Vous êtes sur la page 1sur 13

IN DEPTH

STATE OF THE ART


Acute Coronary Syndromes
The Way Forward From Mechanisms to Precision Treatment

ABSTRACT: Well into the 21st century, we still triage acute myocardial Filippo Crea, MD
infarction on the basis of the presence or absence of ST-segment Peter Libby, MD
elevation, a century-old technology. Meanwhile, we have learned a great
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

deal about the pathophysiology and mechanisms of acute coronary


syndromes (ACS) at the clinical, pathological, cellular, and molecular
levels. Contemporary imaging studies have shed new light on the
mechanisms of ACS. This review discusses these advances and their
implications for clinical management of the ACS for the future. Plaque
rupture has dominated our thinking about ACS pathophysiology for
decades. However, current evidence suggests that a sole focus on plaque
rupture vastly oversimplifies this complex collection of diseases and
obscures other mechanisms that may mandate different management
strategies. We propose segmenting coronary artery thrombosis caused
by plaque rupture into cases with or without signs of concomitant
inflammation. This distinction may have substantial therapeutic
implications as direct anti-inflammatory interventions for atherosclerosis
emerge. Coronary artery thrombosis caused by plaque erosion may be
on the rise in an era of intense lipid lowering. Identification of patients
with of ACS resulting from erosion may permit a less invasive approach to
management than the current standard of care. We also now recognize
ACS that occur without apparent epicardial coronary artery thrombus or
stenosis. Such events may arise from spasm, microvascular disease, or
other pathways. Emerging management strategies may likewise apply
selectively to this category of ACS. We advocate this more mechanistic
approach to the categorization of ACS to provide a framework for future
tailoring, triage, and therapy for patients in a more personalized and
precise manner.
Correspondence to: Peter Libby,
MD, Brigham and Womens
Hospital, Harvard Medical School,
77 Ave Louis Pasteur, NRB-741-G,
Boston, MA 02115. E-mail plibby@
bwh.harvard.edu

Key Words:erosion
inflammation microvessels
myocardial infarction plaque
plaque, atherosclerotic
thrombosis
2017 American Heart
Association, Inc.

Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1155


Crea and Libby

W
illem Einthoven introduced electrocardiogra- Moreover, although we have witnessed wide accep-
phy at the dawn of the 20th century. Well into tance of the formerly controversial concept that inflam-
the 21st century, we still triage acute myocar- mation contributes to atherogenesis,13 inflammation
dial infarction on the basis of this venerable technology: may not drive all transitions from stable atherosclerosis
ST segments up or not. Although this approach remains to acute thrombotic events. In a large study, about half
the appropriate evidence-based strategy today, aiming of ACS occurred in the presence of normal levels of
toward the future goal of more individualized therapy, C-reactive protein (CRP), a marker of inflammation.14
can we apply a more pathophysiologically based cat- Another recent study using optical coherence tomog-
egorization of the acute coronary syndromes (ACS)? In- raphy (OCT) demonstrated that among patients with
deed, we have learned much about the mechanisms of ACS and plaque rupture, two thirds of patients had im-
ACS since we last reviewed this topic in these pages.1,2 aging evidence of inflammatory cell infiltration in the
Moreover, the human disease of atherosclerosis has region of the ruptured fibrous cap but one third did
evolved considerably in the interim.35 This review aims not and the latter had lower levels of CRP.15 In addition,
to work toward a categorization of the ACS that reach- plaque erosion causes up to a third of ACS in the cur-
es beyond equating all ACS with plaque rupture or fis- rent era.16,17 Finally, about one fifth of ACS occur in the
sure, guided by the latest insights into the underlying apparent absence of coronary thrombosis, suggesting
mechanisms of ACS, and points a way forward toward that functional alterations beyond thrombus formation
the eventual clinical application of these considerations. can contribute to ACS pathogenesis.18,19
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

Postmortem studies carried out in the 1980s pro- The ensemble of postmortem studies and in vivo
posed that plaque rupture (also sometimes referred to studies using intravascular imaging point to 4 patho-
as fissure) caused most fatal myocardial infarctions.6 logical pathways to ACS. Although these mechanisms
These findings led to the notion of the vulnerable or may overlap and coexist in some patients, we present
high-risk plaque characterized by a large central lipid them separately for heuristic simplicity: plaque rupture
core, an abundance of inflammatory cells, a paucity of with systemic inflammation, plaque rupture without
systemic inflammation, plaque erosion, and plaque
smooth muscle cells (SMCs), and a thin fibrous cap.7
without thrombus (Figure1).20
These observations spawned the widely accepted con-
This review considers the causes of these 4 predomi-
cept that instability of coronary atheromata resulted
nant pathways to ACS. To strive for a more precision
from fissuring of a thin-capped fibroatheroma caused
approach, we explore the therapeutic implications of
by a weakening of its collagen structure wrought by
these distinct mechanisms that provoke ACS. Moreover,
inflammatory mechanisms.8
we advocate the future goal to move beyond triage of
This concept stimulated manifold attempts to de-
therapy based merely on electrocardiographic current
velop methods to detect the vulnerable plaque, a quest
of injury by harnessing the recent advances in arterial
predicated on the postulate that local interventions
biology and human studies that shed new mechanis-
could preclude plaque thrombosis and possibly prevent tic light on ACS pathogenesis. Further improvements
ACS. However, attempts to identify vulnerable plaques in the appropriate deployment of technologies and in
proved disappointing because of their low predictive outcomes should evolve from a more mechanistically
value.9 Pathological studies of human coronary arter- based classification of ACS.
ies show that plaque rupture occurs commonly in indi-
viduals without symptomatic ACS.10 Moreover, <5% of
plaques with the features of thin-capped fibroatheroma PLAQUE RUPTURE WITH SYSTEMIC
as determined by intravascular ultrasound interroga-
INFLAMMATION
tion actually cause clinical events during over 3 years
of follow-up.11 Thus, most thin-capped fibroatheroma Mechanisms
are clinically quite stable, a realization that renders Several studies have implicated systemic inflammation
vulnerable plaque a misnomer. Indeed, plaques with in ACS as assessed by the biomarker CRP (Figure1A).21
thin-capped fibroatheroma morphology as assessed Laboratory studies and observations on human plaques
by radiofrequency backscatter data from intravascular point to inflammatory mechanisms as key regulators of
ultrasound (virtual histology) commonly convert to an the fragility of the fibrous cap and of the thrombogenic
apparently more stable character during a 1-year fol- potential of the lipid core (Figure2). Macrophages likely
low-up period.12 Such evolution in plaques may reflect pave the way for the rupture of the fibrous cap of the
mutability during the natural history of human athero- plaque: When activated, these cells elaborate enzymes
sclerosis but could also reflect adherence of many of that degrade all components of the arterial extracellular
these patients to a contemporary secondary preven- matrix. These enzymes include matrix metalloproteinases
tion regimen, including smoking cessation, statins, and and certain cathepsins. Multiple mechanisms regulate
agents that target the renin-angiotensin axis. these matrix-degrading proteinases: transcription, trans-

1156 September 19, 2017 Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870


ACS Mechanisms Inform Management

STATE OF THE ART


Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

Figure 1. Four diverse mechanisms cause acute coronary syndromes (ACS).


A, Plaque rupture, also referred to as fissure, traditionally considered the dominant substrate for ACS, usually associates
with both local inflammation, as depicted by the blue monocytes, and systemic inflammation, as indicated by the gauge
showing an increase in blood C-reactive protein (CRP; measured with a high-sensitivity [hsCRP] assay). B, In some cases,
plaque rupture complicates atheromata that do not harbor large collections of intimal macrophages, as identified by optical
coherence tomography criteria, and do not associate with elevations in circulating CRP. Plaque rupture usually provokes
the formation of fibrin-rich red thrombi. C, Plaque erosion appears to account for a growing portion of ACS, often provok-
ing nonST-segmentelevation myocardial infarction. The thrombi overlying patches of intimal erosion generally exhibit
characteristics of white platelet-rich structures. D, Vasospasm can also cause ACS, long recognized as a phenomenon in the
epicardial arteries but also affecting coronary microcirculation.

lation, activation of zymogen precursors, and balance thick collagen fibers, which might increase plaque sta-
with endogenous inhibitors such as the tissue inhibitors bility.25,26 Tregs normally help to maintain homeostasis of
of matrix metalloproteinases or the cystatins. Thus, in- cells involved in adaptive immunity, including antigen-
creased amounts of activated proteinases or reduced lev- presenting cells and effector T cells. Tregs mediate these
els of their corresponding inhibitors can enhance break- modulatory effects by contact-dependent suppression
down of the extracellular matrix of plaque.22 or by releasing anti-inflammatory cytokines such IL-
Adaptive immunity also appears altered in coronary 10 or transforming growth factor-1.27 Patients with
plaque instability.23 Patients with ACS have an increased ACS have a reduced number and suppressive function
population of particularly proinflammatory CD4+ T cells of circulating Tregs compared with patients with stable
characterized by low cell surface expression of CD28, a angina and healthy control subjects.28 The molecular
costimulatory molecule critically involved in determin- mechanisms responsible for the T-cell dysregulation in
ing the outcome of antigen recognition by T cells.24 ACS ACS remain largely unknown. Recent work highlights
also profoundly perturb the numbers of 2 other circulat- the importance of the strength of the stimulation of
ing T-cell subsets: type 17 helper T cells and CD4+CD25+ the T-cell receptor for antigen in the differentiation of
regulatory T cells (Tregs). The net role of interleukin (IL)-17 helper T-cell subsets. The altered activity observed in
in atherosclerosis remains controversial, however; some ACS of proteins involved in the regulation of upstream
experimental studies in mice support a predominantly T-cell receptor for antigen signaling such as CD31 and
proatherogenic function for IL-17, yet type 17 helper protein tyrosine phosphatase N22 may modulate T-cell
T cells activated in plaques promote the formation of functions.29,30

Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1157


Crea and Libby
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

Figure 2. Imbalance in adaptive immune pathways can modulate atherosclerotic plaque activity.
Subsets of T lymphocytes, major participants in adaptive immunity, can either promote local plaque inflammation (effector T
cells) or, in the case of regulatory T cells (Treg), suppress inflammation. Although many pathways regulate T-cell functions, the
markers and mechanisms depicted here illustrate the principle that imbalances in T-cell activities can prevail in plaques. Low
levels of expression of the resurface marker CD31 and high activity of protein tyrosine phosphatase N22 (PTPN22; also known
as Lyp) characterize effector T cells. High levels of activation of CREB (cAMP-responsive element binding protein) character-
ize Tregs that can dampen local adaptive immune responses in the plaque. CCR2 indicates C-C chemokine receptor type 2;
CX3CR1, CX3C chemokine receptor 1; IL, interleukin; IFN-, interferon-; TGF, transforming growth factor-; and TNF, tumor
necrosis factor.

Therapeutic Implications sensitivity CRP in patients in the 7 days after presenta-


tion with ACS.34 A large (n>10 000) phase III trial with
Several anti-inflammatory treatments might provide
benefit in this setting. Colchicine, a time-honored clinical end points that tested a fully humanized anti
anti-inflammatory drug, reduced major cardiovascular IL-1 monoclonal antibody, canakinumab, in patients
events in a medium-sized, unblinded randomized study with previous myocardial infarction and CRP levels >2
in patients with stable ischemic heart disease.31 Two tri- mg/L showed a significant 15% reduction in the prima-
als underway are evaluating colchicine treatment in pa- ry endpoint of MI, stroke, or death, and a 17% fall in
tients with coronary artery disease, although not in the a secondary endpoint that included urgent revascular-
acute phase.32 A phase III trial is testing the potential ization. Indeed, revascularization procedures declined
beneficial effects of low-dose methotrexate in patients 30% in the group that received the interleukin-1 neu-
after myocardial infarction or with coronary artery dis- tralizing antibody canakinumab 150mg subcutaneously
ease and diabetes mellitus or elements of the metabolic every 3 months. This intervention trial substantiates the
syndrome.33 causal contribution of inflammation to atherosclerotic
Targeting proinflammatory cytokines offers an- events.35-37
other approach. Because of the potentially detrimen- T-cell activation signaling offers possible targets. A
tal effects on cardiovascular risk profile or the lack of synthetic CD31-derived peptide can suppresses im-
compelling evidence of benefit in experimental ath- munity in vivo by restoring an inhibitory pathway to a
erosclerosis, monoclonal antibodies that neutralize tu- CD31-negative T-cell subset.38 Moreover, the develop-
mor necrosis factor-, interferon-, or IL-17 may not ment of a novel series of inhibitors of the phosphatase
prove appropriate for treating atherosclerosis. In a small protein tyrosine phosphatase N22 might contribute to
(n=182) phase II study, the IL-1 receptor antagonist a better understanding of its role in immune dysregula-
anakinra reduced the area under the curve of high- tion in patients with ACS.39 Therapeutic strategies that

1158 September 19, 2017 Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870


ACS Mechanisms Inform Management

STATE OF THE ART


target Treg include administration of low doses of IL-2 ity, although the triggers for and effectors of such local
and infusion of autologous Tregs that have undergone inflammation may differ from those that operate in
differentiation and expansion in vitro.40 Various vaccina- patients with systemic inflammation.43 Although many
tion strategies also can limit experimental atherogen- studies have clarified the molecular mechanisms lead-
esis and may act by augmenting the activity of Treg.41 ing to coronary instability in individuals with systemic
inflammation (eg, gauged by CRP elevations), patients
without systemic inflammation have undergone less
PLAQUE RUPTURE WITHOUT extensive investigation. The precise causes of instability
SYSTEMIC INFLAMMATION remain poorly understood, providing a strong stimulus
for further research.
Mechanisms The possible relationship between psychological
When plaque rupture occurs in the absence of systemic stress and plaque rupture may relate to sympathetic
inflammatory activation, other mechanisms may con- nervous system activation and catecholamine release
tribute to pathogenesis, including extreme emotional associated with an increase in heart rate, blood pres-
disturbance ranging from external events of short dura- sure, and coronary vasoconstriction favoring plaque dis-
tion such as earthquakes and a beloved teams loss of a ruption and platelet activation, hypercoagulability, and
football (soccer) match to acute manifestations of more intense coronary microvascular constriction.44 More-
long-term emotional disturbances (Figure 1B). Intense over, 3-adrenergic stimulation can stimulate release
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

physical exertion and local mechanical stress at the level from the bone marrow of proinflammatory monocytes
of the artery wall, either increased circumferential stress that can home to experimental atheromata and amplify
or reduced shear stress, may also predispose to plaque local inflammation.45 Although physical or emotional
rupture.42 In addition, subclinical inflammation in the stress per se may not suffice to cause coronary throm-
microenvironment of the culprit stenosis might com- bosis, it might trigger instability in plaques already pre-
pound the chain of events leading to coronary instabil- disposed to provoke events.46

Figure 3. Cholesterol crystals activate local innate immune pathways in the atherosclerotic plaque.
Plasma low-density lipoprotein can enter the arterial wall and accumulate in mononuclear phagocytes via scavenger receptors.
Lipid-laden macrophage foam cells can die, contributing to the accumulation of extracellular cholesteryl ester and cholesterol
monohydrate crystals in the lipid-rich necrotic core of the plaque. The dying macrophages can also release apoptotic bodies
and microparticles that contain the potent procoagulant tissue factor (TF+). The cholesterol crystals can coactivate the inflam-
masome, an intracellular supramolecular structure that generates the biologically active forms of the proinflammatory cyto-
kines interleukin (IL)-1 and IL-18. Large crystals might also cause mechanical disruption of the fibrous cap.

Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1159


Crea and Libby

Local changes in the equilibrium between esterified the fibrous cap of plaque can commonly cause ACS.63
and free cholesterol might promote plaque rupture47 (Fig- The mechanism of plaque thrombosis described by pa-
ure3). Cholesterol crystal formation in the lipid core could thologists as superficial erosion appears not to involve
heighten the risk of plaque rupture and thrombosis and inflammation mediated by macrophages, as in the case
can coactivate the inflammasome, an intracellular multi- of fibrous cap fracture (Figure1C). Superficial erosion
meric complex that generates active IL-1 and IL-18.48 complicates lesions with a distinct epidemiology and
OCT has colocalized cholesterol crystals in human morphology and involves pathophysiological mecha-
coronary arteries with thin-capped plaques.49 The devel- nisms that differ from rupture of the fibrous cap. In-
opment of micro-OCT with a 2-m resolution may shed deed, neutrophil activation seems to play a pivotal role
new light on this potential mechanism of instability by in thrombosis due to plaque erosion.4 Patients who
assessing its occurrence in vivo.50 Cholesterol crystals presented with ACS associated with plaque erosion
might also theoretically resist plaque rupture by increas- defined by OCT had higher systemic myeloperoxidase
ing the stiffness of the lipid pool, although this mecha- levels compared with patients with plaque rupture.
nism probably contributes little to plaque stability.51 The Moreover, in postmortem coronary artery specimens,
role of calcium mineral as a causal factor in ACS remains luminal thrombi superimposed on eroded plaques con-
controversial. Large collections of calcified tissue do not tain many more myeloperoxidase-positive cells than
augment the biomechanical instability of plaques or as- thrombi associated with ruptured plaques64 (Figure4).
sociate with lesions that provoke ACS.52,53 Small foci of Autopsy studies suggest that fatal plaque erosion as-
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

calcification within the atherosclerotic intima, some caus- sociates more commonly with hypertriglyceridemia, dia-
ing spotty calcification visible on computed tomography, betes mellitus, female sex, and old age.65 The morphology
can also introduce biomechanical inhomogeneities that of lesions that produce thrombosis caused by superficial
can favor plaque destabilization.3,54,55 erosion differs radically from the features associated with
fibrous cap rupture. Superficially eroded lesions contain
few macrophages or T lymphocytes, inflammatory cells
Therapeutic Implications considered pathogenic in plaque rupture. Rather than
Because it is difficult to limit environmental, physical, depleted collagen, the lesions associated with superficial
or emotional triggers, the altered plaque characteristics erosion contain abundant proteoglycan and glycosami-
produced by intensive lipid-lowering treatment might noglycans. Instead of a paucity of SMCs, eroded lesions
protect against this form of plaque destabilization.56 In can contain abundant arterial SMCs. Plaques complicat-
particular, statins and ezetimibe may interfere with cho- ed by fibrous cap fracture typically harbor large necrotic,
lesterol crystal formation.57,58 Cyclodextrin can solubilize lipid-rich cores. In contrast, the more fibrous lesions asso-
cholesterol, limits experimental atherogenesis, and thus ciated with superficial erosion generally lack prominent
might provide a pharmacological approach to combat- central lipid cores.66 A loss of intimal endothelial cells de-
ting cholesterol crystal accumulation in plaques.59 fines lesions that provoke thrombosis by the mechanism
Enhancement of cholesterol efflux might promote denoted as superficial erosion.
plaque stabilization, but strategies to enhance this process Recent work has implicated engagement of the innate
by manipulation of high-density lipoprotein concentra- immune receptor Toll-like receptor 2 in promoting the
tions or by administration of apolipoprotein A-1 have gen- susceptibility of endothelial cells to apoptotic stimuli. Hy-
erally failed to confer clinical benefit.60 A large outcome aluronic acid, a prominent constituent of the extracellular
trial with anacetrapib produced a modest event reduction, matrix of eroded lesions, could serve as an endogenous
but since this agent not only raises high-density lipopro- ligand of Toll-like receptor 2, implicated in promoting
tein but also lowers low-density lipoprotein renders it dif- endothelial cell apoptosis.67 Experiments that have evalu-
ficult to attribute event reduction to raising high-density ated gain and loss of Toll-like receptor 2 function in mice
lipoprotein.61 In addition, the inhibition of cholesteryl ester support these observations.68 Indeed, hyaluronan and its
hydrolase, an enzyme that converts cholesteryl esters to receptor, CD44, colocalize along the plaque/thrombus
free cholesterol, might prevent cholesterol crystallization. interface in eroded plaque but not in ruptured or stable
Inhibition of acetyl-CoA acetyltransferase-1, which in con- plaque.69 Accumulation of hyaluronan and expression
trast promotes cholesterol crystallization, increased ath- of CD44 along the plaque/thrombus interface of erod-
eroma volume and major cardiovascular events.62 ed plaques may promote de-endothelization, resulting
in CD44-dependent platelet adhesion and subsequent
thrombus formation, mediated in part by a direct action
PLAQUE EROSION of hyaluronan on fibrin polymerization.70,71
We have hypothesized that thrombosis caused by su-
Mechanisms perficial erosion occurs in 2 phases. The first hit, mediat-
Current evidence suggests that mechanisms of plaque ed, for example, by Toll-like receptor 2, would jeopardize
disruption distinct from macrophage-driven rupture of endothelial viability or adherence, leading to a breach in

1160 September 19, 2017 Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870


ACS Mechanisms Inform Management

STATE OF THE ART


from the blood compartment by the endothelial lining of
the intima. The second hit, mediated by the release of
granular contents from activated platelets and the local
production of chemoattractants for polymorphonuclear
leukocytes, would beckon these granulocytes to join the
platelets at local sites of endothelial denudation. In turn,
the activation of polymorphonuclear leukocytes could
generate structures known as neutrophil extracellular
traps, made up of strands of unwound DNA released by
the dying granulocytes.72 This uncoiled DNA becomes
decorated with proteases, tissue factor, and pro-oxidant
enzymes that could propagate local amplification of an
innate immune response, thrombin activation, and fibrin
generation and entrap further platelets and fibrin strands
in an evolving thrombus.51
This hypothetical scenario posits a mechanism of
thrombosis that is independent of the chronic inflam-
matory cells typically implicated in fibrous cap rupture:
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

macrophages and T lymphocytes. Rather, this schema


implicates different arms of innate immunity in throm-
bus initiation, propagation, and stabilization. These
mechanistic differences with fibrous cap fracture have
potential therapeutic implications.

Therapeutic Implications
Because statins alter the lipid profile primarily by low-
ering low-density lipoprotein rather than triglycerides,
therapies that address triglyceride-rich lipoproteins may
address the residual risk of events resulting from erosion
that persists in the statin era more effectively than more
intense low-density lipoproteinlowering therapies.73
Moreover, therapies that destabilize neutrophil extra-
Figure 4. Pathways implicated in arterial thrombosis
cellular traps such as deoxyribonuclease administration
caused by superficial erosion.
might limit the propagation of thrombi resulting from
Stimuli such as disturbed flow or engagement of innate im-
mune receptors such as Toll-like receptor-2 (TLR2) can activate superficial erosion.74 Receptors such as CD44 expressed
the endothelial cells that line the arterial intima. These cells can by neutrophils may bind hyaluronan and glycosamino-
undergo cell death, eg, by apoptosis, depicted by the cell with glycan enriched in plaques complicated by erosion and
the interrupted membrane and pyknotic nucleus. Injured or dy- furnish a potential therapeutic target.75
ing endothelial cells can desquamate, uncovering the basement Jia et al76 recently reported a series of patients with
membrane. Neutrophils attracted by chemokines produced ACS with OCT-defined erosion and treated with anti-
by activated endothelial cells can congregate on the denuded coagulant/antiplatelet therapy rather that mechanical
intima and can in turn degranulate and die, releasing neutro- revascularization. About 80% of the patients showed
phil extracellular traps (NETs). These strands of extruded DNA a >50% reduction of thrombus volume after 1 month.
can bind the contents of the neutrophil granules and other
More than a third of patients lacked detectable thrombus
proteins, eg, myeloperoxidase or tissue factor. The NETs can
constitute a solid-state reactor that generates oxidants such as
after 1 month. This study suggests that pharmacological
hypochlorous acid and stimulates coagulation locally. Platelets rather than interventional treatment can effectively man-
interacting with the basement membrane can activate, release age ACS caused by superficial erosion, a proposition that
their granular contents, including chemokines that can recruit merits and requires rigorous testing in outcome trials.77
further leukocytes, and form the nidus of a white thrombus.

the integrity of the endothelial monolayer overlying the PLAQUE WITHOUT THROMBUS
atherosclerotic plaque. The denuded patch on the intimal
surface would then attract platelets that could undergo Mechanisms
activation by contact with collagen and other compo- In patients with ACS without plaque thrombus, a func-
nents of the arterial extracellular matrix usually protected tional alteration of coronary circulation likely causes

Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1161


Crea and Libby

acute ischemia involving large epicardial coronary higher prevalence of coronary spasm than whites for
arteries or the coronary microcirculation (Figure 1D). unknown reasons.
Epicardial coronary vasospasm may occur in patients Microvascular spasm also can cause myocardial isch-
in whom coronary angiography does not demonstrate emia.83 This mechanism likely operates in patients with
an obstructive atherosclerotic plaque.78 In the CASPAR Takotsubo cardiomyopathy,84 which frequently occurs
study (Coronary Artery Spasm in Patients With Acute in the absence of obstructive atherosclerosis, although
Coronary Syndrome), coronary angiography failed to 15% of these patients exhibit concomitant obstructive
show culprit lesions in 30% of patients with sus- atherosclerosis.85 Microvascular spasm can also contrib-
pected ACS.79 Intracoronary acetylcholine administra- ute to ischemia in the face of angiographically normal-
tion elicited coronary spasm in nearly 50% of these appearing epicardial coronary arteries. Microvascular
patients. Similar findings pertained to a Japanese ischemia often manifests as angina pectoris but can
population.80 Coronary artery spasm also may cause also cause nonST-segmentelevation ACS as defined
coronary instability in patients with obstructive ath- by electrocardiographic alterations and biomarker evi-
erosclerosis. Indeed, a classic study by Bertrand et al81 dence of myocyte injury.86 Coronary spasm can also
found that ergonovine induced spasm in 20% of pa- contribute to plaque instability by causing endothelial
tients with recent myocardial infarction and in 40% of damage, as shown in an infarction-prone strain of the
patients with unstable angina. In a more recent study, Watanabe heritable hyperlipidemic rabbit.44
acetylcholine induced spasm in 20% of white patients Either macrovascular or microvascular spasm can
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

and in 60% of patients with myocardial infarction result from impaired vasodilatation or from vasocon-
within the previous 14 days.82 Japanese people have a strictor stimuli acting on hyperreactive vascular SMCs

Figure 5. Cellular mechanisms of arterial epicardial spasm relevant to acute coronary syndrome pathogenesis.
Smooth muscle cells in the media layer of coronary arterial vessels can contract in response to stimuli from the autonomic ner-
vous system (eg, acetylcholine), local responses to autacoids (eg, histamine), and pharmacological stimuli. Local hyperreactivity
of smooth muscle cells mediated mainly by enhanced Rho kinase activity results in spasm in response to constrictor stimuli.
Normal endothelial cells produce endogenous vasodilator substances, including nitric oxide (NO). A large body of literature
supports the contribution of dysfunctional endothelium to inappropriate constriction of coronary and other arteries.

1162 September 19, 2017 Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870


ACS Mechanisms Inform Management

STATE OF THE ART


(Figure 5).87,88 Shimokawa et al89 demonstrated SMC for fibrous cap rupture or of a minority mechanism:
hyperreactivity in a coronary segment in pigs after superficial erosion. We and others have championed
adventitial exposure of a coronary segment to an in- and explored the role of inflammation as a pathway
flammatory stimulus (IL-1). Subsequent experimental to the ACS.
and clinical data suggested that an increase in Rho- However, we now have a greater appreciation that
kinase activity was a major mechanism of SMC hy- not all cases of ACS fit this mold. To move forward, we
perreactivity.9092 Perivascular fibrosis in the coronary need to remain vigilant not to cling to our cherished no-
microvasculature can contribute to myocardial isch- tions and to remain open to shifts in the nature of the
emia.93 This observation highlights the potential contri- diseases we treat and to new vistas opened by scientific
bution of fixed structural changes in coronary arterioles advances. As discussed here, we need to consider more
beyond impaired endothelial vasodilator function as a deeply in the current era arterial spasm, rupture with-
contributor to myocardial ischemia in the absence of out inflammation, and distinct aspects of innate im-
obstructive disease of the epicardial coronary arteries. munity or dyslipidemia as instigators of plaque erosion.
The complexity of clinical ACS surpasses our traditional
categorization of this condition. We should evolve be-
Therapeutic Implications yond the current constrained concepts by expanding
Several vasoconstrictor substances implicated as trig- our understanding.
gers of epicardial coronary spasm may cause ischemia in We have achieved considerable mastery of plaque
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

the same patient under different conditions.94 Accord- rupture with inflammation from a mechanistic and
ingly, the vasoactive substances themselves may not re- therapeutic viewpoint. Control of traditional risk fac-
spond to therapeutic targeting. The current treatment tors such as hypercholesterolemia, hypertension, and
of epicardial spasm uses nonspecific vasodilators such smoking addresses the pathophysiology of this mecha-
as long-acting nitrates and calcium channel blockers.95 nism of ACS. The other categories that we discuss here
Nitrates and calcium channel antagonists may alleviate may account for an increasing proportion of ACS, now
ischemia caused by vasospasm. However, we need to that we are addressing more effectively the traditional
identify the molecular alterations responsible for SMC risk factors.5 We should remain open to the possibility,
hyperreactivity because many patients do not respond even likelihood, that tailored therapeutic approaches
satisfactorily to standard doses of vasodilators.96 Knowl- may apply to the prevention and treatment of the less
edge of the postreceptor mechanisms responsible for well-recognized pathways to acute myocardial ischemia
SMC hyperreactivity and epicardial coronary spasm discussed here.
might enable the development of new therapeutic op- We must aim to link the clinical presentation of
tions for patients who present poor clinical response to ACS more tightly to pathophysiological mechanisms.
nonspecific vasodilator therapy. Fasudil, a Rho kinase More than a century after the introduction of ECG, we
inhibitor, reduces the rate of coronary spasm episodes still triage our patients with ACS on the basis of this
in patients with vasospastic angina.97 Similarly, further mature technology. To move forward in the manage-
efforts are warranted to unravel the molecular mecha- ment of ACS, we should strive for a more personal-
nisms responsible for coronary microvascular dysfunc- ized approach to therapy based on criteria more tightly
tion in Takotsubo cardiomyopathy and in ischemia with linked to the diverse pathophysiological mechanisms
normal coronary arteries.20 The observation that Tako than ECG repolarization abnormalities. We need to
tsubo cardiomyopathy may have worse outcomes than develop and validate soluble and imaging biomarkers
previously considered and the challenges of managing that reflect the underlying mechanism that yields acute
patients with episodic ischemia caused by microvascu- ischemia. Point-of-care assessment of such biomarkers
lar dysfunction render this quest urgent.85 Preventing would help render their use clinically practical in the
or reversing perivascular fibrosis in the coronary arterial triage of patients who present with ACS, with the goal
microvasculature represents another novel target be- of sparing some the need for urgent invasive diagnos-
yond traditional antiatherosclerotic therapies. tic or therapeutic measures.18,67,98 We then need rig-
orous clinical evaluation of targeted therapies guided
by a more precise pathophysiological classification of
CONCLUSIONS AND PERSPECTIVES ACS that reaches beyond our current dichotomous ap-
A dichotomous approach based on the ECG has tradi- proach based on the ECG. If these ventures succeed,
tionally dominated our clinical management of ACS: we may one day look back on todays treatment of ACS
ST-segmentmyocardial infarction versus nonST- as oversimplified and rather primitive. We should strive
segmentmyocardial infarction. Our thinking concern- as a community not to be satisfied with our current ap-
ing the pathophysiological bases of the ACS has also proach to ACS management but reach to realize the
evolved separately and, in parallel, has been dominated promise of a more personalized precision deployment
by the concept of the vulnerable plaque as a substrate of treatment strategies, including new ones based on

Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1163


Crea and Libby

greater mechanistic understanding of the diverse un- D, Maseri A; FAMI Study Investigators. High-sensitivity C-reactive protein
is within normal levels at the very onset of first ST-segment elevation acute
derlying causes of acute myocardial ischemia. myocardial infarction in 41% of cases: a multiethnic case-control study.
J Am Coll Cardiol. 2011;58:26542661. doi: 10.1016/j.jacc.2011.08.055.
15. Scalone G, Niccoli G, Refaat H, Vergallo R, Porto I, Leone AM, Burzotta F,
DISCLOSURES DAmario D, Liuzzo G, Fracassi F, Trani C, Crea F. Not all plaque ruptures
are born equal: an optical coherence tomography study [published online
Dr Libby reports receiving grants from Novartis and holding ahead of print December 24, 2016]. Eur Heart J Cardiovasc Imaging. doi:
scientific advisory board positions for Olatec and Interleukin 10.1093/ehjci/jew208.
16. Arbustini E, Dal Bello B, Morbini P, Burke AP, Bocciarelli M, Specchia G,
Genetics (company no longer operating). Dr Crea reports no
Virmani R. Plaque erosion is a major substrate for coronary thrombosis in
conflicts. acute myocardial infarction. Heart. 1999;82:269272.
17. Bentzon JF, Otsuka F, Virmani R, Falk E. Mechanisms of plaque forma-
tion and rupture. Circ Res. 2014;114:18521866. doi: 10.1161/CIRCRE-
AFFILIATIONS SAHA.114.302721.
18. Niccoli G, Montone RA, Cataneo L, Cosentino N, Gramegna M, Refaat
From Department of Cardiovascular and Thoracic Sciences, H, Porto I, Burzotta F, Trani C, Leone AM, Severino A, Crea F. Morpho-
Catholic University, Rome, Italy (F.C.); and Division of logical-biohumoral correlations in acute coronary syndromes: patho-
genetic implications. Int J Cardiol. 2014;171:463466. doi: 10.1016/j.
Cardiovascular Medicine, Department of Medicine, Brigham
ijcard.2013.12.238.
and Womens Hospital, Harvard Medical School, Boston, 19. Jia H, Abtahian F, Aguirre AD, Lee S, Chia S, Lowe H, Kato K, Yo-
MA (P.L.). netsu T, Vergallo R, Hu S, Tian J, Lee H, Park SJ, Jang YS, Raffel OC,
Mizuno K, Uemura S, Itoh T, Kakuta T, Choi SY, Dauerman HL, Prasad
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

A, Toma C, McNulty I, Zhang S, Yu B, Fuster V, Narula J, Virmani R,


Jang IK. In vivo diagnosis of plaque erosion and calcified nodule in
FOOTNOTES patients with acute coronary syndrome by intravascular optical coherence
Circulation is available at http://circ.ahajournals.org. tomography. J Am Coll Cardiol. 2013;62:17481758. doi: 10.1016/j.
jacc.2013.05.071.
20. Crea F, Liuzzo G. Pathogenesis of acute coronary syndromes. J Am Coll
Cardiol. 2013;61:111. doi: 10.1016/j.jacc.2012.07.064.
REFERENCES 21. Liuzzo G, Biasucci LM, Gallimore JR, Grillo RL, Rebuzzi AG, Pepys MB,
Maseri A. The prognostic value of C-reactive protein and serum amyloid A
1. Libby P. Molecular bases of the acute coronary syndromes. Circulation.
protein in severe unstable angina. N Engl J Med. 1994;331:417424. doi:
1995;91:28442850.
10.1056/NEJM199408183310701.
2. Libby P. Current concepts of the pathogenesis of the acute coronary syn-
22. Schnbeck U, Mach F, Sukhova GK, Murphy C, Bonnefoy JY, Fabunmi
dromes. Circulation. 2001;104:365372.
RP, Libby P. Regulation of matrix metalloproteinase expression in human
3. Libby P. Mechanisms of acute coronary syndromes and their implications
vascular smooth muscle cells by T lymphocytes: a role for CD40 signaling
for therapy. N Engl J Med. 2013;368:20042013. doi: 10.1056/NEJM-
in plaque rupture? Circ Res. 1997;81:448454.
ra1216063.
23. Liuzzo G, Kopecky SL, Frye RL, OFallon WM, Maseri A, Goronzy JJ, Wey-
4. Libby P, Pasterkamp G. Requiem for the vulnerable plaque. Eur Heart J.
and CM. Perturbation of the T-cell repertoire in patients with unstable
2015;36:29842987. doi: 10.1093/eurheartj/ehv349.
angina. Circulation. 1999;100:21352139.
5. Pasterkamp G, den Ruijter HM, Libby P. Temporal shifts in clinical pre-
24. Liuzzo G, Goronzy JJ, Yang H, Kopecky SL, Holmes DR, Frye RL, Weyand
sentation and underlying mechanisms of atherosclerotic disease. Nat Rev
CM. Monoclonal T-cell proliferation and plaque instability in acute coro-
Cardiol. 2017;14:2129. doi: 10.1038/nrcardio.2016.166.
6. Davies MJ, Thomas A. Thrombosis and acute coronary-artery lesions in nary syndromes. Circulation. 2000;101:28832888.
sudden cardiac ischemic death. N Engl J Med. 1984;310:11371140. doi: 25. Chen S, Crother TR, Arditi M. Emerging role of IL-17 in atherosclerosis.
10.1056/NEJM198405033101801. J Innate Immun. 2010;2:325333. doi: 10.1159/000314626.
7. Falk E, Shah PK, Fuster V. Coronary plaque disruption. Circulation. 26. Gister A, Robertson AK, Andersson J, Ketelhuth DF, Ovchinnikova O,
1995;92:657671. Nilsson SK, Lundberg AM, Li MO, Flavell RA, Hansson GK. Transforming
8. Finn AV, Nakano M, Narula J, Kolodgie FD, Virmani R. Concept of vulnera- growth factor- signaling in T cells promotes stabilization of atheroscle-
ble/unstable plaque. Arterioscler Thromb Vasc Biol. 2010;30:12821292. rotic plaques through an interleukin-17-dependent pathway. Sci Transl
doi: 10.1161/ATVBAHA.108.179739. Med. 2013;5:196ra100. doi: 10.1126/scitranslmed.3006133.
9. Motoyama S, Ito H, Sarai M, Kondo T, Kawai H, Nagahara Y, Harigaya H, Kan 27. Brusko TM, Putnam AL, Bluestone JA. Human regulatory T cells: role
S, Anno H, Takahashi H, Naruse H, Ishii J, Hecht H, Shaw LJ, Ozaki Y, Narula J. in autoimmune disease and therapeutic opportunities. Immunol Rev.
Plaque characterization by coronary computed tomography angiography 2008;223:371390. doi: 10.1111/j.1600-065X.2008.00637.x.
and the likelihood of acute coronary events in mid-term follow-up. J Am 28. Liuzzo G, Montone RA, Gabriele M, Pedicino D, Giglio AF, Trotta F, Galiffa
Coll Cardiol. 2015;66:337346. doi: 10.1016/j.jacc.2015.05.069. VA, Previtero M, Severino A, Biasucci LM, Crea F. Identification of unique
10. Mann J, Davies MJ. Mechanisms of progression in native coronary artery adaptive immune system signature in acute coronary syndromes. Int J Car-
disease: role of healed plaque disruption. Heart. 1999;82:265268. diol. 2013;168:564567. doi: 10.1016/j.ijcard.2013.01.009.
11. Stone GW, Maehara A, Lansky AJ, de Bruyne B, Cristea E, Mintz GS, Meh- 29. Flego D, Severino A, Trotta F, Previtero M, Ucci S, Zara C, Pedicino D, Mas-
ran R, McPherson J, Farhat N, Marso SP, Parise H, Templin B, White R, saro G, Biasucci LM, Liuzzo G, Crea F. Altered CD31 expression and activity
Zhang Z, Serruys PW; PROSPECT Investigators. A prospective natural-his- in helper T cells of acute coronary syndrome patients. Basic Res Cardiol.
tory study of coronary atherosclerosis. N Engl J Med. 2011;364:226235. 2014;109:448. doi: 10.1007/s00395-014-0448-3.
doi: 10.1056/NEJMoa1002358. 30. Flego D, Severino A, Trotta F, Previtero M, Ucci S, Zara C, Massaro G,
12. Kubo T, Maehara A, Mintz GS, Doi H, Tsujita K, Choi SY, Katoh O, Nasu K, Pedicino D, Biasucci LM, Liuzzo G, Crea F. Increased PTPN22 expression
Koenig A, Pieper M, Rogers JH, Wijns W, Bse D, Margolis MP, Moses JW, and defective CREB activation impair regulatory T-cell differentiation in
Stone GW, Leon MB. The dynamic nature of coronary artery lesion mor- non-ST-segment elevation acute coronary syndromes. J Am Coll Cardiol.
phology assessed by serial virtual histology intravascular ultrasound tissue 2015;65:11751186. doi: 10.1016/j.jacc.2015.01.027.
characterization. J Am Coll Cardiol. 2010;55:15901597. doi: 10.1016/j. 31. Nidorf SM, Eikelboom JW, Budgeon CA, Thompson PL. Low-dose colchi-
jacc.2009.07.078. cine for secondary prevention of cardiovascular disease. J Am Coll Cardiol.
13. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circula- 2013;61:404410. doi: 10.1016/j.jacc.2012.10.027.
tion. 2002;105:11351143. 32. Tardif J-C, LAllier P. Colchicine Cardiovascular Outcomes Trial (COL-
14. Cristell N, Cianflone D, Durante A, Ammirati E, Vanuzzo D, Banfi M, Calori COT). https://clinicaltrials.gov/ct2/show/NCT02551094. 2017. Accessed
G, Latib A, Crea F, Marenzi G, De Metrio M, Moretti L, Li H, Uren NG, Hu June 1, 2017.

1164 September 19, 2017 Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870


ACS Mechanisms Inform Management

STATE OF THE ART


33. Everett BM, Pradhan AD, Solomon DH, Paynter N, Macfadyen J, Zaharris idemic rabbits. Arterioscler Thromb Vasc Biol. 2013;33:25182523. doi:
E, Gupta M, Clearfield M, Libby P, Hasan AA, Glynn RJ, Ridker PM. Ra- 10.1161/ATVBAHA.113.301303.
tionale and design of the Cardiovascular Inflammation Reduction Trial: 45. Libby P, Nahrendorf M, Swirski FK. Leukocytes link local and systemic in-
a test of the inflammatory hypothesis of atherothrombosis. Am Heart J. flammation in ischemic cardiovascular disease: an expanded cardiovas-
2013;166:199207.e15. doi: 10.1016/j.ahj.2013.03.018. cular continuum. J Am Coll Cardiol. 2016;67:10911103. doi: 10.1016/j.
34. Morton AC, Rothman AM, Greenwood JP, Gunn J, Chase A, Clarke B, Hall jacc.2015.12.048.
AS, Fox K, Foley C, Banya W, Wang D, Flather MD, Crossman DC. The 46. Burke AP, Farb A, Malcom GT, Liang Y, Smialek JE, Virmani R. Plaque rup-
effect of interleukin-1 receptor antagonist therapy on markers of inflam- ture and sudden death related to exertion in men with coronary artery
mation in non-ST elevation acute coronary syndromes: the MRC-ILA Heart disease. JAMA. 1999;281:921926.
Study. Eur Heart J. 2015;36:377384. doi: 10.1093/eurheartj/ehu272. 47. Janoudi A, Shamoun FE, Kalavakunta JK, Abela GS. Cholesterol crystal in-
35. Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1 inhibition and the duced arterial inflammation and destabilization of atherosclerotic plaque.
prevention of recurrent cardiovascular events: rationale and design of the Eur Heart J. 2016;37:19591967. doi: 10.1093/eurheartj/ehv653.
Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). 48. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG,
Am Heart J. 2011;162:597605. doi: 10.1016/j.ahj.2011.06.012. Abela GS, Franchi L, Nuez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA,
36. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne Rock KL, Moore KJ, Wright SD, Hornung V, Latz E. NLRP3 inflammasomes
C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH, are required for atherogenesis and activated by cholesterol crystals. Na-
Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z, ture. 2010;464:13571361. doi: 10.1038/nature08938.
Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi 49. Nakamura S, Inami S, Murai K, Takano M, Takano H, Asai K, Yasutake
PRF, Troquay RPT, Libby P, Glynn RJ and Group CT. Antiinflammatory M, Shimizu W, Mizuno K. Relationship between cholesterol crystals and
Therapy with Canakinumab for Atherosclerotic Disease [published on- culprit lesion characteristics in patients with stable coronary artery disease:
line ahead of print August 27, 2017]. N Engl J Med. 2017. doi: 10.1056/ an optical coherence tomography study. Clin Res Cardiol. 2014;103:1015
NEJMoa1707914. 1021. doi: 10.1007/s00392-014-0748-5.
37. Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P, Glynn RJ, Ridker P, 50. Liu L, Gardecki JA, Nadkarni SK, Toussaint JD, Yagi Y, Bouma BE, Tearney
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

Lorenzatti A, Krum H, Varigos J, Siostrzonek P, Sinnaeve P, Fonseca F, Nico- GJ. Imaging the subcellular structure of human coronary atherosclerosis
lau J, Gotcheva N, Genest J, Yong H, Urina-Triana M, Milicic D, Cifkova R, using micro-optical coherence tomography. Nat Med. 2011;17:1010
Vettus R, Koenig W, Anker SD, Manolis AJ, Wyss F, Forster T, Sigurdsson A, 1014. doi: 10.1038/nm.2409.
Pais P, Fucili A, Ogawa H, Shimokawa H, Veze I, Petrauskiene B, Salvador L, 51. Loree HM, Tobias BJ, Gibson LJ, Kamm RD, Small DM, Lee RT. Mechanical
Kastelein J, Cornel JH, Klemsdal TO, Medina F, Budaj A, Vida-Simiti L, Koba- properties of model atherosclerotic lesion lipid pools. Arterioscler Thromb.
lava Z, Otasevic P, Pella D, Lainscak M, Seung K-B, Commerford P, Dellborg
1994;14:230234.
M, Donath M, Hwang J-J, Kultursay H, Flather M, Ballantyne C, Bilazarian S,
52. Huang H, Virmani R, Younis H, Burke AP, Kamm RD, Lee RT. The impact
Chang W, East C, Everett B, Forgosh L, Glynn R, Harris B, Libby P, Ligueros
of calcification on the biomechanical stability of atherosclerotic plaques.
M, Thuren T, Bohula E, Charmarthi B, Cheng S, Chou S, Danik J, McMahon
Circulation. 2001;103:10511056.
G, Maron B, Ning M, Olenchock B, Pande R, Perlstein T, Pradhan A, Rost N,
53. Virmani R, Joner M, Sakakura K. Recent highlights of ATVB: calcification.
Singhal A, Taqueti V, Wei N, Burris H, Cioffi A, Dalseg AM, Ghosh N, Gralow
Arterioscler Thromb Vasc Biol. 2014;34:13291332. doi: 10.1161/ATVBA-
J, Mayer T, Rugo H, Fowler V, Limaye AP, Cosgrove S, Levine D, Lopes R,
HA.114.304000.
Scott J, Thuren T, Ligueros M, Hilkert R, Tamesby G, Mickel C, Manning B,
54. Ruiz JL, Weinbaum S, Aikawa E, Hutcheson JD. Zooming in on the genesis
Woelcke J, Tan M, Manfreda S, Ponce T, Kam J, Saini R, Banker K, Salko T,
of atherosclerotic plaque microcalcifications. J Physiol. 2016;594:2915
Nandy P, Tawfik R, O'Neil G, Manne S, Jirvankar P, Lal S, Nema D, Jose J,
2927. doi: 10.1113/JP271339.
Collins R, Bailey K, Blumenthal R, Colhoun H, Gersh B and Glynn RJ. Effect
55. Camici PG, Rimoldi OE, Gaemperli O, Libby P. Non-invasive anatomic and
of interleukin-1 inhibition with canakinumab on incident lung cancer in
functional imaging of vascular inflammation and unstable plaque. Eur
patients with atherosclerosis: exploratory results from a randomised, dou-
Heart J. 2012;33:13091317. doi: 10.1093/eurheartj/ehs067.
ble-blind, placebo-controlled trial [published online ahead of print August
56. Libby P. How does lipid lowering prevent coronary events? New insights
25, 2017]. The Lancet. doi: 10.1016/S0140-6736(17)32247-X.
from human imaging trials. Eur Heart J. 2015;36:472474. doi: 10.1093/
38. Fornasa G, Clement M, Groyer E, Gaston AT, Khallou-Laschet J, Morvan
eurheartj/ehu510.
M, Guedj K, Kaveri SV, Tedgui A, Michel JB, Nicoletti A, Caligiuri G. A
57. Abela GS, Vedre A, Janoudi A, Huang R, Durga S, Tamhane U. Effect
CD31-derived peptide prevents angiotensin II-induced atherosclerosis pro-
gression and aneurysm formation. Cardiovasc Res. 2012;94:3037. doi: of statins on cholesterol crystallization and atherosclerotic plaque sta-
10.1093/cvr/cvs076. bilization. Am J Cardiol. 2011;107:17101717. doi: 10.1016/j.amj-
39. Stanford SM, Krishnamurthy D, Falk MD, Messina R, Debnath B, Li S, Liu card.2011.02.336.
T, Kazemi R, Dahl R, He Y, Yu X, Chan AC, Zhang ZY, Barrios AM, Woods 58. Patel R, Janoudi A, Vedre A, Aziz K, Tamhane U, Rubinstein J, Abela
VL Jr, Neamati N, Bottini N. Discovery of a novel series of inhibitors of lym- OG, Berger K, Abela GS. Plaque rupture and thrombosis are reduced
phoid tyrosine phosphatase with activity in human T cells. J Med Chem. by lowering cholesterol levels and crystallization with ezetimibe and are
2011;54:16401654. doi: 10.1021/jm101202j. correlated with fluorodeoxyglucose positron emission tomography. Ar-
40. Putnam AL, Brusko TM, Lee MR, Liu W, Szot GL, Ghosh T, Atkinson MA, terioscler Thromb Vasc Biol. 2011;31:20072014. doi: 10.1161/ATVBA-
Bluestone JA. Expansion of human regulatory T-cells from patients with HA.111.226167.
type 1 diabetes. Diabetes. 2009;58:652662. doi: 10.2337/db08-1168. 59. Zimmer S, Grebe A, Bakke SS, Bode N, Halvorsen B, Ulas T, Skjelland M,
41. Shah PK, Chyu KY, Dimayuga PC, Nilsson J. Vaccine for atherosclerosis. De Nardo D, Labzin LI, Kerksiek A, Hempel C, Heneka MT, Hawxhurst V,
J Am Coll Cardiol. 2014;64:27792791. doi: 10.1016/j.jacc.2014.10.018. Fitzgerald ML, Trebicka J, Bjrkhem I, Gustafsson J, Westerterp M, Tall
42. Koskinas KC, Sukhova GK, Baker AB, Papafaklis MI, Chatzizisis YS, Coskun AR, Wright SD, Espevik T, Schultze JL, Nickenig G, Ltjohann D, Latz E.
AU, Quillard T, Jonas M, Maynard C, Antoniadis AP, Shi GP, Libby P, Edel- Cyclodextrin promotes atherosclerosis regression via macrophage repro-
man ER, Feldman CL, Stone PH. Thin-capped atheromata with reduced gramming. Sci Transl Med. 2016;8:333ra50. doi: 10.1126/scitranslmed.
collagen content in pigs develop in coronary arterial regions exposed to aad6100.
persistently low endothelial shear stress. Arterioscler Thromb Vasc Biol. 60. Mohammadpour AH, Akhlaghi F. Future of cholesteryl ester transfer pro-
2013;33:14941504. doi: 10.1161/ATVBAHA.112.300827. tein (CETP) inhibitors: a pharmacological perspective. Clin Pharmacokinet.
43. Chatzizisis YS, Baker AB, Sukhova GK, Koskinas KC, Papafaklis MI, Beigel 2013;52:615626. doi: 10.1007/s40262-013-0071-8.
R, Jonas M, Coskun AU, Stone BV, Maynard C, Shi GP, Libby P, Feldman CL, 61. HPS/TIMI55-REVEAL Collaborative Group. Effects of Anacetrapib in Pa-
Edelman ER, Stone PH. Augmented expression and activity of extracellular tients with Atherosclerotic Vascular Disease [published online ahead of
matrix-degrading enzymes in regions of low endothelial shear stress colo- print August 28, 2017]. N Engl J Med. doi: 10.1056/NEJMoa1706444.
calize with coronary atheromata with thin fibrous caps in pigs. Circulation. 62. Meuwese MC, de Groot E, Duivenvoorden R, Trip MD, Ose L, Maritz FJ, Ba-
2011;123:621630. doi: 10.1161/CIRCULATIONAHA.110.970038. sart DC, Kastelein JJ, Habib R, Davidson MH, Zwinderman AH, Schwocho
44. Shiomi M, Ishida T, Kobayashi T, Nitta N, Sonoda A, Yamada S, Koike LR, Stein EA; CAPTIVATE Investigators. ACAT inhibition and progression of
T, Kuniyoshi N, Murata K, Hirata K, Ito T, Libby P. Vasospasm of athero- carotid atherosclerosis in patients with familial hypercholesterolemia: the
sclerotic coronary arteries precipitates acute ischemic myocardial damage CAPTIVATE randomized trial. JAMA. 2009;301:11311139. doi: 10.1001/
in myocardial infarction-prone strain of the Watanabe heritable hyperlip- jama.301.11.1131.

Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1165


Crea and Libby

63. Lscher TF. Substrates of acute coronary syndromes: new insights into 81. Bertrand ME, LaBlanche JM, Tilmant PY, Thieuleux FA, Delforge MR, Carre
plaque rupture and erosion. Eur Heart J. 2015;36:13471349. doi: AG, Asseman P, Berzin B, Libersa C, Laurent JM. Frequency of provoked
10.1093/eurheartj/ehv149. coronary arterial spasm in 1089 consecutive patients undergoing coronary
64. Ferrante G, Nakano M, Prati F, Niccoli G, Mallus MT, Ramazzotti V, Mon- arteriography. Circulation. 1982;65:12991306.
tone RA, Kolodgie FD, Virmani R, Crea F. High levels of systemic my- 82. Pristipino C, Beltrame JF, Finocchiaro ML, Hattori R, Fujita M, Mongiardo
eloperoxidase are associated with coronary plaque erosion in patients R, Cianflone D, Sanna T, Sasayama S, Maseri A. Major racial differences
with acute coronary syndromes: a clinicopathological study. Circulation. in coronary constrictor response between Japanese and Caucasians with
2010;122:25052513. doi: 10.1161/CIRCULATIONAHA.110.955302. recent myocardial infarction. Circulation. 2000;101:11021108.
65. Yahagi K, Kolodgie FD, Lutter C, Mori H, Romero ME, Finn AV, Virmani 83. Camici PG, Crea F. Coronary microvascular dysfunction. N Engl J Med.
R. Pathology of human coronary and carotid artery atherosclerosis and 2007;356:830840. doi: 10.1056/NEJMra061889.
vascular calcification in diabetes mellitus. Arterioscler Thromb Vasc Biol. 84. Galiuto L, De Caterina AR, Porfidia A, Paraggio L, Barchetta S, Locorotondo
2017;37:191204. doi: 10.1161/ATVBAHA.116.306256. G, Rebuzzi AG, Crea F. Reversible coronary microvascular dysfunction: a
66. Hansson GK, Libby P, Tabas I. Inflammation and plaque vulnerability. J In- common pathogenetic mechanism in apical ballooning or tako-tsubo syn-
tern Med. 2015;278:483493. doi: 10.1111/joim.12406. drome. Eur Heart J. 2010;31:13191327. doi: 10.1093/eurheartj/ehq039.
67. Quillard T, Arajo HA, Franck G, Shvartz E, Sukhova G, Libby P. TLR2 and 85. Templin C, Ghadri JR, Diekmann J, Napp LC, Bataiosu DR, Jaguszewski M,
neutrophils potentiate endothelial stress, apoptosis and detachment: im- Cammann VL, Sarcon A, Geyer V, Neumann CA, Seifert B, Hellermann J,
plications for superficial erosion. Eur Heart J. 2015;36:13941404. doi: Schwyzer M, Eisenhardt K, Jenewein J, Franke J, Katus HA, Burgdorf C,
10.1093/eurheartj/ehv044. Schunkert H, Moeller C, Thiele H, Bauersachs J, Tschpe C, Schultheiss HP,
68. Franck G, Mawson T, Sausen G, Salinas M, Masson GS, Cole A, Beltrami- Laney CA, Rajan L, Michels G, Pfister R, Ukena C, Bhm M, Erbel R, Cuneo
Moreira M, Chatzizisis Y, Quillard T, Tesmenitsky Y, Shvartz E, Sukhova GK, A, Kuck KH, Jacobshagen C, Hasenfuss G, Karakas M, Koenig W, Rott-
Swirski FK, Nahrendorf M, Aikawa E, Croce KJ, Libby P. Flow perturbation bauer W, Said SM, Braun-Dullaeus RC, Cuculi F, Banning A, Fischer TA, Va-
mediates neutrophil recruitment and potentiates endothelial injury via sankari T, Airaksinen KE, Fijalkowski M, Rynkiewicz A, Pawlak M, Opolski
TLR2 in mice: implications for superficial erosion. Circ Res. 2017;121:31 G, Dworakowski R, MacCarthy P, Kaiser C, Osswald S, Galiuto L, Crea F,
Dichtl W, Franz WM, Empen K, Felix SB, Delmas C, Lairez O, Erne P, Bax JJ,
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

42. doi: 10.1161/CIRCRESAHA.117.310694.


69. Kolodgie FD, Burke AP, Wight TN, Virmani R. The accumulation of specific Ford I, Ruschitzka F, Prasad A, Lscher TF. Clinical features and outcomes
types of proteoglycans in eroded plaques: a role in coronary thrombosis in of takotsubo (stress) cardiomyopathy. N Engl J Med. 2015;373:929938.
the absence of rupture. Curr Opin Lipidol. 2004;15:575582. doi: 10.1056/NEJMoa1406761.
70. Day AJ. The structure and regulation of hyaluronan-binding proteins. Bio- 86. Arrebola-Moreno AL, Arrebola JP, Moral-Ruiz A, Ramirez-Hernandez JA,
Melgares-Moreno R, Kaski JC. Coronary microvascular spasm triggers
chem Soc Trans. 1999;27:115121.
transient ischemic left ventricular diastolic abnormalities in patients with
71. Butler LM, Rainger GE, Nash GB. A role for the endothelial glycosami-
chest pain and angiographically normal coronary arteries. Atherosclerosis.
noglycan hyaluronan in neutrophil recruitment by endothelial cells cul-
2014;236:207214. doi: 10.1016/j.atherosclerosis.2014.07.009.
tured for prolonged periods. Exp Cell Res. 2009;315:34333441. doi:
87. Kaski JC, Maseri A, Vejar M, Crea F, Hackett D. Spontaneous coronary
10.1016/j.yexcr.2009.08.012.
artery spasm in variant angina is caused by a local hyperreactivity to a
72. Dring Y, Soehnlein O, Weber C. Neutrophil extracellular traps in ath-
generalized constrictor stimulus. J Am Coll Cardiol. 1989;14:14561463.
erosclerosis and atherothrombosis. Circ Res. 2017;120:736743. doi:
88. Lanza GA, Pedrotti P, Pasceri V, Lucente M, Crea F, Maseri A. Autonomic
10.1161/CIRCRESAHA.116.309692.
changes associated with spontaneous coronary spasm in patients with
73. Libby P. Triglycerides on the rise: should we swap seats on the seesaw? Eur
variant angina. J Am Coll Cardiol. 1996;28:12491256. doi: 10.1016/
Heart J. 2015;36:774776. doi: 10.1093/eurheartj/ehu500.
S0735-1097(96)00309-9.
74. Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzenbck A,
89. Shimokawa H, Tomoike H, Nabeyama S, Yamamoto H, Araki H, Nakamura
Simon D, Laimer D, Bangert C, Kammerlander A, Mascherbauer J, Winter
M, Ishii Y, Tanaka K. Coronary artery spasm induced in atherosclerotic
MP, Distelmaier K, Adlbrecht C, Preissner KT, Lang IM. Coronary neutro-
miniature swine. Science. 1983;221:560562.
phil extracellular trap burden and deoxyribonuclease activity in ST-eleva-
90. Ito A, Shimokawa H, Nakaike R, Fukai T, Sakata M, Takayanagi T, Egashira K,
tion acute coronary syndrome are predictors of ST-segment resolution
Takeshita A. Role of protein kinase C-mediated pathway in the pathogenesis
and infarct size. Circ Res. 2015;116:11821192. doi: 10.1161/CIRCRE-
of coronary artery spasm in a swine model. Circulation. 1994;90:24252431.
SAHA.116.304944.
91. Shimokawa H, Seto M, Katsumata N, Amano M, Kozai T, Yamawaki T, Ku-
75. Aguirre-Alvarado C, Segura-Cabrera A, Velzquez-Quesada I, Hernndez- wata K, Kandabashi T, Egashira K, Ikegaki I, Asano T, Kaibuchi K, Takeshita
Esquivel MA, Garca-Prez CA, Guerrero-Rodrguez SL, Ruiz-Moreno AJ, A. Rho-kinase-mediated pathway induces enhanced myosin light chain
Rodrguez-Moreno A, Prez-Tapia SM, Velasco-Velzquez MA. Virtual phosphorylations in a swine model of coronary artery spasm. Cardiovasc
screening-driven repositioning of etoposide as CD44 antagonist in Res. 1999;43:10291039.
breast cancer cells. Oncotarget. 2016;7:2377223784. doi: 10.18632/ 92. Shimokawa H. 2014 Williams Harvey Lecture: importance of coro-
oncotarget.8180. nary vasomotion abnormalities: from bench to bedside. Eur Heart J.
76. Jia H, Dai J, Hou J, Xing L, Ma L, Liu H, Xu M, Yao Y, Hu S, Yamamoto E, 2014;35:31803193. doi: 10.1093/eurheartj/ehu427.
Lee H, Zhang S, Yu B, Jang IK. Effective anti-thrombotic therapy without 93. Dai Z, Aoki T, Fukumoto Y, Shimokawa H. Coronary perivascular fibrosis
stenting: intravascular optical coherence tomography-based management is associated with impairment of coronary blood flow in patients with
in plaque erosion (the EROSION study). Eur Heart J. 2017;38:792800. non-ischemic heart failure. J Cardiol. 2012;60:416421. doi: 10.1016/j.
doi: 10.1093/eurheartj/ehw381. jjcc.2012.06.009.
77. Libby P. Superficial erosion and the precision management of acute coro- 94. Kaski JC, Crea F, Meran D, Rodriguez L, Araujo L, Chierchia S, Davies G,
nary syndromes: not one-size-fits-all. Eur Heart J. 2017;38:801803. doi: Maseri A. Local coronary supersensitivity to diverse vasoconstrictive stimuli
10.1093/eurheartj/ehw599. in patients with variant angina. Circulation. 1986;74:12551265.
78. Beltrame JF, Crea F, Kaski JC, Ogawa H, Ong P, Sechtem U, Shimokawa 95. Lanza GA, Careri G, Crea F. Mechanisms of coronary artery spasm. Circulation.
H, Bairey Merz CN; Coronary Vasomotion Disorders International Study 2011;124:17741782. doi: 10.1161/CIRCULATIONAHA.111.037283.
Group (COVADIS). The who, what, why, when, how and where of vaso- 96. Abbate A, Hamza M, Cassano AD, Melchior R, Roberts C, Grizzard J, Shah
spastic angina. Circ J. 2016;80:289298. doi: 10.1253/circj.CJ-15-1202. K, Hastillo A, Kasirajan V, Crea F, Lanza GA, Vetrovec GW. Sympathec-
79. Ong P, Athanasiadis A, Borgulya G, Voehringer M, Sechtem U. 3-Year fol- tomy as a treatment for refractory coronary artery spasm. Int J Cardiol.
low-up of patients with coronary artery spasm as cause of acute coronary 2012;161:e7e9. doi: 10.1016/j.ijcard.2012.03.006.
syndrome: the CASPAR (Coronary Artery Spasm in Patients With Acute 97. Masumoto A, Mohri M, Shimokawa H, Urakami L, Usui M, Takeshita A.
Coronary Syndrome) study follow-up. J Am Coll Cardiol. 2011;57:147 Suppression of coronary artery spasm by the Rho-kinase inhibitor fasudil
152. doi: 10.1016/j.jacc.2010.08.626. in patients with vasospastic angina. Circulation. 2002;105:15451547.
80. Sato K, Kaikita K, Nakayama N, Horio E, Yoshimura H, Ono T, Ohba K, 98. King K, Grazette LP, Paltoo DN, McDevitt JT, Sia SK, Barrett PM, Apple
Tsujita K, Kojima S, Tayama S, Hokimoto S, Matsui K, Sugiyama S, Yamabe FS, Gurbel PA, Weissleder R, Leeds H, Iturriaga EJ, Rao A, Adhikari B,
H, Ogawa H. Coronary vasomotor response to intracoronary acetylcholine Desvigne-Nickens P, Galis ZS, Libby P. Point-of-care technologies for preci-
injection, clinical features, and long-term prognosis in 873 consecutive pa- sion cardiovascular care and clinical research: National Heart, Lung, and
tients with coronary spasm: analysis of a single-center study over 20 years. Blood Institute Working Group. JACC Basic Transl Sci. 2016;1:7386. doi:
J Am Heart Assoc. 2013;2:e000227. doi: 10.1161/JAHA.113.000227. 10.1016/j.jacbts.2016.01.008.

1166 September 19, 2017 Circulation. 2017;136:11551166. DOI: 10.1161/CIRCULATIONAHA.117.029870


Acute Coronary Syndromes: The Way Forward From Mechanisms to Precision
Treatment
Filippo Crea and Peter Libby

Circulation. 2017;136:1155-1166
Downloaded from http://circ.ahajournals.org/ by guest on September 18, 2017

doi: 10.1161/CIRCULATIONAHA.117.029870
Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright 2017 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7322. Online ISSN: 1524-4539

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circ.ahajournals.org/content/136/12/1155

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial
Office. Once the online version of the published article for which permission is being requested is located,
click Request Permissions in the middle column of the Web page under Services. Further information about
this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation is online at:


http://circ.ahajournals.org//subscriptions/

Vous aimerez peut-être aussi