet2004;

    364:583 591.

    114. Assmann G, Gotto AM, Jr. HDL cholesterol and protective factors in

    atherosclerosis. Circulation 2004;109:III8 III14.

    115. Meier B. Plaque sealing by coronary angioplasty. Heart

    2004;90:1395 1398.

    116. Spratt JC, Camenzind E. Plaque stabilisation by systemic and local drug

    administration. Heart2004;90:1392 1394.

    117. Davies JR, Rudd JH, Weissberg PL. Molecular and metabolic imaging of

    atherosclerosis. J Nucl Med2004;45:1898 1907.

    118. Rudd JHF, Warburton EA, Fryer TD, et al. Imaging atherosclerotic

    plaque inflammation with [18F]-fluorodeoxyglucose positron emission

    tomography. Circulation 2002;105:2708 2711.

    Page 26 of 26Ovid: Textbook of Cardiovascular Medicine