restenosis following implantation of bare metal coronary stents: pathophysiology and pathways...

19
Restenosis following implantation of bare metal coronary stents: Pathophysiology and pathways involved in the vascular response to injury B Neal A. Scott * Camino Cardiovascular Associates, 525 South Drive, Suite 107, Mountain View, CA 94040, USA Received 17 February 2005; accepted 31 January 2006 Available online 6 March 2006 Abstract This review summarizes the restenotic process that occurs after the implantation of bare metal coronary stents. The pathology of in-stent restenosis is distinct from that seen after balloon angioplasty and is characterized by neointimal proliferation and extracellular matrix deposition. The degree of neointimal proliferation is proportional to the amount of injury, the intensity of the inflammatory infiltrate and the association of stent struts with lipid-filled plaque. In-stent restenosis also appears to be associated with systemic markers of inflammation. Shear stress has an important influence on restenosis as does the presence and adhesiveness of vascular progenitor cells. Clinical predictors (e.g., artery size, stent length, diabetes, and gender) may affect the incidence of restenosis seen after stent placement. A number of catheter-based interventions have been used to treat in-stent restenosis. Although preliminary data suggest that the use of drug-eluting stents may be effective, only intracoronary radiation has shown consistent efficacy in randomized trials. D 2006 Elsevier B.V. All rights reserved. Keywords: In-stent restenosis; Pathology; Bare metal stents; Vascular responses; Vascular injury Contents 1. Introduction .................................................... 359 2. Pathology of in-stent restenosis .......................................... 360 3. Is the response to stenting similar to wound healing? Role of the extracellular matrix .............. 361 4. Mechanism of restenosis after balloon angioplasty ................................ 361 0169-409X/$ - see front matter D 2006 Elsevier B.V. All rights reserved. doi:10.1016/j.addr.2006.01.015 B This review is part of the Advanced Drug Delivery Reviews theme issue on bDrug-Eluting Stents: an Innovative Multidisciplinary Drug Delivery PlatformQ, Vol. 58/3, 2006. * Tel.: +1 650 961 7021; fax: +1 650 969 8679. E-mail address: [email protected]. Advanced Drug Delivery Reviews 58 (2006) 358 – 376 www.elsevier.com/locate/addr

Upload: neal-a-scott

Post on 25-Aug-2016

214 views

Category:

Documents


1 download

TRANSCRIPT

  • ry stents:

    ular

    gender) may affect the incidence of restenosis seen after stent placement. A number of catheter-based interventions have been

    e Multidisciplinary Drug

    Advanced Drug Delivery Reviews 58 (2006) 358376

    www.elsevier.com/locate/addrB This review is part of the Advanced Drug Delivery Reviews theused to treat in-stent restenosis. Although preliminary data suggest that the use of drug-eluting stents may be effective, only

    intracoronary radiation has shown consistent efficacy in randomized trials.

    D 2006 Elsevier B.V. All rights reserved.

    Keywords: In-stent restenosis; Pathology; Bare metal stents; Vascular responses; Vascular injury

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 359

    2. Pathology of in-stent restenosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 360

    3. Is the response to stenting similar to wound healing? Role of the extracellular matrix . . . . . . . . . . . . . . 361

    4. Mechanism of restenosis after balloon angioplasty . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 361response to injuryB

    Neal A. Scott *

    Camino Cardiovascular Associates, 525 South Drive, Suite 107, Mountain View, CA 94040, USA

    Received 17 February 2005; accepted 31 January 2006

    Available online 6 March 2006

    Abstract

    This review summarizes the restenotic process that occurs after the implantation of bare metal coronary stents. The

    pathology of in-stent restenosis is distinct from that seen after balloon angioplasty and is characterized by neointimal

    proliferation and extracellular matrix deposition. The degree of neointimal proliferation is proportional to the amount of injury,

    the intensity of the inflammatory infiltrate and the association of stent struts with lipid-filled plaque. In-stent restenosis also

    appears to be associated with systemic markers of inflammation. Shear stress has an important influence on restenosis as does

    the presence and adhesiveness of vascular progenitor cells. Clinical predictors (e.g., artery size, stent length, diabetes, andRestenosis following implantation of bare metal corona

    Pathophysiology and pathways involved in the vasc0169-409X/$ - s

    doi:10.1016/j.ad

    Delivery Platfor

    * Tel.: +1 650

    E-mail addreme issue on bDrug-Eluting Stents: an InnovativmQ, Vol. 58/3, 2006.ee front matter D 2006 Elsevier B.V. All rights reserved.

    dr.2006.01.015

    961 7021; fax: +1 650 969 8679.

    ss: [email protected].

  • . . .

    . . .

    ular

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    . . .

    n in 1987 when

    lacement of 24

    y arteries of 19

    ary-artery reste-

    =4) after coro-

    gioplasty. They

    the group with

    occlusion of a

    sure, a second

    uccessfully with

    d after bypass

    ound occlusion.

    ithout evidence

    he stented seg-

    heir preliminary

    may effectively

    fter angioplasty,

    be required to

    eir preliminary

    se of coronary

    eliverThe catheter-based treatment of obstructive coro-

    nary atherosclerosis was pioneered by Andreas

    Gruentzig in the late 1970s when he and Senning

    described the procedure they named percutaneous

    transluminal coronary angioplasty. They reported that

    6 of the 32 patients (19%) who had successful

    angioplasty suffered restenosis, or re-narrowing of

    the vessel, several months after the initial procedure

    [1]. Subsequent registry studies of large numbers of

    coronary angioplasty patients documented a restenosis

    rate closer to 33% [2]. One of the first pathological

    descriptions of restenosis after coronary angioplasty

    was published by Essed et al. [3]., who described a

    proliferative fibrocellular response that occluded the

    coronary lumen. This report was later followed by

    additional case reports confirming the fibrocellular

    response to coronary angioplasty [4,5] that has

    characterized restenosis. Since the early days of

    coronary angioplasty, a myriad of devices (stents,

    atherectomy, laser, rotablator, etc.) was developed

    The era of coronary stenting bega

    Sigwart et al. [9] described the p

    self-expanding stents in the coronar

    patients who presented with coron

    noses (n =17) or abrupt closure (n

    nary or coronary-bypass graft an

    observed three complications in

    coronary disease. One thrombotic

    stent resulted in asymptomatic clo

    acute thrombosis was managed s

    thrombolysis, and one patient die

    surgery for a suspected but unf

    Follow-up continued for 9 months w

    of any further restenoses within t

    ments. The authors noted that t

    experience suggested that stents

    prevent occlusion and restenosis a

    and that long-term follow-up would

    validate the early success of th

    results. The widespread elective u1. Introduction to show any significant reduction in coronary reste-

    nosis is the stent [68].5. Mechanism of in-stent restenosis . . . . . . . . . . .

    6. Inflammation . . . . . . . . . . . . . . . . . . . . .

    6.1. Inflammatory mechanisms involved in the vasc

    6.2. Circulating monocytes . . . . . . . . . . . . .

    6.3. Mac-1 receptor. . . . . . . . . . . . . . . . .

    6.4. Monocyte chemoattractant protein (MCP-1) . .

    6.5. C-reactive protein . . . . . . . . . . . . . . .

    7. Shear stress . . . . . . . . . . . . . . . . . . . . . .

    8. Stent endothelialization . . . . . . . . . . . . . . . .

    9. Progenitor cells . . . . . . . . . . . . . . . . . . . .

    10. Clinical predictors of in-stent restenosis . . . . . . .

    10.1. Procedural characteristics . . . . . . . . . . .

    10.2. Diabetes . . . . . . . . . . . . . . . . . . .

    10.3. Angiotensin converting enzyme . . . . . . .

    10.4. Gender . . . . . . . . . . . . . . . . . . . .

    11. Angiographic patterns of in-stent restenosis . . . . .

    12. Treatment of in-stent restenosis . . . . . . . . . . . .

    12.1. Radiation . . . . . . . . . . . . . . . . . . .

    12.2. Rotational atherectomy . . . . . . . . . . . .

    12.3. Cutting balloon . . . . . . . . . . . . . . . .

    12.4. Laser angioplasty . . . . . . . . . . . . . . .

    12.5. Drug-eluting stents . . . . . . . . . . . . . .

    References. . . . . . . . . . . . . . . . . . . . . . . . . .

    N.A. Scott / Advanced Drug Dwith the primary goal of decreasing the incidence of

    restenosis after coronary angioplasty. The only device. . . . . . . . . . . . . . . . . . . . . . . . . . . 362

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 362

    response to injury . . . . . . . . . . . . . . . . . . 362

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 363

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 363

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 364

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 364

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 365

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 367

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 367

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 368

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 368

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 368

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 369

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 369

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 369

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 370

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 370

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 371

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 371

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 371

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 371

    . . . . . . . . . . . . . . . . . . . . . . . . . . . 371

    y Reviews 58 (2006) 358376 359stents occurred after they were shown to provide a

    significant decrease in restenosis over elective

  • tion, morphological characteristics of the lesion, stent

    design and method of analysis.

    eliver2. Pathology of in-stent restenosis

    Despite the very large number of coronary stents

    used in patients over the past decade, there have been

    relatively few pathologic studies of the course of these

    devices after implantation. Komatsu et al. published

    the first systematic examination of the neointimal

    tissue response to stenting in 11 patients who died

    between 2 days and 21 months after stenting. The four

    patients who died within 12 days had occlusive

    thrombus formation while the patients who died after

    64 days had a distinct layer of neointima, albeit to

    varying degrees. In non-restenotic lesions, neointimal

    thickening was markedly less than in restenotic

    lesions but without qualitative differences; the neo-

    intima contained macrophages but was composed

    predominantly of alpha-actin positive smooth muscle

    cells [12].

    In a more extensive study, Farb et al. [13] made

    histological observations on 55 stents in 35 coronary

    vessels (32 native arteries and 3 vein grafts) from 32balloon angioplasty in two randomized trials.

    Restenosis occurred in 3242% of patients treated

    with balloon angioplasty and 2232% of patients

    treated with stents [7,8].

    The major complications associated with coronary

    stent placement are stent thrombosis and restenosis.

    Stent thrombosis is a life-threatening condition that

    involves the occlusion of the stent by thrombus. This

    is rarely seen at the time of stent placement and more

    commonly occurs within 130 days after the proce-

    dure. This condition is usually associated with

    myocardial infarction and commonly requires emer-

    gent angioplasty or bypass surgery. The use of

    improved stent delivery techniques involving high

    deployment pressures to fully appose the stent to the

    vessel wall, accompanied by the administration of

    anti-platelet agents has decreased the incidence of this

    complication to less than 1% [10,11]. Restenosis is the

    most common complication associated with coronary

    stent placement. The incidence of restenosis is

    influenced by many factors including patient selec-

    N.A. Scott / Advanced Drug D360patients. They examined plaquestent interaction,

    thrombus formation, inflammation, and the presenceof a neointima. The mean duration of stent placement

    was 39F82 days. Platelet-rich thrombi were acharacteristic of early lesions. Fibrin-rich thrombi

    were commonly seen around stent struts, especially

    early after stenting.

    Acute inflammatory cells (neutrophils) associated

    with stent struts were present more frequently in stents

    implanted for V3 days and were related to theunderlying arterial wall morphology. There was more

    of an inflammatory cellular response when the stent

    struts were embedded in damaged media or a lipid

    core when compared to fibrous plaque.

    A neointima consisting of spindle-shaped mesen-

    chymal cells (a-actin positive smooth muscle cells)within a proteoglycan matrix associated with stents

    struts was not present in any of the 18 patients with

    b11 days implant duration, however, this finding waspresent in 45% of patients who died at 1230 days

    after stent placement and 100% of patients at N30days. In stents implanted for N30 days, neointimalthickness at stent strut sites was greater when medial

    damage (medial laceration or rupture) was present

    than when struts were in contact with plaque or intact

    media. Based on their findings, the authors concluded

    that the morphological changes following coronary

    stenting involve early thrombus formation and an

    acute inflammatory response followed by neointimal

    growth. Medial injury and lipid core penetration by

    struts resulted in an increased inflammatory response.

    The relationship between inflammation and neo-

    intimal formation was further examined by these

    authors in a subsequent study [14]. Detailed histology

    was performed on 116 stents implanted N90 days in87 coronary arteries from 56 patients. The mean

    duration of stent implant was 10 months. In-stent

    restenosis was defined as a stent area stenosis of

    N75%. The inflammatory infiltrates associated withstenting were composed predominantly of macro-

    phages with smaller numbers of T cells and rare B

    cells. Neointimal inflammatory cell density correlated

    with increased neointimal thickness, and the mean

    number of inflammatory cells was 2.4-fold higher in

    restenosis versus no restenosis cases. Peri-strut in-

    flammatory cell density was increased in regions of

    medial disruption compared with struts in contact with

    fibrous plaque or an intact fibrous cap. The percent of

    y Reviews 58 (2006) 358376the neointima occupied by macrophages was 3-fold

    higher in restenosis versus no-restenosis cases. Lipid

  • a granulation tissue phase (macrophage infiltration,

    myofibroblast in-growth and angiogenesis). The

    reduced staining seen at later time points. Type I

    collagen staining was weakest early after stent

    eliverearly extracellular matrix consists of fibrin, fibro-

    nectin, hyaluronan, and versican. Hyaluronan pro-

    vides the matrix into which mesenchymal cells

    migrate, promotes cell proliferation, and supplies

    feedback regulation of growth factor synthesis.

    Versican binds to hyaluronan and affords viscoelas-

    ticity to healing tissues. Over time there is progres-

    sive hyaluronan degradation, reabsorption of a

    portion of the type III collagen, and synthesis of

    type I collagen, decorin, and biglycan. These later

    changes, along with the cross-linking of type I

    collagen, are associated with wound contraction. In

    uncomplicated dermal wounds, the healing is usually

    complete by 2 weeks [16].

    When postmortem human coronary arteries con-

    taining stents underwent histological assessment of

    neointimal proteoglycans, hyaluronan, collagen (typescore penetration was associated with an increased

    neointimal thickness and increased numbers of

    inflammatory cells compared with struts in contact

    with fibrous plaque.

    Similar findings were noted in an analysis of

    saphenous vein bypass grafts [15]. Segments were

    surgically retrieved from 10 patients (21 stents) at 3

    days to 10 months after implantation of a self-

    expanding Wallstent. Early observations revealed that

    large amounts of platelets and leukocytes adhered to

    the stent wires during the first few days. At 3 months,

    the wires were embedded in a layered new intimal

    thickening, consisting of smooth muscle cells and

    collagenous matrix. In addition, foam cells were

    abundant near the wires. Abnormal adherence of

    leukocytes was noted as late as 10 months after

    implantation.

    3. Is the response to stenting similar to wound

    healing? Role of the extracellular matrix

    Similarities have been found between the cellular

    and molecular mechanisms involved in stent reste-

    nosis and wound healing. In wound healing, best

    characterized by dermal injury responses, a throm-

    botic and acute inflammatory reaction is followed by

    N.A. Scott / Advanced Drug DI and III), smooth muscle cells, and CD44 (a cell

    surface receptor for hyaluronan), neointimal versicanplacement, with progressively stronger staining with

    time. Smooth muscle cell density and stent stenosis

    were significantly reduced in stents implanted for over

    18 months. CD44 staining co-localized with macro-

    phages and was associated with increased neointimal

    thickness. These findings suggested that the formation

    of the extracellular matrix within human coronary

    stents resembles that seen with wound healing. An

    important finding of this study was that the response

    to a coronary stent does not show complete healing

    until 18 months after deployment [17].

    In a study of atherectomy samples taken from

    patients with in-stent restenosis, Chung et al. found

    that the cellularity of the neointima decreased in time

    after stenting. Early lesions (b3 months) were hyper-cellular, while late lesions (N3 months) were hypo-cellular. The cells seen were predominantly smooth

    muscle cells, however, inflammatory cells were also

    observed. Of the cell-depleted areas seen in the late

    lesions, the extracellular matrix was composed of

    either collagen-rich or collagen-poor areas. The

    collagen-rich areas also stained for biglycan, while

    the collagen-poor areas stained for versican and

    hyaluronan [18]. These data suggest that enhanced

    extracellular matrix rather than cellular proliferation

    contribute to the later stages of in-stent restenosis.

    4. Mechanism of restenosis after balloon

    angioplasty

    Three factors play a major role in restenosis after

    balloon angioplasty: recoil, neointimal formation and

    chronic remodeling. Recoil is defined as the elastic

    response that occurs after overstretch of the vessel

    and is usually seen immediately after balloonand hyaluronan staining was strongly positive, co-

    localized with alpha-actin-positive smooth muscle

    cells, and was greater in intensity in stents implanted

    for less than 18 months when compared to stents

    implanted for over 18 months. Conversely, decorin

    staining was strongest in stents implanted for over 18

    months. The neointima of stents implanted for less

    than 18 months was rich in type III collagen, with

    y Reviews 58 (2006) 358376 361deflation. Neointimal formation has a time course

    that spans over several weeks to months. The

  • postulated. In animal models, chronic remodeling

    appears to be the major component of restenosis

    molecules that bind ligands on leukocytes. Selectins

    mediate the initial attachment of platelets and the

    eliverafter balloon angioplasty [20,21]. Serial ultrasound

    studies performed in patients immediately after

    angioplasty and 6 months later confirmed the

    animals studies, showing that (1) a decrease in total

    arterial cross-sectional area accounted for 70% to

    75% of late lumen loss and (2) late lumen loss

    correlated better with a decrease in the cross-

    sectional area of the external elastic lamina than

    with an increase in the plaque plus medial area

    (neointimal proliferation) [22].

    5. Mechanism of in-stent restenosis

    A similar serial intravascular ultrasound study

    performed on patients with stents found that in stented

    segments, late lumen area loss correlated strongly

    with tissue growth but only weakly with remodeling.

    The authors concluded that since stents appear to

    withstand the extrinsic compression of the artery from

    arterial remodeling (the dominant mechanism of late

    lumen loss in nonstented lesions) that in-stent

    restenosis was the result of neointimal tissue forma-

    tion (i.e., cellular proliferation and accumulation of

    extracellular matrix) [23].

    The existence of positive, peri-stent remodeling

    was initially suggested by Hoffman et al. who

    showed that stents induce proliferation both withinneointima is formed by cells and extracellular

    matrix. It appears that the source of most of these

    cells may be the adventitia, since intense prolifera-

    tion occurs there 2 to 3 days after balloon injury in

    porcine coronaries. When these cells are labeled,

    they are found in the neointima several weeks later.

    Seven days after injury, some proliferation is noted

    in the medial layer [19]. The third component of

    restenosis is chronic remodeling. Chronic remodeling

    is due to extrinsic compression of the vessel, best

    characterized by a decrease in the outer circumfer-

    ence of the vessel, the external elastic lamina. The

    cause of the chronic remodeling is likely fibrosis

    although other causes such as changes in the

    extracellular matrix composition and structure, and

    chronic changes in vascular tone have also been

    N.A. Scott / Advanced Drug D362the endoluminal surface of the stent and in the tissue

    layers outside of the stent. They also showed thatrolling interaction of leukocytes with the luminal

    endothelium. The integrin class of adhesion molecules

    mediates the firm adhesion and transendothelial

    migration. The beta-2 integrin molecule Mac-1

    (CD11b/CD18) is present on neutrophils and mono-

    cytes and appears to be of central importance in

    leukocyte recruitment after vascular injury. In additionthe tissue increase outside of the stent was accom-

    panied by positive remodeling (increased cross-

    sectional area of the external elastic lamina) [24].

    Nakamura et al. later found that the peri-stent

    positive remodeling occurs to a variable extent and

    is inversely correlated with the degree of neointimal

    formation [25].

    6. Inflammation

    Several recent reviews have summarized the data

    suggesting an important relationship between inflam-

    mation and stent restenosis [26,27]. As noted above,

    analyses of human restenotic tissue from autopsy

    studies identified an inflammatory component in the

    arterial response to stent placement [13,12] suggesting

    a strong link between the extent of medial damage,

    inflammation, and restenosis.

    In animal models, a brisk early inflammatory

    response was produced after balloon injury or stent

    placement with abundant surface-adherent leukocytes

    of monocyte and granulocyte lineage [28,29]. Days

    and weeks later, macrophages invaded the forming

    neointima and were observed clustering around the

    struts, forming giant cells. Blockade of early mono-

    cyte recruitment with anti-inflammatory agents

    resulted in reduced late neointimal thickening

    [30,31]. Rogers et al. have demonstrated a linear

    relationship between tissue monocyte content and

    neointimal area, suggesting a causal role for mono-

    cytes in restenosis [29].

    6.1. Inflammatory mechanisms involved in the

    vascular response to injury

    Upon injury, endothelial cells express adhesion

    y Reviews 58 (2006) 358376to promoting the accumulation of leukocytes at sites

    of vascular injury and the binding of platelets to

  • eliverneutrophils, Mac-1 amplifies the inflammatory re-

    sponse by inducing neutrophil activation, upregulat-

    ing cell adhesion molecule expression, and generating

    signals that promote integrin activation and chemo-

    kine synthesis. Proinflammatory cytokines provide a

    chemotactic stimulus to the adherent leukocytes,

    directing their migration into the intima. Recent

    research has identified candidate chemoattractant

    molecules responsible for this transmigration. For

    example, monocyte chemoattractant protein-1 (MCP-

    1) appears responsible for the direct migration of

    monocytes into the intima at sites of lesion formation.

    In addition to MCP-1, macrophage colony-stimu-

    lating factor (M-CSF) contributes to the differentia-

    tion of the blood monocyte into the macrophage foam

    cell. Inflammatory mediators such as M-CSF augment

    expression of macrophage scavenger receptors lead-

    ing to the formation of lipid-laden macrophages. M-

    CSF and other mediators can promote the replication

    of macrophages within the intima as well. T lympho-

    cytes also join macrophages in the intima during

    lesion development. T cells encounter signals that

    cause them to elaborate inflammatory cytokines such

    as g-interferon and lymphotoxin (tumor necrosisfactor [TNF]-h) that in turn can stimulate macro-phages as well as vascular endothelial cells and

    smooth muscle cells. These leukocytes, as well as

    resident vascular wall cells, secrete cytokines and

    growth factors (such as platelet-derived growth factor,

    basic fibroblast growth factor and epidermal growth

    factor) that can promote the migration and prolifera-

    tion of smooth muscle cells. Medial smooth muscle

    cells express specialized enzymes that can degrade

    elastin and collagen in response to inflammatory

    stimulation. This degradation of the arterial extracel-

    lular matrix permits the penetration of the smooth

    muscle cells through the elastic laminae and collag-

    enous matrix of the injured artery.

    6.2. Circulating monocytes

    Fukuda et al. examined the circulating monocyte

    count from peripheral blood samples taken immedi-

    ately before stent implantation and daily for 7 days

    after the intervention in 107 patients. All patients

    underwent angiography and volumetric intravascular

    N.A. Scott / Advanced Drug Dultrasound analysis 6 months later. They found that

    the circulating monocyte count increased and reachedits peak 2 days after stent placement. The maximum

    monocyte count after stent implantation showed a

    significant positive correlation with neointimal stent

    volume at 6-month follow-up. Angiographic resteno-

    sis was observed in 22 patients and these patients had

    a significantly higher maximum monocyte count than

    patients without restenosis [32].

    6.3. Mac-1 receptor

    Leukocyte adhesion to injured arteries may occur

    through a variety of selectin- and integrin-dependent

    mechanisms involving platelets and extracellular

    matrix proteins. In particular, leukocyte recruitment

    to areas of extravascular inflammation is mediated by

    the b2-integrin family of receptors. Of these receptors,

    Mac-1 (CD11b/CD18/aLb2) is most commonly asso-

    ciated with restenosis.

    The multivalent binding properties of Mac-1 make

    this receptor uniquely poised to regulate adhesive and

    inflammatory processes after vascular injury. Mac-1 is

    capable of binding fibrinogen, intercellular adhesion

    molecule-1, and factor X, ligands that are abundant in

    the injured wall. Mac-1 is, in fact, the primary

    fibrinogen receptor on leukocytes, facilitating the

    adhesion and transmigration of neutrophils and

    monocytes at sites of fibrin and platelet deposition.

    Recent clinical reports have implicated up-regulation

    of Mac-1 with restenosis after coronary angioplasty

    [3335].

    When careful analysis of the transcardiac gradient

    (coronary sinus blood minus the value of peripheral

    blood) of CD11b (alpha-subunit of Mac-1) was

    compared in patients who underwent coronary angio-

    plasty or stenting, there were profound differences

    between the two procedures. The gradient for neutro-

    phil surface expression of CD11b increased 48 h after

    coronary stenting, but this change showed less

    significance 48 h after balloon angioplasty alone.

    The gradient 48 h after the procedures for CD11b was

    independently correlated with restenosis in both the

    stent and angioplasty groups [36].

    Inoue et al. examined the expression of CD11b and

    binding of a monoclonal antibody against an activa-

    tion-dependent neo-epitope of Mac-1 (8B2) on the

    surface of polymorphonuclear leukocytes in 62

    y Reviews 58 (2006) 358376 363patients undergoing coronary stenting. Transcardiac

    CD11b expression increased significantly at 24 h and

  • elivermaximally at 48 h after stenting; 8B2 began to

    increase at 10 min and was maximally increased at

    48 h after stenting. These changes were more

    prominent in patients with subsequent restenosis.

    Multiple regression analysis showed that the late

    lumen loss by quantitative coronary angiographic

    analysis was independently correlated with the

    CD11b increase and the 8B2 increase 48 h after the

    procedure. Mac-1 activation, as assessed by 8B2

    binding, was the most powerful predictor of late

    lumen loss [37].

    Rogers et al. [30] administered an antibody

    directed against CD11b to rabbits immediately before,

    and every 48 h after balloon or stent injury to the iliac

    vessels. They found a marked decrease in neointimal

    growth in the animals treated with the specific

    antibody, suggesting that leukocyte recruitment and

    infiltration is an important component of the neo-

    intimal response to balloon and stent injury.

    6.4. Monocyte chemoattractant protein (MCP-1)

    Chemokines are a group of chemoattractant cyto-

    kines produced by a number of somatic cells, including

    endothelial cells, smooth muscle cells and leukocytes.

    They include monocyte chemoattractant protein

    (MCP-1), and interleukin (IL)-8, both of which recruit

    leukocytes to areas of vascular injury. MCP-1 is the

    prototype of the C-C chemokine-beta subfamily and

    exhibits its most potent chemotactic activity toward

    monocytes and T lymphocytes. In addition to promot-

    ing the transmigration of circulating monocytes into

    tissues, MCP-1 exerts various other effects on mono-

    cytes, including superoxide anion induction, cytokine

    production and adhesion molecule expression. MCP-1

    expression is induced by inflammatory cytokines,

    peptide growth factors, endothelial cells or vascular

    smooth muscle cells. Since elevated levels of MCP-1

    have been demonstrated in myocardial infarction, heart

    failure and after angioplasty [38], this chemokine is

    probably a key factor in the initiation of the inflam-

    matory process and maintaining the proliferative

    response to vascular injury.

    After balloon angioplasty, plasma levels of MCP-1

    increase and remain elevated in those patients who

    develop restenosis [38]. Similarly, following stent

    N.A. Scott / Advanced Drug D364placement, MCP-1 levels in plasma increase after

    several days and are more likely to be elevated atfollow-up 6 months later in patients who have

    restenosis [39]. When balloon overstretch injury and

    stent placement are compared, stent implantation is

    associated with a more intense acute and chronic, low-

    grade inflammatory response [40]. In balloon-injured

    arteries, leukocyte recruitment was confined to early

    neutrophil infiltration. IL-8 and MCP-1 mRNA levels

    peaked within hours and were undetectable at 14 days.

    In contrast, in stented arteries, early neutrophil

    recruitment was followed by prolonged macrophage

    accumulation. IL-8 and MCP-1 mRNA levels peaked

    within hours but were still detectable 14 days after

    injury. In contrast to balloon injury, stent-induced

    injury results in sustained chemokine expression and

    leukocyte recruitment [41].

    In animals, Horvath et al. used antibodies directed

    against neutrophils or monocytes to determine the role

    played by each cell type after either balloon angio-

    plasty or stenting in a primate iliac model. They found

    that monocyte-specific blockade achieved via block-

    ade of the MCP-1 receptor was effective at reducing

    neointimal hyperplasia after stenting. In contrast,

    combined neutrophil and monocyte blockade

    achieved by targeting the leukocyte beta (2)-integrin

    beta-subunit CD18 was required to reduce neointimal

    hyperplasia after balloon injury [42]. These studies

    suggest that monocytes, and not polymorphonuclear

    leukocytes, may play the more important role in stent

    restenosis.

    Ohtani et al. [43] recently devised a new strategy

    for anti-MCP-1 gene therapy by transfecting an N-

    terminal deletion mutant of the MCP-1 gene into

    skeletal muscles. They used this strategy to investigate

    the role of MCP-1 in experimental in-stent restenosis

    in hypercholesterolemic rabbits and monkeys. Trans-

    fection of the mutant MCP-1 gene suppressed

    monocyte infiltration and activation in the stented

    arterial wall and markedly reduced the development

    of neointimal hyperplasia. This strategy also sup-

    pressed local expression of MCP-1 and inflammatory

    cytokines. They concluded that inhibition of MCP-1-

    mediated inflammation is effective in reducing exper-

    imental in-stent restenosis.

    6.5. C-reactive protein

    y Reviews 58 (2006) 358376Formerly considered solely as a biomarker for

    inflammation, C-reactive protein (CRP) is now

  • Activation of various signaling cascades and

    transcription factor systems, as well as the identifica-

    eliverviewed as a prominent component of the vascular

    inflammatory process. CRP is a protein that binds to

    the C-polysaccharide of the pneumococcal cell wall. It

    is part of the innate immunity that activates the

    classical complement pathway after aggregation or

    binding to ligands. CRP also binds to phospholipids

    of damaged cells, with subsequent limited activation

    of the complement system and enhanced uptake of

    these cells by macrophages.

    CRP also induces the secretion of interleukin-6 and

    endothelin-1 and decreases the expression and bio-

    availability of endothelial nitric oxide synthase in

    human endothelial cells. In addition, CRP activates

    macrophages to express cytokine and tissue factor and

    enhances the uptake of LDL. CRP also amplifies the

    proinflammatory effects of several other mediators,

    including endotoxin.

    Immunohistochemical staining for CRP on athe-

    rectomy samples obtained from patients who under-

    went directional coronary atherectomy or stenting as

    their initial procedure demonstrated more staining for

    CRP and macrophages in patients with in-stent

    restenosis when compared to patients with restenosis

    after atherectomy [44].

    Although there have been a number of negative

    and positive studies on the association between

    plasma levels of CRP and restenosis, the study by

    Versaci et al. strongly suggests a link between

    elevated CRP levels and stent restenosis. The inves-

    tigators enrolled 83 patients who underwent success-

    ful stenting and had plasma CRP levels that were

    elevated (N0.5 mg/dl) 72 h after the procedure. Thepatients were randomized to treatment with oral

    prednisone or placebo for 45 days. Six-month

    restenosis rates were lower in the prednisone group

    (7% vs. 33%). The prednisone group also had a

    better event free survival rate 1 year after the

    procedure (93% vs. 65%) [45]. These data suggest

    that there is a subpopulation of patients that can be

    identified by systemic markers of inflammation and

    that when these patients are treated with agents that

    diminish the inflammatory process, restenosis can be

    inhibited. Although other studies failed to demon-

    strate a beneficial effect of corticosteroid adminis-

    tration on restenosis, most of these studies were

    performed on patients who underwent balloon

    N.A. Scott / Advanced Drug Dangioplasty rather than stenting. In addition, either

    a single corticosteroid dose or a much shorter coursetion of shear-stress-response elements in the promoters

    of several genes relevant to both atherosclerotic and

    restenotic processes (e.g., platelet-derived growth

    factors A and B, macrophage chemoattractant pro-

    tein-1, and vascular cell adhesion molecule-1 have

    helped to provide insight into the cellular mechanismsof treatment was used. Also, no prior attempt was

    made to target patients with high inflammatory

    markers for treatment. A study by Walter et al.

    [46] found that administration statins to patients with

    elevated CRP levels could also decrease restenosis

    rates.

    7. Shear stress

    Neointimal hyperplasia resulting in restenosis can

    be observed within any discreet location of the stented

    segment or can appear in a diffuse pattern. Although

    certain systemic characteristics (e.g., the presence of

    diabetes) or anatomical variables (small vessel diam-

    eter, long lesion length) increase the probability of

    restenosis, the presence or absence of these factors

    does not explain the specific location of a site of

    neointimal proliferation within a stent or its discrete or

    diffuse pattern.

    In a study that examined the role of blood flow in

    the progression of atherosclerosis, Glagov et al.

    determined that alterations in shear stress could cause

    important compensatory changes in both luminal and

    vessel diameter [47]. Biomechanical forces such as

    fluid shear stresses stimulate the production in

    endothelium of a large and diverse array of potent

    biological mediators [48,49]. Some of these agents

    involve gene regulation at the transcriptional level and

    thus are analogous to endothelial activation by

    humeral factors. The endothelial cell appears capable

    of responding not only to the magnitude of the applied

    forces but also to their temporal and spatial fluctua-

    tions (e.g., steady versus pulsatile flow, uniform

    laminar, disturbed laminar, or turbulent flow regions),

    thus suggesting the existence of primary flow sensors

    (receptors) that are coupled via distinct signaling

    pathways to nuclear events [50,49].

    y Reviews 58 (2006) 358376 365linking shear stress stimuli and genetic regulatory

    events [51,49,48].

  • eliverCultured human endothelial cells exposed to two

    well defined biomechanical stimulia steady laminar

    shear stress and a turbulent shear stress of equivalent

    spatial and temporal average intensity revealed

    distinctive patterns of up- and down-regulation

    associated with each type of stimulus in many of the

    11,397 unique genes examined [51]. Different cyto-

    skeletal morphologies were also observed in the cells

    exposed to the two types of shear stress. Thus,

    endothelial cells have the capacity to discriminate

    among specific biomechanical forces and to translate

    these input stimuli into distinctive phenotypes.

    In vitro, endothelial cell proliferation increases

    significantly when subjected to weak shear stress,

    while strong shear stresses are associated with

    increased nitric oxide production, and inhibition of

    endothelial cell proliferation [52,53]. In addition,

    exposure of endothelium to low and oscillating shear

    stress is associated with atherosclerotic lesion devel-

    opment [5457].

    In vitro modeling studies [58] have demonstrated

    focal areas within stents that have low shear stress

    values and may provide a milieu for endothelial cell

    activation and vascular proliferation. In vivo, coro-

    nary stent implantation can change the three-

    dimensional geometry and the shear stress distribu-

    tion of the vessel. In addition to changes at the

    entrance and exit zones of the stent [54], stagnation

    zones around the stent struts have been demonstrat-

    ed, along with a decrease of minimum wall shear

    stress by 77% in stented compared to un-stented

    vessels [59].

    In an attempt to assess the effect of a device that,

    when implanted within a stent, could increase wall

    shear stress and thereby decrease restenosis, Carlier et

    al. induced a local augmentation of wall shear stress

    with a new device, the Anti-Restenotic Diffuser flow

    divider. The investigators placed the flow divider

    randomly in one of two identical stents placed in the

    external iliac vessels of hypercholesterolemic rabbits.

    The study was controlled for confounding factors

    such as degree of hypercholesterolemia, blood

    pressure and stent design. They demonstrated that

    this device could locally increase wall shear stress

    and induce a reduction in neointimal hyperplasia.

    They also found a significant reduction in inflamma-

    N.A. Scott / Advanced Drug D366tion that was associated with the increase in shear

    stress [60].Although most studies that examined the effects

    of shear stress on restenosis were performed in

    animal models, the combination of intravascular

    ultrasound, biplane angiography and computational

    fluid dynamics has allowed investigators the unique

    opportunity to acquire detailed measurements of

    local wall geometry from a three-dimensional recon-

    struction of a human coronary artery along with local

    shear stresses. Since both biplane angiography and

    intravascular ultrasound can be performed at the time

    of stent placement and at follow-up, shear stress

    values can be calculated, followed over time and

    correlated to the observed changes in vascular

    geometry.

    Wentzel et al. [61] studied 14 patients 6 months

    after implantation of a self-expanding coronary Wall-

    stent. They performed three-dimensional reconstruc-

    tion with a combined angiographic and intravascular

    ultrasound technique. The bare stent reconstruction

    was used to calculate in-stent shear stress at implan-

    tation, by applying computational fluid dynamics.

    They found that neointimal thickness measured 6

    months after stent implantation was inversely related

    to shear stress.

    Thury et al. [62] calculated wall shear stress after

    balloon angioplasty and determined its predictive

    value for long-term outcome. Wall shear stress

    values measured proximal to and in the dilated

    lesion were higher in vessels that developed reste-

    nosis. In-lesion wall shear stress was predictive of

    angiographic restenosis and the proximal wall shear

    stress value was an independent predictor of target

    lesion revascularization.

    Stone et al. [63] used also performed three-

    dimensional coronary reconstruction with biplane

    angiography and intravascular ultrasound to examine

    changes in shear stress after coronary intervention.

    They examined six stented arteries that underwent

    intravascular profiling initially and at follow-up 6

    months later. In these stented segments, there was

    evidence of neointimal hyperplasia, with a decrease in

    lumen radius and increase in endothelial shear stress

    at all levels of baseline shear stress. Evidence of in-

    stent restenosis appeared to occur to some degree in

    each category of baseline endothelial shear stress in

    this small group of patients. These exciting prelimi-

    y Reviews 58 (2006) 358376nary studies suggest that the effect of endothelial

    shear stress on the process of in-stent restenosis in

  • with other catheter-based interventions. Camori et al.

    [70] studied patients who underwent a catheter-based

    eliverintervention (stent placement, balloon angioplasty or

    directional catheter atherectomy) on the left anterior

    descending coronary artery. Several months after thehumans may be less clear than in animal studies and

    will require more extensive investigation in larger

    numbers of patients.

    8. Stent endothelialization

    An intact endothelial layer overlying the stent is

    thought to be required for the inhibition of neointimal

    growth. In addition to focal deep injury from struts

    [6466] and overall arterial strain, stent deployment

    also causes partial denudation of the endothelium in a

    pattern unique to each stent configuration, suggesting

    balloon-related injury. Rogers et al. used a finite

    element analysis to determine that higher inflation

    pressures, wider stent-strut openings, and more com-

    pliant balloon materials cause larger surface-contact

    areas and contact stresses between stent struts [67].

    Regrowth of the endothelial layer after stent

    placement occurs within 1 month in rabbit external

    iliac vessels [68], however, this process may require

    significantly more time in humans. Grewe et al. [69]

    used scanning electron microscopy to examine

    coronary artery segments from 18 patients who died

    between 1 and 340 days after stent implantation.

    They described three phases of stent integration. In

    the acute phase (b6 weeks), the border between thevascular lumen and arterial wall was constituted by a

    thin, multilayered thrombus. No endothelial cells

    were found in the implantation zone, however,

    smooth muscle cells and extracellular matrix were

    detected. In the intermediate phase (612 weeks), the

    neointima consisted of extracellular matrix and

    increasing numbers of smooth muscle cells. Endo-

    thelial cells were also found on the luminal surface

    of the stent neointima. Complete re-endothelializa-

    tion occurred in the chronic phase (3 months after

    stent placement).

    There is evidence to suggest that the response of

    these regenerated endothelial cells to vasoactive

    agents may differ significantly from the response seen

    N.A. Scott / Advanced Drug Dintervention, endothelial reactivity was measured at a

    segment distal to the treated lesion and quantified by9. Progenitor cells

    In the past, the regeneration of injured endothelium

    had been attributed to the migration and proliferation

    of neighboring endothelial cells. Accumulating evi-

    dence now indicates that bone marrow-derived cells

    are involved in repair processes throughout the

    cardiovascular system. These cells normally circulate

    throughout the vascular system in very low levels.

    However, following acute vascular injury, a rapid

    increase in circulating levels of these endothelial

    progenitor cells is seen [72]. Additionally, circulating

    endothelial precursor cells can home to denuded parts

    of the artery after balloon injury [73].

    George et al. [74] examined 16 patients with

    angiographically demonstrated in-stent restenosis

    and compared them with eleven patients with

    similar clinical presentation that exhibited patent

    stents. The groups were similar with respect to the

    use of drugs that could potentially influence

    endothelial progenitor cell numbers. Circulating

    endothelial progenitor cell numbers were determinedthe degree of angiographic spasm or dilation induced

    by infused intracoronary acetylcholine, which acts

    directly on the endothelium. There was persistent

    endothelial dysfunction in the stented coronary artery.

    More severe endothelium-dependent vasoconstriction

    was observed in the left anterior descending of stented

    patients than in the LAD of patients who had

    undergone balloon angioplasty or atherectomy. The

    stent group had more than twice as much distal spasm

    as the other two groups (22% vs. 9%). Stenting was

    the only variable associated with this marker of

    endothelial dysfunction.

    Vascular endothelial growth factor accelerates

    endothelial repair by stimulating endothelial cell

    migration and proliferation. Hedman et al. [71]

    performed a randomized placebo-controlled, double

    blind study to determine if VEGF gene transfer could

    prevent in-stent restenosis. Although the gene transfer

    procedure was tolerated well, there was no difference

    in restenosis between the patients who received

    vascular endothelial growth factor and the patients

    who received placebo.

    y Reviews 58 (2006) 358376 367by the colony-forming unit assay, and their pheno-

    type was characterized by endothelial-cell markers.

  • origin of neointimal cells in in-stent restenosis,

    atherectomy samples from patients with in-stent

    eliverrestenosis were compared to samples from athero-

    sclerotic primary lesions. Whereas alpha-smooth

    muscle actin positive cells constituted the largest

    intimal cell pool, immunohistochemical staining for

    bone marrow- and neural crest-derived cells was

    consistently present in in-stent restenosis. These data

    suggest that in addition to endothelial progenitor

    cells, other extravascular cells are recruited to the

    neointima in the formation of human in-stent

    restenosis [75].

    10. Clinical predictors of in-stent restenosis

    As with balloon angioplasty, clinical restenosis has

    many definitions. Angiographic restenosis is usually

    based on a dichotomous distinction of greater than

    50% diameter stenosis at the site of stent placement.

    Typically, the follow-up angiographic procedure is

    performed at least 6 months after stent placement.

    However, while angiographic parameters of restenosis

    have added to the understanding of the mechanisms of

    restenosis, clinical outcomes must be regarded as the

    true measure of treatment success [76]. Clinical

    definitions of success involve repeat procedures that

    are usually based on symptoms or other signs of

    myocardial ischemia. Target lesion revascularization

    (TLR) is usually defined as any repeat percutaneousAdhesiveness of the endothelial progenitor cells

    from both groups to extracellular matrix and to

    endothelial cells was also assayed. Overall, patients

    with in-stent restenosis and with patent stents

    displayed a similar number of circulating endothelial

    progenitor cells. Fibronectin-binding was compro-

    mised in patients with in-stent restenosis as com-

    pared with their controls exhibiting patent stents.

    Patients with diffuse in-stent restenosis exhibited

    reduced numbers of circulating endothelial progen-

    itor cells in comparison with subjects with focal in-

    stent lesions. These data suggest that reduced

    numbers of circulating endothelial progenitor cells

    and impaired adhesion of these cells to fibronectin

    may contribute to diffuse in-stent restenosis.

    In a study to assess the cellularity, cell type and

    N.A. Scott / Advanced Drug D368revascularization or surgical bypass of the original

    target lesion site that occurs 30 days after the indexprocedure. Target vessel revascularization (TVR)

    usually describes a percutaneous revascularization or

    bypass of the target lesion or any segment of the

    epicardial coronary artery containing the target lesion

    or more proximal vessels that may have been

    traversed by the angioplasty guidewire. In a study of

    over 6000 patients who underwent coronary stenting

    and clinical and angiographic follow-up over a 12-

    month period, Cutlip et al. [77] found that only one

    half of patients with angiographic restenosis mani-

    fested clinical restenosis. The likelihood of TLR

    correlated with the severity of the angiographic

    stenosis. An important observation was that the

    duration of follow-up influenced the perceived rate

    of clinical restenosis. They noted that the rate of TLR

    was 7% at 6 months but increased to 12% at 1 year.

    These findings suggest that the clinical response to

    stenting may significantly differ from balloon angio-

    plasty and require longer observation for clinical end-

    points than the 6-month period usually used with

    PTCA.

    10.1. Procedural characteristics

    Goldberg et al. [78] evaluated a consecutive series

    of 456 coronary lesions with in-stent restenosis. They

    defined diffuse restenosis as a follow-up lesion length

    z10 mm and aggressive restenosis as either anincrease in lesion length from the original lesion or

    a restenotic narrowing tighter than the original. They

    found that diffuse restenosis was associated with a

    smaller reference artery diameter, longer lesion length,

    female gender, longer stent length and the use of coil

    stents. Aggressive restenosis was more common in

    women, with the use of Wallstents and with long stent

    to lesion length ratios. Aggressive restenosis occurred

    earlier and was more closely associated with symp-

    toms and myocardial infarctions than non-aggressive

    restenotic lesions. Several other studies have demon-

    strated an association between restenosis and stent

    length [7982], multiple stents [81,82], smaller final

    minimal lumen diameter [79,80,82,83], and lack of

    use of intravascular ultrasound [79].

    10.2. Diabetes

    y Reviews 58 (2006) 358376A number of large studies have suggested that

    diabetics have a higher incidence of restenosis after

  • of co-morbid diseases, such as diabetes. In addition,

    coronary diameter tends to be smaller in women.

    eliverstent placement when compared to non-diabetics

    [77,82,8486]. Gilbert et al. performed a multivariate

    analysis on six studies including 1166 patients with

    diabetes and 5070 without [87]. The average reste-

    nosis rates among patients with and without diabetes

    were 36.7% and 25.9%, respectively. Restenosis rates

    were also shown to be higher among older patients

    with and without diabetes. The authors found that the

    diabetic patients were older and that the incidence of

    restenosis after stenting is reduced by approximately

    half after adjusting for age. They concluded that the

    apparent effect of diabetes on restenosis is overstated.

    A subsequent study retrospectively analyzed all

    stented diabetic patients in 16 studies of percutaneous

    coronary intervention. Within these studies, 418 of

    3090 (14%) stented patients with 6-month angio-

    graphic follow-up had diabetes. Restenosis occurred

    in 21% of the non-diabetic and 31% of the diabetic

    patients. There was no significant difference in age

    between the two groups [88], suggesting that diabetes

    is associated with an increased incidence of stent

    restenosis. Another study suggested that an important

    contributor to the increased incidence of revascular-

    ization procedures seen in diabetics is not only

    restenosis but also progression of coronary athero-

    sclerotic disease [89]. At this time, it appears safe to

    assume from the myriad of smaller studies conducted,

    that diabetes does predispose to in-stent restenosis,

    however, the amount of influence diabetes has on

    restenosis is controversial.

    10.3. Angiotensin converting enzyme

    Since 1993, a number of studies have suggested a

    correlation between angiotensin converting enzyme

    genotype insertion or deletion polymorphism and

    restenosis after coronary intervention. Elevated levels

    of the DD genotype for angiotensin converting

    enzyme (which is associated with high plasma

    angiotensin converting enzyme levels) have been

    associated with restenosis, however the data are

    conflicting. Bonnici et al. [90] conducted a meta-

    analysis of 16 studies involving 11 without stenting

    (2535 patients) and 5 with stenting (4631 patients)

    that examined the relationship between ACE genotype

    and restenosis. There was no significant heterogeneity

    N.A. Scott / Advanced Drug Dbetween studies of percutaneous intervention with

    stenting and those without stenting. However, whenGiven these two predisposing factors, it is not

    surprising that the incidence of in-stent restenosis

    has been reported to be higher in women [78].

    However, one study was unable to demonstrate an

    increased risk of in-stent restenosis [91] and, in a

    study comprising over 4000 patients (1025 women),

    there was a lower risk of restenosis in women after

    coronary stenting despite a preponderance of diabetes

    and small vessel size [92]. Thus, there is controversy

    as to whether gender in a significant influence on in-

    stent restenosis. In an interesting study, Ferrero et al.

    examined polymorphisms of the alpha-estrogen re-

    ceptor gene in men and women and found that women

    who were homozygous for the T-allele of the PvuII

    polymorphism of the alpha-estrogen receptor gene

    have a higher risk of in stent restenosis than men [93].

    11. Angiographic patterns of in-stent restenosis

    Mehran et al. [84] examined the angiographic

    images of 288 in-stent restenosis lesions in 245

    patients who were treated with tubular slotted stents

    (primarily Palmaz-Schatz). All patients underwent

    intravascular ultrasound in addition to angiography.

    Class I Focal in-stent restenosis group. Lesions

    are V10 mm in length and are positionedat the unscaffolded segment (i.e., articu-

    lation or gap), the body of the stent, the

    proximal or distal margin (but not both),the studies were grouped by size, the odds ratios

    decreased with increasing study size. Also, blinding of

    the laboratory staff also decreased the odds ratios. The

    authors found weaker associations between the

    angiotensin converting enzyme DD genotype and

    restenosis in the larger and more rigorous studies and

    suggested that much of this association may be

    artifactual.

    10.4. Gender

    Women tend to develop coronary disease at a later

    age then men, and as a result, have a higher incidence

    y Reviews 58 (2006) 358376 369or a combination of these sites (multifocal

    in-stent restenosis).

  • myocardial infarction. However, a significant increase

    occurred in target lesion revascularization with

    eliverincreasing levels of in-stent restenosis classification

    (class I, 19%; class II, 35%; class III, 50%; and class

    IV, 83%; P b0.0001). In a multivariate analysis, theonly parameters that predicted target lesion revascu-

    larization after treatment of in-stent restenosis were

    the pattern according to the angiographic classifica-

    tion, the occurrence of previous in-stent restenosis and

    the presence of diabetes mellitus.

    12. Treatment of in-stent restenosis

    Comprehensive reviews of the literature on the

    treatment of in-stent restenosis have been published

    [9496]. In an effort to identify the most effective

    treatment modality for in-stent restenosis, Radke et

    al. [97] reviewed all papers published between 1987

    and March 2001 that reported clinical results of

    treatment of in-stent restenosis with one of six

    different treatment modalities: balloon angioplasty,

    stentin-stent therapy, rotational atherectomy, direc-

    tional atherectomy, excimer laser angioplasty, and

    intracoronary radiation. They found 28 papers that

    included a total of 3012 patients with a clinical

    follow up of 9F64 months. They performed ameta-analysis and found that treatment of in-stent

    restenosis was associated with a 30% rate of majorClass II bDiffuse intrastentQ in-stent restenosis.Lesions are N10 mm in length and areconfined to the stent(s), without extending

    outside the margins of the stent(s).

    Class III bDiffuse proliferativeQ in-stent restenosis.Lesions are N10 mm in length and extendbeyond the margin(s) of the stent(s).

    Class IV In-stent restenosis with btotal occlusion.QLesions have a TIMI flow grade of 0.

    Of the 288 lesions analyzed, 42% (n =121) were

    focal (Class I), 21% (n =61) diffuse intrastent (Class

    II), 30% (n =86) diffuse proliferative (Class III), and

    7% (n =20) total occlusions.

    The investigators found that the 1-year clinical

    event rates were uniformly high, without significant

    differences among the groups with respect to death or

    N.A. Scott / Advanced Drug D370adverse cardiac events (death, myocardial infarction

    or target lesion revascularization), regardless of thetype of device used. Repeat balloon angioplasty

    was judged to be safe, associated with a high rate

    of procedural success, cost-effective and had favor-

    able long-term results in long lesions. After

    adjustment of confounding factors (lesion length,

    pre-procedural diameter stenosis, diabetes) the only

    treatment modality felt to be superior to balloon

    angioplasty was intracoronary radiation. Since 2001,

    a number of multicenter studies have been pub-

    lished that closely examine the efficacy of catheter-

    based technologies in the treatment of in-stent

    restenosis.

    12.1. Radiation

    Intravascular brachytherapy with both beta and

    gamma sources has been proven in a number of

    randomized trials to be an effective treatment for in-

    stent restenosis [98]. The beneficial effect of radiation

    is sustained, out to at least 5 years [99,100].

    Restenosis rates after radiation were higher in longer

    lesions, but still significantly less than controls

    [98,101,102]. Diabetic patients, despite having longer

    and more complex lesions, have been reported to have

    no increase in target lesion revascularization when

    compared to non-diabetic patients [103,104]. Al-

    though some studies have shown a failure rate in

    diabetics that was higher than non-diabetics, reste-

    nosis rates were still lower than controls [105]. Sabate

    et al. have demonstrated that the incidence of

    neointimal proliferation after brachytherapy is similar

    between diabetics and non-diabetics but the incidence

    of late stent thrombosis and stenoses proximal or

    distal to the stent were higher in diabetic patients and

    were frequently the cause of the target lesion

    revascularization [106].

    Success after brachytherapy can be predicted by

    the presenting angiographic pattern of the in-stent

    restenosis. After treatment with gamma vascular

    brachytherapy, the binary angiographic restenosis

    rate increased with worsening in-stent restenosis

    patterns; however, target lesion revascularization

    and major adverse cardiac event rates increased for

    focal, diffuse, and proliferative patterns of in-stent

    restenosis but not for total occlusions [107]. In those

    patients who develop restenosis after radiation for in-

    y Reviews 58 (2006) 358376stent restenosis, the predominant angiographic pat-

    tern of the lesions is focal restenosis, and these

  • eliverlesions respond well to conventional revasculariza-

    tion methods [108].

    12.2. Rotational atherectomy

    A single-center randomized trial of rotational

    atherectomy versus balloon angioplasty for diffuse

    in-stent restenosis demonstrated a lower incidence of

    target lesion revascularization in the rotational athe-

    rectomy group [109]. However, a multicenter, ran-

    domized, prospective trial comprising 298 patients

    with in-stent restenosis showed no significant benefit

    over balloon angioplasty [110].

    12.3. Cutting balloon

    The cutting balloon is a balloon catheter with three

    or four microsurgical blades bonded longitudinally to

    the balloon surface. When the balloon is expanded,

    the blades incise and facilitate redistribution of the

    lesion. In a randomized, prospective, multicenter trial

    of cutting balloon angioplasty to conventional angio-

    plasty, there was no difference in angiographic

    restenosis rates or clinical events between the two

    groups [111].

    12.4. Laser angioplasty

    In a multicenter registry that compared laser to

    conventional balloon angioplasty in patients with in-

    stent restenosis, there was no difference in the

    incidence of major cardiac events or target lesion

    revascularization [112]. However, the authors noted

    that the lesions in the laser group were longer and

    more complex.

    12.5. Drug-eluting stents

    The use of drug-eluting stents will be discussed

    in detail in other chapters. However, several

    relatively small trials have demonstrated that signif-

    icant benefit may be obtained with implanting a

    paclitaxel- [113] or rapamycin- [114116] eluting

    stent as a treatment of for in-stent restenosis. The

    benefit obtained may be similar to that seen with

    intracoronary radiation [117]. In addition, high-dose

    N.A. Scott / Advanced Drug Doral rapamycin has efficacy in prevention of in-stent

    restenosis [118].References

    [1] A.R. Gruntzig, A. Senning, W.E. Siegenthaler, Nonoperative

    dilatation of coronary-artery stenosis: percutaneous trans-

    luminal coronary angioplasty, N. Engl. J. Med. 301 (1979)

    6168.

    [2] D.R. Holmes Jr., R.E. Vlietstra, H.C. Smith, G.W. Vetrovec,

    K.M. Kent, M.J. Cowley, D.P. Faxon, A.R. Gruentzig, S.F.

    Kelsey, K.M. Detre, Restenosis after percutaneous trans-

    luminal coronary angioplasty (PTCA): a report from the

    PTCA Registry of the National Heart, Lung, and Blood

    Institute, Am. J. Cardiol. 53 (1984) 77C81C.

    [3] C.E. Essed, B.M. van den, A.E. Becker, Transluminal

    coronary angioplasty and early restenosis. Fibrocellular

    occlusion after wall laceration, Br. Heart J. 49 (1983)

    393396.

    [4] J. Schneider, A.R. Gruntzig, Percutaneous transluminal

    coronary angioplasty: morphological findings in 3 patients,

    Pathol. Res. Pract. 180 (1985) 348352.

    [5] G.E. Austin, N.B. Ratliff, J. Hollman, S. Tabei, D.F. Phillips,

    Intimal proliferation of smooth muscle cells as an explanation

    for recurrent coronary artery stenosis after percutaneous

    transluminal coronary angioplasty, J. Am. Coll. Cardiol. 6

    (1985) 369375.

    [6] J.A. Bittl, D.P. Chew, E.J. Topol, D.F. Kong, R.M. Califf,

    Meta-analysis of randomized trials of percutaneous trans-

    luminal coronary angioplasty versus atherectomy, cutting

    balloon atherotomy, or laser angioplasty, J. Am. Coll.

    Cardiol. 43 (2004) 936942.

    [7] P.W. Serruys, P. de Jaegere, F. Kiemeneij, C. Macaya, W.

    Rutsch, G. Heyndrickx, H. Emanuelsson, J. Marco, V.

    Legrand, P. Materne, A comparison of balloon-expandable-

    stent implantation with balloon angioplasty in patients with

    coronary artery disease. Benestent Study Group, N. Engl. J.

    Med. 331 (1994) 489495.

    [8] D.L. Fischman, M.B. Leon, D.S. Baim, R.A. Schatz, M.P.

    Savage, I. Penn, K. Detre, L. Veltri, D. Ricci, M. Nobuyoshi,

    A randomized comparison of coronary-stent placement and

    balloon angioplasty in the treatment of coronary artery

    disease. Stent Restenosis Study Investigators, N. Engl. J.

    Med. 331 (1994) 496501.

    [9] U. Sigwart, J. Puel, V. Mirkovitch, F. Joffre, L. Kappen-

    berger, Intravascular stents to prevent occlusion and reste-

    nosis after transluminal angioplasty, N. Engl. J. Med. 316

    (1987) 701706.

    [10] P. Hall, S. Nakamura, L. Maiello, A. Itoh, S. Blengino, G.

    Martini, M. Ferraro, A. Colombo, A randomized compar-

    ison of combined ticlopidine and aspirin therapy versus

    aspirin therapy alone after successful intravascular ultra-

    sound-guided stent implantation, Circulation 93 (1996)

    215222.

    [11] M.B. Leon, D.S. Baim, J.J. Popma, P.C. Gordon, D.E. Cutlip,

    K.K. Ho, A. Giambartolomei, D.J. Diver, D.M. Lasorda,

    D.O. Williams, S.J. Pocock, R.E. Kuntz, A clinical trial

    comparing three antithrombotic-drug regimens after coro-

    y Reviews 58 (2006) 358376 371nary-artery stenting. Stent Anticoagulation Restenosis Study

    Investigators, N. Engl. J. Med. 339 (1998) 16651671.

  • N.A. Scott / Advanced Drug Delivery Reviews 58 (2006) 358376372[12] R. Komatsu, M. Ueda, T. Naruko, A. Kojima, A.E. Becker,

    Neointimal tissue response at sites of coronary stenting in

    humans: macroscopic, histological, and immunohistochemi-

    cal analyses, Circulation 98 (1998) 224233.

    [13] A. Farb, G. Sangiorgi, A.J. Carter, V.M. Walley, W.D.

    Edwards, R.S. Schwartz, R. Virmani, Pathology of acute and

    chronic coronary stenting in humans, Circulation 99 (1999)

    4452.

    [14] A. Farb, D.K. Weber, F.D. Kolodgie, A.P. Burke, R. Virmani,

    Morphological predictors of restenosis after coronary stenting

    in humans, Circulation 105 (2002) 29742980.

    [15] H.M. van Beusekom, W.J. van der Giessen, R. van Suylen, E.

    Bos, F.T. Bosman, P.W. Serruys, Histology after stenting of

    human saphenous vein bypass grafts: observations from

    surgically excised grafts 3 to 320 days after stent implanta-

    tion, J. Am. Coll. Cardiol. 21 (1993) 4554.

    [16] P. Martin, Wound healingaiming for perfect skin regener-

    ation, Science 276 (1997) 7581.

    [17] A. Farb, F.D. Kolodgie, J.Y. Hwang, A.P. Burke, K. Tefera,

    D.K. Weber, T.N. Wight, R. Virmani, Extracellular matrix

    changes in stented human coronary arteries, Circulation 110

    (2004) 940947.

    [18] I.M. Chung, H.K. Gold, S.M. Schwartz, Y. Ikari, M.A. Reidy,

    T.N. Wight, Enhanced extracellular matrix accumulation in

    restenosis of coronary arteries after stent deployment, J. Am.

    Coll. Cardiol. 40 (2002) 20722081.

    [19] N.A. Scott, G.D. Cipolla, C.E. Ross, B. Dunn, F.H. Martin,

    L. Simonet, J.N. Wilcox, Identification of a potential role for

    the adventitia in vascular lesion formation after balloon

    overstretch injury of porcine coronary arteries, Circulation 93

    (1996) 21782187.

    [20] M.J. Post, C. Borst, R.E. Kuntz, The relative importance of

    arterial remodeling compared with intimal hyperplasia in

    lumen renarrowing after balloon angioplasty. A study in the

    normal rabbit and the hypercholesterolemic Yucatan micro-

    pig, Circulation 89 (1994) 28162821.

    [21] M. Nakatani, Y. Takeyama, M. Shibata, M. Yorozuya, H.

    Suzuki, S. Koba, T. Katagiri, Mechanisms of restenosis

    after coronary intervention: difference between plain old

    balloon angioplasty and stenting, Cardiovasc. Pathol. 12

    (2003) 4048.

    [22] G.S. Mintz, J.J. Popma, A.D. Pichard, K.M. Kent, L.F. Satler,

    C. Wong, M.K. Hong, J.A. Kovach, M.B. Leon, Arterial

    remodeling after coronary angioplasty: a serial intravascular

    ultrasound study, Circulation 94 (1996) 3543.

    [23] R. Hoffmann, G.S. Mintz, G.R. Dussaillant, J.J. Popma,

    A.D. Pichard, L.F. Satler, K.M. Kent, J. Griffin, M.B.

    Leon, Patterns and mechanisms of in-stent restenosis. A

    serial intravascular ultrasound study, Circulation 94 (1996)

    12471254.

    [24] R. Hoffmann, G.S. Mintz, J.J. Popma, L.F. Satler, A.D.

    Pichard, K.M. Kent, C. Walsh, P. Mackell, M.B. Leon, Chronic

    arterial responses to stent implantation: a serial intravascular

    ultrasound analysis of Palmaz-Schatz stents in native coronary

    arteries, J. Am. Coll. Cardiol. 28 (1996) 11341139.

    [25] M. Nakamura, P.G. Yock, H.N. Bonneau, K. Kitamura,T. Aizawa, H. Tamai, P.J. Fitzgerald, Y. Honda, Impactof peri-stent remodeling on restenosis: a volumetric

    intravascular ultrasound study, Circulation 103 (2001)

    21302132.

    [26] F.G. Welt, C. Rogers, Inflammation and restenosis in the stent

    era, Arterioscler. Thromb. Vasc. Biol. 22 (2002) 17691776.

    [27] A. Colombo, G. Sangiorgi, The monocyte: the key in the lock

    to reduce stent hyperplasia? J. Am. Coll. Cardiol. 43 (2004)

    2426.

    [28] F.G. Welt, E.R. Edelman, D.I. Simon, C. Rogers, Neutrophil,

    not macrophage, infiltration precedes neointimal thickening

    in balloon-injured arteries, Arterioscler. Thromb. Vasc. Biol.

    20 (2000) 25532558.

    [29] C. Rogers, F.G. Welt, M.J. Karnovsky, E.R. Edelman,

    Monocyte recruitment and neointimal hyperplasia in rabbits.

    Coupled inhibitory effects of heparin, Arterioscler. Thromb.

    Vasc. Biol. 16 (1996) 13121318.

    [30] C. Rogers, E.R. Edelman, D.I. Simon, A mAb to the beta2-

    leukocyte integrin Mac-1 (CD11b/CD18) reduces intimal

    thickening after angioplasty or stent implantation in rabbits,

    Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 1013410139.

    [31] E. Mori, K. Komori, T. Yamaoka, M. Tanii, C. Kataoka, A.

    Takeshita, M. Usui, K. Egashira, K. Sugimachi, Essential role

    of monocyte chemoattractant protein-1 in development of

    restenotic changes (neointimal hyperplasia and constrictive

    remodeling) after balloon angioplasty in hypercholesterol-

    emic rabbits, Circulation 105 (2002) 29052910.

    [32] D. Fukuda, K. Shimada, A. Tanaka, T. Kawarabayashi, M.

    Yoshiyama, J. Yoshikawa, Circulating monocytes and in-

    stent neointima after coronary stent implantation, J. Am.

    Coll. Cardiol. 43 (2004) 1823.

    [33] F.J. Neumann, I. Ott, M. Gawaz, G. Puchner, A. Schomig,

    Neutrophil and platelet activation at balloon-injured coronary

    artery plaque in patients undergoing angioplasty, J. Am. Coll.

    Cardiol. 27 (1996) 819824.

    [34] J.K. Mickelson, N.M. Lakkis, G. Villarreal-Levy, B.J.

    Hughes, C.W. Smith, Leukocyte activation with platelet

    adhesion after coronary angioplasty: a mechanism for

    recurrent disease? J. Am. Coll. Cardiol. 28 (1996) 345353.

    [35] T. Inoue, Y. Sakai, S. Morooka, T. Hayashi, K.

    Takayanagi, Y. Takabatake, Expression of polymorphonu-

    clear leukocyte adhesion molecules and its clinical

    significance in patients treated with percutaneous trans-

    luminal coronary angioplasty, J. Am. Coll. Cardiol. 28

    (1996) 11271133.

    [36] T. Inoue, R. Sohma, T. Miyazaki, Y. Iwasaki, I. Yaguchi, S.

    Morooka, Comparison of activation process of platelets and

    neutrophils after coronary stent implantation versus balloon

    angioplasty for stable angina pectoris, Am. J. Cardiol. 86

    (2000) 10571062.

    [37] T. Inoue, T. Uchida, I. Yaguchi, Y. Sakai, K. Takayanagi,

    S. Morooka, Stent-induced expression and activation of

    the leukocyte integrin Mac-1 is associated with neo-

    intimal thickening and restenosis, Circulation 107 (2003)

    17571763.

    [38] F. Cipollone, M. Marini, M. Fazia, B. Pini, A. Iezzi, M.

    Reale, L. Paloscia, G. Materazzo, E. DAnnunzio, P. Conti, F.Chiarelli, F. Cuccurullo, A. Mezzetti, Elevated circulating

  • N.A. Scott / Advanced Drug Delivery Reviews 58 (2006) 358376 373levels of monocyte chemoattractant protein-1 in patients with

    restenosis after coronary angioplasty, Arterioscler. Thromb.

    Vasc. Biol. 21 (2001) 327334.

    [39] S. Oshima, H. Ogawa, S. Hokimoto, S. Nakamura, K. Noda,

    T. Saito, H. Soejima, K. Takazoe, F. Ishibashi, H. Yasue,

    Plasma monocyte chemoattractant protein-1 antigen levels

    and the risk of restenosis after coronary stent implantation,

    Jpn. Circ. J. 65 (2001) 261264.

    [40] J.F. Tanguay, T. Hammoud, P. Geoffroy, Y. Merhi, Chronic

    platelet and neutrophil adhesion: a causal role for neointimal

    hyperplasia in in-stent restenosis, J. Endovasc. Ther. 10

    (2003) 968977.

    [41] F.G. Welt, C. Tso, E.R. Edelman, M.A. Kjelsberg, J.F.

    Paolini, P. Seifert, C. Rogers, Leukocyte recruitment and

    expression of chemokines following different forms of

    vascular injury, Vasc. Med. 8 (2003) 17.

    [42] C. Horvath, F.G. Welt, M. Nedelman, P. Rao, C. Rogers,

    Targeting CCR2 or CD18 inhibits experimental in-stent

    restenosis in primates: inhibitory potential depends on type

    of injury and leukocytes targeted, Circ. Res. 90 (2002)

    488494.

    [43] K. Ohtani, M. Usui, K. Nakano, Y. Kohjimoto, S. Kitajima,

    Y. Hirouchi, X.H. Li, S. Kitamoto, A. Takeshita, K.

    Egashira, Antimonocyte chemoattractant protein-1 gene

    therapy reduces experimental in-stent restenosis in hyper-

    cholesterolemic rabbits and monkeys, Gene Ther. 11 (2004)

    12731282.

    [44] R. Kawamoto, K. Hatakeyama, T. Imamura, T. Ishikawa, H.

    Date, Y. Shibata, M. Takenaga, Y. Asada, T. Eto, Relation of

    C-reactive protein to restenosis after coronary stent implan-

    tation and to restenosis after coronary atherectomy, Am. J.

    Cardiol. 94 (2004) 104107.

    [45] F. Versaci, A. Gaspardone, F. Tomai, F. Ribichini, P. Russo,

    I. Proietti, A.S. Ghini, V. Ferrero, L. Chiariello, P.A. Gioffre,

    F. Romeo, F. Crea, Immunosuppressive therapy for the

    prevention of restenosis after coronary artery stent implan-

    tation (IMPRESS Study), J. Am. Coll. Cardiol. 40 (2002)

    19351942.

    [46] D.H. Walter, S. Fichtlscherer, M. Sellwig, W. Auch-Schwelk,

    V. Schachinger, A.M. Zeiher, Preprocedural C-reactive

    protein levels and cardiovascular events after coronary stent

    implantation, J. Am. Coll. Cardiol. 37 (2001) 839846.

    [47] S. Glagov, E. Weisenberg, C.K. Zarins, R. Stankunavicius,

    G.J. Kolettis, Compensatory enlargement of human athero-

    sclerotic coronary arteries, N. Engl. J. Med. 316 (1987)

    13711375.

    [48] N. Resnick, M.A. Gimbrone Jr., Hemodynamic forces are

    complex regulators of endothelial gene expression, FASEB J.

    9 (1995) 874882.

    [49] P.F. Davies, Flow-mediated endothelial mechanotransduc-

    tion, Physiol. Rev. 75 (1995) 519560.

    [50] M.A. Gimbrone Jr., N. Resnick, T. Nagel, L.M. Khachigian,

    T. Collins, J.N. Topper, Hemodynamics, endothelial gene

    expression, and atherogenesis, Ann. N.Y. Acad. Sci. 811

    (1997) 110.

    [51] G. Garcia-Cardena, J. Comander, K.R. Anderson, B.R.Blackman, M.A. Gimbrone Jr., Biomechanical activation ofvascular endothelium as a determinant of its functional

    phenotype, Proc. Natl. Acad. Sci. U. S. A. 98 (2001)

    44784485.

    [52] K. Lin, P.P. Hsu, B.P. Chen, S. Yuan, S. Usami, J.Y. Shyy,

    Y.S. Li, S. Chien, Molecular mechanism of endothelial

    growth arrest by laminar shear stress, Proc. Natl. Acad. Sci.

    U. S. A. 97 (2000) 93859389.

    [53] M.J. Levesque, R.M. Nerem, E.A. Sprague, Vascular

    endothelial cell proliferation in culture and the influence of

    flow, Biomaterials 11 (1990) 702707.

    [54] J.N. Oshinski, D.N. Ku, S. Mukundan Jr., F. Loth, R.I.

    Pettigrew, Determination of wall shear stress in the aorta with

    the use of MR phase velocity mapping, J. Magn. Reson.

    Imaging 5 (1995) 640647.

    [55] K.S. Cunningham, A.I. Gotlieb, The role of shear stress in the

    pathogenesis of atherosclerosis, Lab. Invest. 85 (2004) 923.

    [56] C. Cheng, R. de Crom, R. van Haperen, F. Helderman, B.M.

    Gourabi, L.C. van Damme, S.W. Kirschbaum, C.J. Slager,

    A.F. van der Steen, R. Krams, The role of shear stress in

    atherosclerosis: action through gene expression and inflam-

    mation? Cell Biochem. Biophys. 41 (2004) 279294.

    [57] A.M. Malek, S.L. Alper, S. Izumo, Hemodynamic shear

    stress and its role in atherosclerosis, JAMA 282 (1999)

    20352042.

    [58] N. Benard, D. Coisne, E. Donal, R. Perrault, Experimental

    study of laminar blood flow through an artery treated by a

    stent implantation: characterisation of intra-stent wall shear

    stress, J. Biomech. 36 (2003) 991998.

    [59] J.F. LaDisa Jr., I. Guler, L.E. Olson, D.A. Hettrick, J.R.

    Kersten, D.C. Warltier, P.S. Pagel, Three-dimensional com-

    putational fluid dynamics modeling of alterations in coronary

    wall shear stress produced by stent implantation, Ann.

    Biomed. Eng. 31 (2003) 972980.

    [60] S.G. Carlier, L.C. van Damme, C.P. Blommerde, J.J.

    Wentzel, G. van Langehove, S. Verheye, M.M. Kockx,

    M.W. Knaapen, C. Cheng, F. Gijsen, D.J. Duncker, N.

    Stergiopulos, C.J. Slager, P.W. Serruys, R. Krams,

    Augmentation of wall shear stress inhibits neointimal

    hyperplasia after stent implantation: inhibition through

    reduction of inflammation? Circulation 107 (2003)

    27412746.

    [61] J.J. Wentzel, R. Krams, J.C. Schuurbiers, J.A. Oomen, J.

    Kloet, W.J. Der Giessen, P.W. Serruys, C.J. Slager, Relation-

    ship between neointimal thickness and shear stress after

    Wallstent implantation in human coronary arteries, Circula-

    tion 103 (2001) 17401745.

    [62] A. Thury, J.J. Wentzel, R.V. Vinke, F.J. Gijsen, J.C.

    Schuurbiers, R. Krams, P.J. de Feyter, P.W. Serruys, C.J.

    Slager, Images in cardiovascular medicine. Focal in-stent

    restenosis near step-up: roles of low and oscillating shear

    stress? Circulation 105 (2002) e185e187.

    [63] P.H. Stone, A.U. Coskun, S. Kinlay, M.E. Clark, M. Sonka,

    A. Wahle, O.J. Ilegbusi, Y. Yeghiazarians, J.J. Popma, J.

    Orav, R.E. Kuntz, C.L. Feldman, Effect of endothelial shear

    stress on the progression of coronary artery disease, vascular

    remodeling, and in-stent restenosis in humans: in vivo 6-month follow-up study, Circulation 108 (2003) 438444.

  • N.A. Scott / Advanced Drug Delivery Reviews 58 (2006) 358376374[64] R.S. Schwartz, K.C. Huber, J.G. Murphy, W.D. Edwards,

    A.R. Camrud, R.E. Vlietstra, D.R. Holmes, Restenosis and

    the proportional neointimal response to coronary artery

    injury: results in a porcine model, J. Am. Coll. Cardiol. 19

    (1992) 267274.

    [65] S.P. Karas, M.B. Gravanis, E.C. Santoian, K.A. Robinson,

    K.A. Anderberg, S.B. King III, Coronary intimal prolif-

    eration after balloon injury and stenting in swine: an

    animal model of restenosis, J. Am. Coll. Cardiol. 20

    (1992) 467474.

    [66] C. Rogers, E.R. Edelman, Endovascular stent design dictates

    experimental restenosis and thrombosis, Circulation 91

    (1995) 29953001.

    [67] C. Rogers, D.Y. Tseng, J.C. Squire, E.R. Edelman, Balloon-

    artery interactions during stent placement: a finite element

    analysis approach to pressure, compliance, and stent design

    as contributors to vascular injury, Circ. Res. 84 (1999)

    378383.

    [68] E. Van Belle, F.O. Tio, T. Couffinhal, L. Maillard, J. Passeri,

    J.M. Isner, Stent endothelialization. Time course, impact of

    local catheter delivery, feasibility of recombinant protein

    administration, and response to cytokine expedition, Circu-

    lation 95 (1997) 438448.

    [69] P.H. Grewe, T. Deneke, S.K. Holt, A. Machraoui, J.

    Barmeyer, K.M. Muller, Scanning electron microscopic

    analysis of vessel wall reactions after coronary stenting, Z.

    Kardiol. 89 (2000) 2127.

    [70] P.R. Caramori, V.C. Lima, P.H. Seidelin, G.E. Newton, J.D.

    Parker, A.G. Adelman, Long-term endothelial dysfunction

    after coronary artery stenting, J. Am. Coll. Cardiol. 34 (1999)

    16751679.

    [71] M. Hedman, J. Hartikainen, M. Syvanne, J. Stjernvall, A.

    Hedman, A. Kivela, E. Vanninen, H. Mussalo, E. Kauppila,

    S. Simula, O. Narvanen, A. Rantala, K. Peuhkurinen, M.S.

    Nieminen, M. Laakso, S. Yla-Herttuala, Safety and feasi-

    bility of catheter-based local intracoronary vascular endo-

    thelial growth factor gene transfer in the prevention of

    postangioplasty and in-stent restenosis and in the treatment

    of chronic myocardial ischemia: phase II results of the

    Kuopio Angiogenesis Trial (KAT), Circulation 107 (2003)

    26772683.

    [72] C. Urbich, S. Dimmeler, Endothelial progenitor cells:

    characterization and role in vascular biology, Circ. Res. 95

    (2004) 343353.

    [73] D.H. Walter, K. Rittig, F.H. Bahlmann, R. Kirchmair, M.

    Silver, T. Murayama, H. Nishimura, D.W. Losordo, T.

    Asahara, J.M. Isner, Statin therapy accelerates reendothelial-

    ization: a novel effect involving mobilization and incorpora-

    tion of bone marrow-derived endothelial progenitor cells,

    Circulation 105 (2002) 30173024.

    [74] J. George, I. Herz, E. Goldstein, S. Abashidze, V. Deutch, A.

    Finkelstein, Y. Michowitz, H. Miller, G. Keren, Number and

    adhesive properties of circulating endothelial progenitor cells

    in patients with in-stent restenosis, Arterioscler. Thromb.

    Vasc. Biol. 23 (2003) e57e60.

    [75] D. Skowasch, A. Jabs, R. Andrie, S. Dinkelbach, B. Luderitz,G. Bauriedel, Presence of bone-marrow- and neural-crest-derived cells in intimal hyperplasia at the time of clinical in-

    stent restenosis, Cardiovasc. Res. 60 (2003) 684691.

    [76] U. Rosenschein, E.J. Topol, Uncoupling clinical outcomes

    and coronary angiography: a review and perspective of recent

    trials in coronary artery disease, Am. Heart J. 132 (1996)

    910920.

    [77] D.E. Cutlip, A.G. Chhabra, D.S. Baim, M.S. Chauhan, S.

    Marulkar, J. Massaro, A. Bakhai, D.J. Cohen, R.E. Kuntz,

    K.K.L. Ho, Beyond restenosis: five-year clinical outcomes

    from second-generation coronary stent trials, Circulation 110

    (2004) 12261230.

    [78] S.L. Goldberg, A. Loussararian, J. De Gregorio, C. di

    Mario, R. Albiero, A. Colombo, Predictors of diffuse and

    aggressive intra-stent restenosis, J. Am. Coll. Cardiol. 37

    (2001) 10191025.

    [79] S. Kasaoka, J.M. Tobis, T. Akiyama, B. Reimers, C. di

    Mario, N.D. Wong, A. Colombo, Angiographic and intra-

    vascular ultrasound predictors of in-stent restenosis, J. Am.

    Coll. Cardiol. 32 (1998) 16301635.

    [80] P.J. de Feyter, P. Kay, C. Disco, P.W. Serruys, Reference

    chart derived from post-stent-implantation intravascular

    ultrasound predictors of 6-month expected restenosis on

    quantitative coronary angiography, Circulation 100 (1999)

    17771783.

    [81] C. Bauters, E. Hubert, A. Prat, K. Bougrimi, E. Van Belle,

    E.P. McFadden, P. Amouyel, J.M. Lablanche, M. Bertrand,

    Predictors of restenosis after coronary stent implantation, J.

    Am. Coll. Cardiol. 31 (1998) 12911298.

    [82] S.G. Lee, C.W. Lee, M.K. Hong, H.K. Park, J.J. Kim, S.W.

    Park, S.J. Park, Predictors of diffuse-type in-stent restenosis

    after coronary stent implantation, Catheter. Cardiovasc.

    Interv. 47 (1999) 406409.

    [83] N. Mercado, E. Boersma, W. Wijns, B.J. Gersh, C.A.

    Morillo, G.A. van Es, D.E. Grobbee, P.W. Serruys, Clinical

    and quantitative coronary angiographic predictors of coro-

    nary restenosis: a comparative analysis from the balloon-to-

    stent era, J. Am. Coll. Cardiol. 38 (2001) 645652.

    [84] R. Mehran, G. Dangas, A.S. Abizaid, G.S. Mintz, A.J.

    Lansky, L.F. Satler, A.D. Pichard, K.M. Kent, G.W. Stone,

    M.B. Leon, Angiographic patterns of in-stent restenosis:

    classification and implications for long-term outcome,

    Circulation 100 (1999) 18721878.

    [85] V. Mathew, B.J. Gersh, B.A. Williams, W.K. Laskey, J.T.

    Willerson, R.T. Tilbury, B.R. Davis, D.R. Holmes Jr.,

    Outcomes in patients with diabetes mellitus undergoing

    percutaneous coronary intervention in the current era: a

    report from the prevention of REStenosis with tranilast

    and its outcomes (PRESTO) trial, Circulation 109 (2004)

    476480.

    [86] W.K. Laskey, F. Selzer, H.A. Vlachos, J. Johnston, A.

    Jacobs, S.B. King III, D.R. Holmes, J. Douglas, P. Block,

    R. Wilensky, D.O. Williams, K. Detre, Comparison of in-

    hospital and one-year outcomes in patients with and

    without diabetes mellitus undergoing percutaneous catheter

    intervention (from the National Heart, Lung, and Blood

    Institute Dynamic Registry), Am. J. Cardiol. 90 (2002)10621067.

  • N.A. Scott / Advanced Drug Delivery Reviews 58 (2006) 358376 375[87] J. Gilbert, J. Raboud, B. Zinman, Meta-analysis of the effect

    of diabetes on res