SlideShare a Scribd company logo
STATEOFTHEART
Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1155
Filippo Crea, MD
Peter Libby, MD
ABSTRACT: Well into the 21st century, we still triage acute myocardial
infarction on the basis of the presence or absence of ST-segment
elevation, a century-old technology. Meanwhile, we have learned a great
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.
Acute Coronary Syndromes
The Way Forward From Mechanisms to Precision Treatment
© 2017 American Heart
Association, Inc.
Correspondence to: Peter Libby,
MD, Brigham and Women’s
Hospital, Harvard Medical School,
77 Ave Louis Pasteur, NRB-741-G,
Boston, MA 02115. E-mail plibby@
bwh.harvard.edu
IN DEPTH
Key Words: erosion
◼ inflammation ◼ microvessels
◼ myocardial infarction ◼ plaque
◼ plaque, atherosclerotic
◼ thrombosis
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
Crea and Libby
September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701156
W
illem Einthoven introduced electrocardiogra-
phy at the dawn of the 20th century. Well into
the 21st century, we still triage acute myocar-
dial infarction on the basis of this venerable technology:
ST segments up or not. Although this approach remains
the appropriate evidence-based strategy today, aiming
toward the future goal of more individualized therapy,
can we apply a more pathophysiologically based cat-
egorization of the acute coronary syndromes (ACS)? In-
deed, we have learned much about the mechanisms of
ACS since we last reviewed this topic in these pages.1,2
Moreover, the human disease of atherosclerosis has
evolved considerably in the interim.3–5
This review aims
to work toward a categorization of the ACS that reach-
es beyond equating all ACS with plaque rupture or fis-
sure, guided by the latest insights into the underlying
mechanisms of ACS, and points a way forward toward
the eventual clinical application of these considerations.
Postmortem studies carried out in the 1980s pro-
posed that plaque rupture (also sometimes referred to
as fissure) caused most fatal myocardial infarctions.6
These findings led to the notion of the vulnerable or
high-risk plaque characterized by a large central lipid
core, an abundance of inflammatory cells, a paucity of
smooth muscle cells (SMCs), and a thin fibrous cap.7
These observations spawned the widely accepted con-
cept that instability of coronary atheromata resulted
from fissuring of a thin-capped fibroatheroma caused
by a weakening of its collagen structure wrought by
inflammatory mechanisms.8
This concept stimulated manifold attempts to de-
velop methods to detect the vulnerable plaque, a quest
predicated on the postulate that local interventions
could preclude plaque thrombosis and possibly prevent
ACS. However, attempts to identify vulnerable plaques
proved disappointing because of their low predictive
value.9
Pathological studies of human coronary arter-
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
as determined by intravascular ultrasound interroga-
tion actually cause clinical events during over 3 years
of follow-up.11
Thus, most thin-capped fibroatheroma
are clinically quite stable, a realization that renders
vulnerable plaque a misnomer. Indeed, plaques with
thin-capped fibroatheroma morphology as assessed
by radiofrequency backscatter data from intravascular
ultrasound (virtual histology) commonly convert to an
apparently more stable character during a 1-year fol-
low-up period.12
Such evolution in plaques may reflect
mutability during the natural history of human athero-
sclerosis but could also reflect adherence of many of
these patients to a contemporary secondary preven-
tion regimen, including smoking cessation, statins, and
agents that target the renin-angiotensin axis.
Moreover, although we have witnessed wide accep-
tance of the formerly controversial concept that inflam-
mation contributes to atherogenesis,13
inflammation
may not drive all transitions from stable atherosclerosis
to acute thrombotic events. In a large study, about half
of ACS occurred in the presence of normal levels of
C-reactive protein (CRP), a marker of inflammation.14
Another recent study using optical coherence tomog-
raphy (OCT) demonstrated that among patients with
ACS and plaque rupture, two thirds of patients had im-
aging evidence of inflammatory cell infiltration in the
region of the ruptured fibrous cap but one third did
not and the latter had lower levels of CRP.15
In addition,
plaque erosion causes up to a third of ACS in the cur-
rent era.16,17
Finally, about one fifth of ACS occur in the
apparent absence of coronary thrombosis, suggesting
that functional alterations beyond thrombus formation
can contribute to ACS pathogenesis.18,19
The ensemble of postmortem studies and in vivo
studies using intravascular imaging point to 4 patho-
logical pathways to ACS. Although these mechanisms
may overlap and coexist in some patients, we present
them separately for heuristic simplicity: plaque rupture
with systemic inflammation, plaque rupture without
systemic inflammation, plaque erosion, and plaque
without thrombus (Figure 1).20
This review considers the causes of these 4 predomi-
nant pathways to ACS. To strive for a more precision
approach, we explore the therapeutic implications of
these distinct mechanisms that provoke ACS. Moreover,
we advocate the future goal to move beyond triage of
therapy based merely on electrocardiographic current
of injury by harnessing the recent advances in arterial
biology and human studies that shed new mechanis-
tic light on ACS pathogenesis. Further improvements
in the appropriate deployment of technologies and in
outcomes should evolve from a more mechanistically
based classification of ACS.
PLAQUE RUPTURE WITH SYSTEMIC
INFLAMMATION
Mechanisms
Several studies have implicated systemic inflammation
in ACS as assessed by the biomarker CRP (Figure 1A).21
Laboratory studies and observations on human plaques
point to inflammatory mechanisms as key regulators of
the fragility of the fibrous cap and of the thrombogenic
potential of the lipid core (Figure 2). Macrophages likely
pave the way for the rupture of the fibrous cap of the
plaque: When activated, these cells elaborate enzymes
that degrade all components of the arterial extracellular
matrix. These enzymes include matrix metalloproteinases
and certain cathepsins. Multiple mechanisms regulate
these matrix-degrading proteinases: transcription, trans-
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
ACS Mechanisms Inform Management
STATEOFTHEART
Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1157
lation, activation of zymogen precursors, and balance
with endogenous inhibitors such as the tissue inhibitors
of matrix metalloproteinases or the cystatins. Thus, in-
creased amounts of activated proteinases or reduced lev-
els of their corresponding inhibitors can enhance break-
down of the extracellular matrix of plaque.22
Adaptive immunity also appears altered in coronary
plaque instability.23
Patients with ACS have an increased
population of particularly proinflammatory CD4+
T cells
characterized by low cell surface expression of CD28, a
costimulatory molecule critically involved in determin-
ing the outcome of antigen recognition by T cells.24
ACS
also profoundly perturb the numbers of 2 other circulat-
ing T-cell subsets: type 17 helper T cells and CD4+
CD25+
regulatory T cells (Tregs
). The net role of interleukin (IL)-17
in atherosclerosis remains controversial, however; some
experimental studies in mice support a predominantly
proatherogenic function for IL-17, yet type 17 helper
T cells activated in plaques promote the formation of
thick collagen fibers, which might increase plaque sta-
bility.25,26
Tregs
normally help to maintain homeostasis of
cells involved in adaptive immunity, including antigen-
presenting cells and effector T cells. Tregs
mediate these
modulatory effects by contact-dependent suppression
or by releasing anti-inflammatory cytokines such IL-
10 or transforming growth factor-β1.27
Patients with
ACS have a reduced number and suppressive function
of circulating Tregs
compared with patients with stable
angina and healthy control subjects.28
The molecular
mechanisms responsible for the T-cell dysregulation in
ACS remain largely unknown. Recent work highlights
the importance of the strength of the stimulation of
the T-cell receptor for antigen in the differentiation of
helper T-cell subsets. The altered activity observed in
ACS of proteins involved in the regulation of upstream
T-cell receptor for antigen signaling such as CD31 and
protein tyrosine phosphatase N22 may modulate T-cell
functions.29,30
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 non–ST-segment–elevation 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.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
Crea and Libby
September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701158
Therapeutic Implications
Several anti-inflammatory treatments might provide
benefit in this setting. Colchicine, a time-honored
anti-inflammatory drug, reduced major cardiovascular
events in a medium-sized, unblinded randomized study
in patients with stable ischemic heart disease.31
Two tri-
als underway are evaluating colchicine treatment in pa-
tients with coronary artery disease, although not in the
acute phase.32
A phase III trial is testing the potential
beneficial effects of low-dose methotrexate in patients
after myocardial infarction or with coronary artery dis-
ease and diabetes mellitus or elements of the metabolic
syndrome.33
Targeting proinflammatory cytokines offers an-
other approach. Because of the potentially detrimen-
tal effects on cardiovascular risk profile or the lack of
compelling evidence of benefit in experimental ath-
erosclerosis, monoclonal antibodies that neutralize tu-
mor necrosis factor-α, interferon-γ, or IL-17 may not
prove appropriate for treating atherosclerosis. In a small
(n=182) phase II study, the IL-1 receptor antagonist
anakinra reduced the area under the curve of high-
sensitivity CRP in patients in the 7 days after presenta-
tion with ACS.34
A large (n10 000) phase III trial with
clinical end points that tested a fully humanized anti–
IL-1β monoclonal antibody, canakinumab, in patients
with previous myocardial infarction and CRP levels 2
mg/L showed a significant 15% reduction in the prima-
ry endpoint of MI, stroke, or death, and a 17% fall in
a secondary endpoint that included urgent revascular-
ization. Indeed, revascularization procedures declined
30% in the group that received the interleukin-1β neu-
tralizing antibody canakinumab 150mg subcutaneously
every 3 months. This intervention trial substantiates the
causal contribution of inflammation to atherosclerotic
events.35-37
T-cell activation signaling offers possible targets. A
synthetic CD31-derived peptide can suppresses im-
munity in vivo by restoring an inhibitory pathway to a
CD31-negative T-cell subset.38
Moreover, the develop-
ment of a novel series of inhibitors of the phosphatase
protein tyrosine phosphatase N22 might contribute to
a better understanding of its role in immune dysregula-
tion in patients with ACS.39
Therapeutic strategies that
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-γ, interferon-γ; TGFβ, transforming growth factor-β; and TNF, tumor
necrosis factor.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
ACS Mechanisms Inform Management
STATEOFTHEART
Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1159
target Treg
include administration of low doses of IL-2
and infusion of autologous Tregs
that have undergone
differentiation and expansion in vitro.40
Various vaccina-
tion strategies also can limit experimental atherogen-
esis and may act by augmenting the activity of Treg
.41
PLAQUE RUPTURE WITHOUT
SYSTEMIC INFLAMMATION
Mechanisms
When plaque rupture occurs in the absence of systemic
inflammatory activation, other mechanisms may con-
tribute to pathogenesis, including extreme emotional
disturbance ranging from external events of short dura-
tion such as earthquakes and a beloved team’s loss of a
football (soccer) match to acute manifestations of more
long-term emotional disturbances (Figure 1B). Intense
physical exertion and local mechanical stress at the level
of the artery wall, either increased circumferential stress
or reduced shear stress, may also predispose to plaque
rupture.42
In addition, subclinical inflammation in the
microenvironment of the culprit stenosis might com-
pound the chain of events leading to coronary instabil-
ity, although the triggers for and effectors of such local
inflammation may differ from those that operate in
patients with systemic inflammation.43
Although many
studies have clarified the molecular mechanisms lead-
ing to coronary instability in individuals with systemic
inflammation (eg, gauged by CRP elevations), patients
without systemic inflammation have undergone less
extensive investigation. The precise causes of instability
remain poorly understood, providing a strong stimulus
for further research.
The possible relationship between psychological
stress and plaque rupture may relate to sympathetic
nervous system activation and catecholamine release
associated with an increase in heart rate, blood pres-
sure, and coronary vasoconstriction favoring plaque dis-
ruption and platelet activation, hypercoagulability, and
intense coronary microvascular constriction.44
More-
over, β3
-adrenergic stimulation can stimulate release
from the bone marrow of proinflammatory monocytes
that can home to experimental atheromata and amplify
local inflammation.45
Although physical or emotional
stress per se may not suffice to cause coronary throm-
bosis, it might trigger instability in plaques already pre-
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)-1β and IL-18. Large crystals might also cause mechanical disruption of the fibrous cap.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
Crea and Libby
September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701160
Local changes in the equilibrium between esterified
and free cholesterol might promote plaque rupture47
(Fig-
ure 3). Cholesterol crystal formation in the lipid core could
heighten the risk of plaque rupture and thrombosis and
can coactivate the inflammasome, an intracellular multi-
meric complex that generates active IL-1β and IL-18.48
OCT has colocalized cholesterol crystals in human
coronary arteries with thin-capped plaques.49
The devel-
opment of micro-OCT with a 2-μm resolution may shed
new light on this potential mechanism of instability by
assessing its occurrence in vivo.50
Cholesterol crystals
might also theoretically resist plaque rupture by increas-
ing the stiffness of the lipid pool, although this mecha-
nism probably contributes little to plaque stability.51
The
role of calcium mineral as a causal factor in ACS remains
controversial. Large collections of calcified tissue do not
augment the biomechanical instability of plaques or as-
sociate with lesions that provoke ACS.52,53
Small foci of
calcification within the atherosclerotic intima, some caus-
ing spotty calcification visible on computed tomography,
can also introduce biomechanical inhomogeneities that
can favor plaque destabilization.3,54,55
Therapeutic Implications
Because it is difficult to limit environmental, physical,
or emotional triggers, the altered plaque characteristics
produced by intensive lipid-lowering treatment might
protect against this form of plaque destabilization.56
In
particular, statins and ezetimibe may interfere with cho-
lesterol crystal formation.57,58
Cyclodextrin can solubilize
cholesterol, limits experimental atherogenesis, and thus
might provide a pharmacological approach to combat-
ting cholesterol crystal accumulation in plaques.59
Enhancement of cholesterol efflux might promote
plaque stabilization, but strategies to enhance this process
by manipulation of high-density lipoprotein concentra-
tions or by administration of apolipoprotein A-1 have gen-
erally failed to confer clinical benefit.60
A large outcome
trial with anacetrapib produced a modest event reduction,
but since this agent not only raises high-density lipopro-
tein but also lowers low-density lipoprotein renders it dif-
ficult to attribute event reduction to raising high-density
lipoprotein.61
In addition, the inhibition of cholesteryl ester
hydrolase, an enzyme that converts cholesteryl esters to
free cholesterol, might prevent cholesterol crystallization.
Inhibition of acetyl-CoA acetyltransferase-1, which in con-
trast promotes cholesterol crystallization, increased ath-
eroma volume and major cardiovascular events.62
PLAQUE EROSION
Mechanisms
Current evidence suggests that mechanisms of plaque
disruption distinct from macrophage-driven rupture of
the fibrous cap of plaque can commonly cause ACS.63
The mechanism of plaque thrombosis described by pa-
thologists as superficial erosion appears not to involve
inflammation mediated by macrophages, as in the case
of fibrous cap fracture (Figure 1C). Superficial erosion
complicates lesions with a distinct epidemiology and
morphology and involves pathophysiological mecha-
nisms that differ from rupture of the fibrous cap. In-
deed, neutrophil activation seems to play a pivotal role
in thrombosis due to plaque erosion.4
Patients who
presented with ACS associated with plaque erosion
defined by OCT had higher systemic myeloperoxidase
levels compared with patients with plaque rupture.
Moreover, in postmortem coronary artery specimens,
luminal thrombi superimposed on eroded plaques con-
tain many more myeloperoxidase-positive cells than
thrombi associated with ruptured plaques64
(Figure 4).
Autopsy studies suggest that fatal plaque erosion as-
sociates more commonly with hypertriglyceridemia, dia-
betes mellitus, female sex, and old age.65
The morphology
of lesions that produce thrombosis caused by superficial
erosion differs radically from the features associated with
fibrous cap rupture. Superficially eroded lesions contain
few macrophages or T lymphocytes, inflammatory cells
considered pathogenic in plaque rupture. Rather than
depleted collagen, the lesions associated with superficial
erosion contain abundant proteoglycan and glycosami-
noglycans. Instead of a paucity of SMCs, eroded lesions
can contain abundant arterial SMCs. Plaques complicat-
ed by fibrous cap fracture typically harbor large necrotic,
lipid-rich cores. In contrast, the more fibrous lesions asso-
ciated with superficial erosion generally lack prominent
central lipid cores.66
A loss of intimal endothelial cells de-
fines lesions that provoke thrombosis by the mechanism
denoted as superficial erosion.
Recent work has implicated engagement of the innate
immune receptor Toll-like receptor 2 in promoting the
susceptibility of endothelial cells to apoptotic stimuli. Hy-
aluronic acid, a prominent constituent of the extracellular
matrix of eroded lesions, could serve as an endogenous
ligand of Toll-like receptor 2, implicated in promoting
endothelial cell apoptosis.67
Experiments that have evalu-
ated gain and loss of Toll-like receptor 2 function in mice
support these observations.68
Indeed, hyaluronan and its
receptor, CD44, colocalize along the plaque/thrombus
interface in eroded plaque but not in ruptured or stable
plaque.69
Accumulation of hyaluronan and expression
of CD44 along the plaque/thrombus interface of erod-
ed plaques may promote de-endothelization, resulting
in CD44-dependent platelet adhesion and subsequent
thrombus formation, mediated in part by a direct action
of hyaluronan on fibrin polymerization.70,71
We have hypothesized that thrombosis caused by su-
perficial erosion occurs in 2 phases. The first hit, mediat-
ed, for example, by Toll-like receptor 2, would jeopardize
endothelial viability or adherence, leading to a breach in
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
ACS Mechanisms Inform Management
STATEOFTHEART
Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1161
the integrity of the endothelial monolayer overlying the
atherosclerotic plaque. The denuded patch on the intimal
surface would then attract platelets that could undergo
activation by contact with collagen and other compo-
nents of the arterial extracellular matrix usually protected
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:
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 lipoprotein–lowering therapies.73
Moreover, therapies that destabilize neutrophil extra-
cellular traps such as deoxyribonuclease administration
might limit the propagation of thrombi resulting from
superficial erosion.74
Receptors such as CD44 expressed
by neutrophils may bind hyaluronan and glycosamino-
glycan enriched in plaques complicated by erosion and
furnish a potential therapeutic target.75
Jia et al76
recently reported a series of patients with
ACS with OCT-defined erosion and treated with anti-
coagulant/antiplatelet therapy rather that mechanical
revascularization. About 80% of the patients showed
a 50% reduction of thrombus volume after 1 month.
More than a third of patients lacked detectable thrombus
after 1 month. This study suggests that pharmacological
rather than interventional treatment can effectively man-
age ACS caused by superficial erosion, a proposition that
merits and requires rigorous testing in outcome trials.77
PLAQUE WITHOUT THROMBUS
Mechanisms
In patients with ACS without plaque thrombus, a func-
tional alteration of coronary circulation likely causes
Figure 4. Pathways implicated in arterial thrombosis
caused by superficial erosion.
Stimuli such as disturbed flow or engagement of innate im-
mune receptors such as Toll-like receptor-2 (TLR2) can activate
the endothelial cells that line the arterial intima. These cells can
undergo cell death, eg, by apoptosis, depicted by the cell with
the interrupted membrane and pyknotic nucleus. Injured or dy-
ing endothelial cells can desquamate, uncovering the basement
membrane. Neutrophils attracted by chemokines produced
by activated endothelial cells can congregate on the denuded
intima and can in turn degranulate and die, releasing neutro-
phil extracellular traps (NETs). These strands of extruded DNA
can bind the contents of the neutrophil granules and other
proteins, eg, myeloperoxidase or tissue factor. The NETs can
constitute a solid-state reactor that generates oxidants such as
hypochlorous acid and stimulates coagulation locally. Platelets
interacting with the basement membrane can activate, release
their granular contents, including chemokines that can recruit
further leukocytes, and form the nidus of a white thrombus.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
Crea and Libby
September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701162
acute ischemia involving large epicardial coronary
arteries or the coronary microcirculation (Figure 1D).
Epicardial coronary vasospasm may occur in patients
in whom coronary angiography does not demonstrate
an obstructive atherosclerotic plaque.78
In the CASPAR
study (Coronary Artery Spasm in Patients With Acute
Coronary Syndrome), coronary angiography failed to
show culprit lesions in ≈30% of patients with sus-
pected ACS.79
Intracoronary acetylcholine administra-
tion elicited coronary spasm in nearly 50% of these
patients. Similar findings pertained to a Japanese
population.80
Coronary artery spasm also may cause
coronary instability in patients with obstructive ath-
erosclerosis. Indeed, a classic study by Bertrand et al81
found that ergonovine induced spasm in 20% of pa-
tients with recent myocardial infarction and in 40% of
patients with unstable angina. In a more recent study,
acetylcholine induced spasm in 20% of white patients
and in 60% of patients with myocardial infarction
within the previous 14 days.82
Japanese people have a
higher prevalence of coronary spasm than whites for
unknown reasons.
Microvascular spasm also can cause myocardial isch-
emia.83
This mechanism likely operates in patients with
Takotsubo cardiomyopathy,84
which frequently occurs
in the absence of obstructive atherosclerosis, although
≈15% of these patients exhibit concomitant obstructive
atherosclerosis.85
Microvascular spasm can also contrib-
ute to ischemia in the face of angiographically normal-
appearing epicardial coronary arteries. Microvascular
ischemia often manifests as angina pectoris but can
also cause non–ST-segment–elevation ACS as defined
by electrocardiographic alterations and biomarker evi-
dence of myocyte injury.86
Coronary spasm can also
contribute to plaque instability by causing endothelial
damage, as shown in an infarction-prone strain of the
Watanabe heritable hyperlipidemic rabbit.44
Either macrovascular or microvascular spasm can
result from impaired vasodilatation or from vasocon-
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.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
ACS Mechanisms Inform Management
STATEOFTHEART
Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1163
(Figure  5).87,88
Shimokawa et al89
demonstrated SMC
hyperreactivity in a coronary segment in pigs after
adventitial exposure of a coronary segment to an in-
flammatory stimulus (IL-1β). Subsequent experimental
and clinical data suggested that an increase in Rho-
kinase activity was a major mechanism of SMC hy-
perreactivity.90–92
Perivascular fibrosis in the coronary
microvasculature can contribute to myocardial isch-
emia.93
This observation highlights the potential contri-
bution of fixed structural changes in coronary arterioles
beyond impaired endothelial vasodilator function as a
contributor to myocardial ischemia in the absence of
obstructive disease of the epicardial coronary arteries.
Therapeutic Implications
Several vasoconstrictor substances implicated as trig-
gers of epicardial coronary spasm may cause ischemia in
the same patient under different conditions.94
Accord-
ingly, the vasoactive substances themselves may not re-
spond to therapeutic targeting. The current treatment
of epicardial spasm uses nonspecific vasodilators such
as long-acting nitrates and calcium channel blockers.95
Nitrates and calcium channel antagonists may alleviate
ischemia caused by vasospasm. However, we need to
identify the molecular alterations responsible for SMC
hyperreactivity because many patients do not respond
satisfactorily to standard doses of vasodilators.96
Knowl-
edge of the postreceptor mechanisms responsible for
SMC hyperreactivity and epicardial coronary spasm
might enable the development of new therapeutic op-
tions for patients who present poor clinical response to
nonspecific vasodilator therapy. Fasudil, a Rho kinase
inhibitor, reduces the rate of coronary spasm episodes
in patients with vasospastic angina.97
Similarly, further
efforts are warranted to unravel the molecular mecha-
nisms responsible for coronary microvascular dysfunc-
tion in Takotsubo cardiomyopathy and in ischemia with
normal coronary arteries.20
The observation that Tako­
tsubo cardiomyopathy may have worse outcomes than
previously considered and the challenges of managing
patients with episodic ischemia caused by microvascu-
lar dysfunction render this quest urgent.85
Preventing
or reversing perivascular fibrosis in the coronary arterial
microvasculature represents another novel target be-
yond traditional antiatherosclerotic therapies.
CONCLUSIONS AND PERSPECTIVES
A dichotomous approach based on the ECG has tradi-
tionally dominated our clinical management of ACS:
ST-segment–myocardial infarction versus non–ST-
segment–myocardial infarction. Our thinking concern-
ing the pathophysiological bases of the ACS has also
evolved separately and, in parallel, has been dominated
by the concept of the vulnerable plaque as a substrate
for fibrous cap rupture or of a minority mechanism:
superficial erosion. We and others have championed
and explored the role of inflammation as a pathway
to the ACS.
However, we now have a greater appreciation that
not all cases of ACS fit this mold. To move forward, we
need to remain vigilant not to cling to our cherished no-
tions and to remain open to shifts in the nature of the
diseases we treat and to new vistas opened by scientific
advances. As discussed here, we need to consider more
deeply in the current era arterial spasm, rupture with-
out inflammation, and distinct aspects of innate im-
munity or dyslipidemia as instigators of plaque erosion.
The complexity of clinical ACS surpasses our traditional
categorization of this condition. We should evolve be-
yond the current constrained concepts by expanding
our understanding.
We have achieved considerable mastery of plaque
rupture with inflammation from a mechanistic and
therapeutic viewpoint. Control of traditional risk fac-
tors such as hypercholesterolemia, hypertension, and
smoking addresses the pathophysiology of this mecha-
nism of ACS. The other categories that we discuss here
may account for an increasing proportion of ACS, now
that we are addressing more effectively the traditional
risk factors.5
We should remain open to the possibility,
even likelihood, that tailored therapeutic approaches
may apply to the prevention and treatment of the less
well-recognized pathways to acute myocardial ischemia
discussed here.
We must aim to link the clinical presentation of
ACS more tightly to pathophysiological mechanisms.
More than a century after the introduction of ECG, we
still triage our patients with ACS on the basis of this
mature technology. To move forward in the manage-
ment of ACS, we should strive for a more personal-
ized approach to therapy based on criteria more tightly
linked to the diverse pathophysiological mechanisms
than ECG repolarization abnormalities. We need to
develop and validate soluble and imaging biomarkers
that reflect the underlying mechanism that yields acute
ischemia. Point-of-care assessment of such biomarkers
would help render their use clinically practical in the
triage of patients who present with ACS, with the goal
of sparing some the need for urgent invasive diagnos-
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
ACS that reaches beyond our current dichotomous ap-
proach based on the ECG. If these ventures succeed,
we may one day look back on today’s treatment of ACS
as oversimplified and rather primitive. We should strive
as a community not to be satisfied with our current ap-
proach to ACS management but reach to realize the
promise of a more personalized precision deployment
of treatment strategies, including new ones based on
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
Crea and Libby
September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701164
greater mechanistic understanding of the diverse un-
derlying causes of acute myocardial ischemia.
DISCLOSURES
Dr Libby reports receiving grants from Novartis and holding
scientific advisory board positions for Olatec and Interleukin
Genetics (company no longer operating). Dr Crea reports no
conflicts.
AFFILIATIONS
From Department of Cardiovascular and Thoracic Sciences,
Catholic University, Rome, Italy (F.C.); and Division of
Cardiovascular Medicine, Department of Medicine, Brigham
and Women’s Hospital, Harvard Medical School, Boston,
MA (P.L.).
FOOTNOTES
Circulation is available at http://circ.ahajournals.org.
REFERENCES
	 1.	 Libby P. Molecular bases of the acute coronary syndromes. Circulation.
1995;91:2844–2850.
	 2.	 Libby P. Current concepts of the pathogenesis of the acute coronary syn-
dromes. Circulation. 2001;104:365–372.
	 3.	 Libby P. Mechanisms of acute coronary syndromes and their implications
for therapy. N Engl J Med. 2013;368:2004–2013. doi: 10.1056/NEJM-
ra1216063.
	 4.	 Libby P, Pasterkamp G. Requiem for the “vulnerable plaque.” Eur Heart J.
2015;36:2984–2987. doi: 10.1093/eurheartj/ehv349.
	 5.	Pasterkamp G, den Ruijter HM, Libby P. Temporal shifts in clinical pre-
sentation and underlying mechanisms of atherosclerotic disease. Nat Rev
Cardiol. 2017;14:21–29. doi: 10.1038/nrcardio.2016.166.
	 6.	 Davies MJ, Thomas A. Thrombosis and acute coronary-artery lesions in
sudden cardiac ischemic death. N Engl J Med. 1984;310:1137–1140. doi:
10.1056/NEJM198405033101801.
	 7.	Falk E, Shah PK, Fuster V. Coronary plaque disruption. Circulation.
1995;92:657–671.
	 8.	 Finn AV, Nakano M, Narula J, Kolodgie FD, Virmani R. Concept of vulnera-
ble/unstable plaque. Arterioscler Thromb Vasc Biol. 2010;30:1282–1292.
doi: 10.1161/ATVBAHA.108.179739.
	 9.	 Motoyama S, Ito H, Sarai M, Kondo T, Kawai H, Nagahara Y, Harigaya H, Kan
S, Anno H, Takahashi H, Naruse H, Ishii J, Hecht H, Shaw LJ, Ozaki Y, Narula J.
Plaque characterization by coronary computed tomography angiography
and the likelihood of acute coronary events in mid-term follow-up. J Am
Coll Cardiol. 2015;66:337–346. doi: 10.1016/j.jacc.2015.05.069.
	10.	 Mann J, Davies MJ. Mechanisms of progression in native coronary artery
disease: role of healed plaque disruption. Heart. 1999;82:265–268.
	11.	 Stone GW, Maehara A, Lansky AJ, de Bruyne B, Cristea E, Mintz GS, Meh-
ran R, McPherson J, Farhat N, Marso SP, Parise H, Templin B, White R,
Zhang Z, Serruys PW; PROSPECT Investigators. A prospective natural-his-
tory study of coronary atherosclerosis. N Engl J Med. 2011;364:226–235.
doi: 10.1056/NEJMoa1002358.
	12.	 Kubo T, Maehara A, Mintz GS, Doi H, Tsujita K, Choi SY, Katoh O, Nasu K,
Koenig A, Pieper M, Rogers JH, Wijns W, Böse D, Margolis MP, Moses JW,
Stone GW, Leon MB. The dynamic nature of coronary artery lesion mor-
phology assessed by serial virtual histology intravascular ultrasound tissue
characterization. J Am Coll Cardiol. 2010;55:1590–1597. doi: 10.1016/j.
jacc.2009.07.078.
	13.	 Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circula-
tion. 2002;105:1135–1143.
	14.	 Cristell N, Cianflone D, Durante A, Ammirati E, Vanuzzo D, Banfi M, Calori
G, Latib A, Crea F, Marenzi G, De Metrio M, Moretti L, Li H, Uren NG, Hu
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
myocardial infarction in 41% of cases: a multiethnic case-control study.
J Am Coll Cardiol. 2011;58:2654–2661. doi: 10.1016/j.jacc.2011.08.055.
	15.	 Scalone G, Niccoli G, Refaat H, Vergallo R, Porto I, Leone AM, Burzotta F,
D’Amario D, Liuzzo G, Fracassi F, Trani C, Crea F. Not all plaque ruptures
are born equal: an optical coherence tomography study [published online
ahead of print December 24, 2016]. Eur Heart J Cardiovasc Imaging. doi:
10.1093/ehjci/jew208.
	16.	 Arbustini E, Dal Bello B, Morbini P, Burke AP, Bocciarelli M, Specchia G,
Virmani R. Plaque erosion is a major substrate for coronary thrombosis in
acute myocardial infarction. Heart. 1999;82:269–272.
	17.	 Bentzon JF, Otsuka F, Virmani R, Falk E. Mechanisms of plaque forma-
tion and rupture. Circ Res. 2014;114:1852–1866. doi: 10.1161/CIRCRE-
SAHA.114.302721.
	18.	 Niccoli G, Montone RA, Cataneo L, Cosentino N, Gramegna M, Refaat
H, Porto I, Burzotta F, Trani C, Leone AM, Severino A, Crea F. Morpho-
logical-biohumoral correlations in acute coronary syndromes: patho-
genetic implications. Int J Cardiol. 2014;171:463–466. doi: 10.1016/j.
ijcard.2013.12.238.
	19.	Jia H, Abtahian F, Aguirre AD, Lee S, Chia S, Lowe H, Kato K, Yo-
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
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
patients with acute coronary syndrome by intravascular optical coherence
tomography. J Am Coll Cardiol. 2013;62:1748–1758. doi: 10.1016/j.
jacc.2013.05.071.
	20.	 Crea F, Liuzzo G. Pathogenesis of acute coronary syndromes. J Am Coll
Cardiol. 2013;61:1–11. doi: 10.1016/j.jacc.2012.07.064.
	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
protein in severe unstable angina. N Engl J Med. 1994;331:417–424. doi:
10.1056/NEJM199408183310701.
	22.	 Schönbeck U, Mach F, Sukhova GK, Murphy C, Bonnefoy JY, Fabunmi
RP, Libby P. Regulation of matrix metalloproteinase expression in human
vascular smooth muscle cells by T lymphocytes: a role for CD40 signaling
in plaque rupture? Circ Res. 1997;81:448–454.
	23.	 Liuzzo G, Kopecky SL, Frye RL, O’Fallon WM, Maseri A, Goronzy JJ, Wey-
and CM. Perturbation of the T-cell repertoire in patients with unstable
angina. Circulation. 1999;100:2135–2139.
	24.	 Liuzzo G, Goronzy JJ, Yang H, Kopecky SL, Holmes DR, Frye RL, Weyand
CM. Monoclonal T-cell proliferation and plaque instability in acute coro-
nary syndromes. Circulation. 2000;101:2883–2888.
	25.	 Chen S, Crother TR, Arditi M. Emerging role of IL-17 in atherosclerosis.
J Innate Immun. 2010;2:325–333. doi: 10.1159/000314626.
	26.	Gisterå A, Robertson AK, Andersson J, Ketelhuth DF, Ovchinnikova O,
Nilsson SK, Lundberg AM, Li MO, Flavell RA, Hansson GK. Transforming
growth factor-β signaling in T cells promotes stabilization of atheroscle-
rotic plaques through an interleukin-17-dependent pathway. Sci Transl
Med. 2013;5:196ra100. doi: 10.1126/scitranslmed.3006133.
	27.	Brusko TM, Putnam AL, Bluestone JA. Human regulatory T cells: role
in autoimmune disease and therapeutic opportunities. Immunol Rev.
2008;223:371–390. doi: 10.1111/j.1600-065X.2008.00637.x.
	28.	 Liuzzo G, Montone RA, Gabriele M, Pedicino D, Giglio AF, Trotta F, Galiffa
VA, Previtero M, Severino A, Biasucci LM, Crea F. Identification of unique
adaptive immune system signature in acute coronary syndromes. Int J Car-
diol. 2013;168:564–567. doi: 10.1016/j.ijcard.2013.01.009.
	29.	 Flego D, Severino A, Trotta F, Previtero M, Ucci S, Zara C, Pedicino D, Mas-
saro G, Biasucci LM, Liuzzo G, Crea F. Altered CD31 expression and activity
in helper T cells of acute coronary syndrome patients. Basic Res Cardiol.
2014;109:448. doi: 10.1007/s00395-014-0448-3.
	30.	Flego D, Severino A, Trotta F, Previtero M, Ucci S, Zara C, Massaro G,
Pedicino D, Biasucci LM, Liuzzo G, Crea F. Increased PTPN22 expression
and defective CREB activation impair regulatory T-cell differentiation in
non-ST-segment elevation acute coronary syndromes. J Am Coll Cardiol.
2015;65:1175–1186. doi: 10.1016/j.jacc.2015.01.027.
	31.	 Nidorf SM, Eikelboom JW, Budgeon CA, Thompson PL. Low-dose colchi-
cine for secondary prevention of cardiovascular disease. J Am Coll Cardiol.
2013;61:404–410. doi: 10.1016/j.jacc.2012.10.027.
	32.	Tardif J-C, L’Allier P. Colchicine Cardiovascular Outcomes Trial (COL-
COT). https://clinicaltrials.gov/ct2/show/NCT02551094. 2017. Accessed
June 1, 2017.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
ACS Mechanisms Inform Management
STATEOFTHEART
Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1165
	33.	 Everett BM, Pradhan AD, Solomon DH, Paynter N, Macfadyen J, Zaharris
E, Gupta M, Clearfield M, Libby P, Hasan AA, Glynn RJ, Ridker PM. Ra-
tionale and design of the Cardiovascular Inflammation Reduction Trial:
a test of the inflammatory hypothesis of atherothrombosis. Am Heart J.
2013;166:199–207.e15. doi: 10.1016/j.ahj.2013.03.018.
	34.	 Morton AC, Rothman AM, Greenwood JP, Gunn J, Chase A, Clarke B, Hall
AS, Fox K, Foley C, Banya W, Wang D, Flather MD, Crossman DC. The
effect of interleukin-1 receptor antagonist therapy on markers of inflam-
mation in non-ST elevation acute coronary syndromes: the MRC-ILA Heart
Study. Eur Heart J. 2015;36:377–384. doi: 10.1093/eurheartj/ehu272.
	35.	 Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1β inhibition and the
prevention of recurrent cardiovascular events: rationale and design of the
Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS).
Am Heart J. 2011;162:597–605. doi: 10.1016/j.ahj.2011.06.012.
	36.	 Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne
C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH,
Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z,
Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi
PRF, Troquay RPT, Libby P, Glynn RJ and Group CT. Antiinflammatory
Therapy with Canakinumab for Atherosclerotic Disease [published on-
line ahead of print August 27, 2017]. N Engl J Med. 2017. doi: 10.1056/
NEJMoa1707914.
	37.	 Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P, Glynn RJ, Ridker P,
Lorenzatti A, Krum H, Varigos J, Siostrzonek P, Sinnaeve P, Fonseca F, Nico-
lau J, Gotcheva N, Genest J, Yong H, Urina-Triana M, Milicic D, Cifkova R,
Vettus R, Koenig W, Anker SD, Manolis AJ, Wyss F, Forster T, Sigurdsson A,
Pais P, Fucili A, Ogawa H, Shimokawa H, Veze I, Petrauskiene B, Salvador L,
Kastelein J, Cornel JH, Klemsdal TO, Medina F, Budaj A, Vida-Simiti L, Koba-
lava Z, Otasevic P, Pella D, Lainscak M, Seung K-B, Commerford P, Dellborg
M, Donath M, Hwang J-J, Kultursay H, Flather M, Ballantyne C, Bilazarian S,
Chang W, East C, Everett B, Forgosh L, Glynn R, Harris B, Libby P, Ligueros
M, Thuren T, Bohula E, Charmarthi B, Cheng S, Chou S, Danik J, McMahon
G, Maron B, Ning M, Olenchock B, Pande R, Perlstein T, Pradhan A, Rost N,
Singhal A, Taqueti V, Wei N, Burris H, Cioffi A, Dalseg AM, Ghosh N, Gralow
J, Mayer T, Rugo H, Fowler V, Limaye AP, Cosgrove S, Levine D, Lopes R,
Scott J, Thuren T, Ligueros M, Hilkert R, Tamesby G, Mickel C, Manning B,
Woelcke J, Tan M, Manfreda S, Ponce T, Kam J, Saini R, Banker K, Salko T,
Nandy P, Tawfik R, O'Neil G, Manne S, Jirvankar P, Lal S, Nema D, Jose J,
Collins R, Bailey K, Blumenthal R, Colhoun H, Gersh B and Glynn RJ. Effect
of interleukin-1β inhibition with canakinumab on incident lung cancer in
patients with atherosclerosis: exploratory results from a randomised, dou-
ble-blind, placebo-controlled trial [published online ahead of print August
25, 2017]. The Lancet. doi: 10.1016/S0140-6736(17)32247-X.
	38.	 Fornasa G, Clement M, Groyer E, Gaston AT, Khallou-Laschet J, Morvan
M, Guedj K, Kaveri SV, Tedgui A, Michel JB, Nicoletti A, Caligiuri G. A
CD31-derived peptide prevents angiotensin II-induced atherosclerosis pro-
gression and aneurysm formation. Cardiovasc Res. 2012;94:30–37. doi:
10.1093/cvr/cvs076.
	39.	 Stanford SM, Krishnamurthy D, Falk MD, Messina R, Debnath B, Li S, Liu
T, Kazemi R, Dahl R, He Y, Yu X, Chan AC, Zhang ZY, Barrios AM, Woods
VL Jr, Neamati N, Bottini N. Discovery of a novel series of inhibitors of lym-
phoid tyrosine phosphatase with activity in human T cells. J Med Chem.
2011;54:1640–1654. doi: 10.1021/jm101202j.
	40.	 Putnam AL, Brusko TM, Lee MR, Liu W, Szot GL, Ghosh T, Atkinson MA,
Bluestone JA. Expansion of human regulatory T-cells from patients with
type 1 diabetes. Diabetes. 2009;58:652–662. doi: 10.2337/db08-1168.
	41.	 Shah PK, Chyu KY, Dimayuga PC, Nilsson J. Vaccine for atherosclerosis.
J Am Coll Cardiol. 2014;64:2779–2791. doi: 10.1016/j.jacc.2014.10.018.
	42.	 Koskinas KC, Sukhova GK, Baker AB, Papafaklis MI, Chatzizisis YS, Coskun
AU, Quillard T, Jonas M, Maynard C, Antoniadis AP, Shi GP, Libby P, Edel-
man ER, Feldman CL, Stone PH. Thin-capped atheromata with reduced
collagen content in pigs develop in coronary arterial regions exposed to
persistently low endothelial shear stress. Arterioscler Thromb Vasc Biol.
2013;33:1494–1504. doi: 10.1161/ATVBAHA.112.300827.
	43.	 Chatzizisis YS, Baker AB, Sukhova GK, Koskinas KC, Papafaklis MI, Beigel
R, Jonas M, Coskun AU, Stone BV, Maynard C, Shi GP, Libby P, Feldman CL,
Edelman ER, Stone PH. Augmented expression and activity of extracellular
matrix-degrading enzymes in regions of low endothelial shear stress colo-
calize with coronary atheromata with thin fibrous caps in pigs. Circulation.
2011;123:621–630. doi: 10.1161/CIRCULATIONAHA.110.970038.
	44.	Shiomi M, Ishida T, Kobayashi T, Nitta N, Sonoda A, Yamada S, Koike
T, Kuniyoshi N, Murata K, Hirata K, Ito T, Libby P. Vasospasm of athero-
sclerotic coronary arteries precipitates acute ischemic myocardial damage
in myocardial infarction-prone strain of the Watanabe heritable hyperlip-
idemic rabbits. Arterioscler Thromb Vasc Biol. 2013;33:2518–2523. doi:
10.1161/ATVBAHA.113.301303.
	45.	 Libby P, Nahrendorf M, Swirski FK. Leukocytes link local and systemic in-
flammation in ischemic cardiovascular disease: an expanded “cardiovas-
cular continuum”. J Am Coll Cardiol. 2016;67:1091–1103. doi: 10.1016/j.
jacc.2015.12.048.
	46.	 Burke AP, Farb A, Malcom GT, Liang Y, Smialek JE, Virmani R. Plaque rup-
ture and sudden death related to exertion in men with coronary artery
disease. JAMA. 1999;281:921–926.
	47.	 Janoudi A, Shamoun FE, Kalavakunta JK, Abela GS. Cholesterol crystal in-
duced arterial inflammation and destabilization of atherosclerotic plaque.
Eur Heart J. 2016;37:1959–1967. doi: 10.1093/eurheartj/ehv653.
	48.	 Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG,
Abela GS, Franchi L, Nuñez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA,
Rock KL, Moore KJ, Wright SD, Hornung V, Latz E. NLRP3 inflammasomes
are required for atherogenesis and activated by cholesterol crystals. Na-
ture. 2010;464:1357–1361. doi: 10.1038/nature08938.
	49.	 Nakamura S, Inami S, Murai K, Takano M, Takano H, Asai K, Yasutake
M, Shimizu W, Mizuno K. Relationship between cholesterol crystals and
culprit lesion characteristics in patients with stable coronary artery disease:
an optical coherence tomography study. Clin Res Cardiol. 2014;103:1015–
1021. doi: 10.1007/s00392-014-0748-5.
	50.	 Liu L, Gardecki JA, Nadkarni SK, Toussaint JD, Yagi Y, Bouma BE, Tearney
GJ. Imaging the subcellular structure of human coronary atherosclerosis
using micro-optical coherence tomography. Nat Med. 2011;17:1010–
1014. doi: 10.1038/nm.2409.
	51.	 Loree HM, Tobias BJ, Gibson LJ, Kamm RD, Small DM, Lee RT. Mechanical
properties of model atherosclerotic lesion lipid pools. Arterioscler Thromb.
1994;14:230–234.
	52.	 Huang H, Virmani R, Younis H, Burke AP, Kamm RD, Lee RT. The impact
of calcification on the biomechanical stability of atherosclerotic plaques.
Circulation. 2001;103:1051–1056.
	53.	 Virmani R, Joner M, Sakakura K. Recent highlights of ATVB: calcification.
Arterioscler Thromb Vasc Biol. 2014;34:1329–1332. doi: 10.1161/ATVBA-
HA.114.304000.
	54.	 Ruiz JL, Weinbaum S, Aikawa E, Hutcheson JD. Zooming in on the genesis
of atherosclerotic plaque microcalcifications. J Physiol. 2016;594:2915–
2927. doi: 10.1113/JP271339.
	55.	 Camici PG, Rimoldi OE, Gaemperli O, Libby P. Non-invasive anatomic and
functional imaging of vascular inflammation and unstable plaque. Eur
Heart J. 2012;33:1309–1317. doi: 10.1093/eurheartj/ehs067.
	56.	 Libby P. How does lipid lowering prevent coronary events? New insights
from human imaging trials. Eur Heart J. 2015;36:472–474. doi: 10.1093/
eurheartj/ehu510.
	57.	Abela GS, Vedre A, Janoudi A, Huang R, Durga S, Tamhane U. Effect
of statins on cholesterol crystallization and atherosclerotic plaque sta-
bilization. Am J Cardiol. 2011;107:1710–1717. doi: 10.1016/j.amj-
card.2011.02.336.
	58.	Patel R, Janoudi A, Vedre A, Aziz K, Tamhane U, Rubinstein J, Abela
OG, Berger K, Abela GS. Plaque rupture and thrombosis are reduced
by lowering cholesterol levels and crystallization with ezetimibe and are
correlated with fluorodeoxyglucose positron emission tomography. Ar-
terioscler Thromb Vasc Biol. 2011;31:2007–2014. doi: 10.1161/ATVBA-
HA.111.226167.
	59.	 Zimmer S, Grebe A, Bakke SS, Bode N, Halvorsen B, Ulas T, Skjelland M,
De Nardo D, Labzin LI, Kerksiek A, Hempel C, Heneka MT, Hawxhurst V,
Fitzgerald ML, Trebicka J, Björkhem I, Gustafsson JÅ, Westerterp M, Tall
AR, Wright SD, Espevik T, Schultze JL, Nickenig G, Lütjohann D, Latz E.
Cyclodextrin promotes atherosclerosis regression via macrophage repro-
gramming. Sci Transl Med. 2016;8:333ra50. doi: 10.1126/scitranslmed.
aad6100.
	60.	 Mohammadpour AH, Akhlaghi F. Future of cholesteryl ester transfer pro-
tein (CETP) inhibitors: a pharmacological perspective. Clin Pharmacokinet.
2013;52:615–626. doi: 10.1007/s40262-013-0071-8.
	61.	HPS/TIMI55-REVEAL Collaborative Group. Effects of Anacetrapib in Pa-
tients with Atherosclerotic Vascular Disease [published online ahead of
print August 28, 2017]. N Engl J Med. doi: 10.1056/NEJMoa1706444.
	62.	 Meuwese MC, de Groot E, Duivenvoorden R, Trip MD, Ose L, Maritz FJ, Ba-
sart DC, Kastelein JJ, Habib R, Davidson MH, Zwinderman AH, Schwocho
LR, Stein EA; CAPTIVATE Investigators. ACAT inhibition and progression of
carotid atherosclerosis in patients with familial hypercholesterolemia: the
CAPTIVATE randomized trial. JAMA. 2009;301:1131–1139. doi: 10.1001/
jama.301.11.1131.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
Crea and Libby
September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701166
	63.	Lüscher TF. Substrates of acute coronary syndromes: new insights into
plaque rupture and erosion. Eur Heart J. 2015;36:1347–1349. doi:
10.1093/eurheartj/ehv149.
	64.	 Ferrante G, Nakano M, Prati F, Niccoli G, Mallus MT, Ramazzotti V, Mon-
tone RA, Kolodgie FD, Virmani R, Crea F. High levels of systemic my-
eloperoxidase are associated with coronary plaque erosion in patients
with acute coronary syndromes: a clinicopathological study. Circulation.
2010;122:2505–2513. doi: 10.1161/CIRCULATIONAHA.110.955302.
	65.	 Yahagi K, Kolodgie FD, Lutter C, Mori H, Romero ME, Finn AV, Virmani
R. Pathology of human coronary and carotid artery atherosclerosis and
vascular calcification in diabetes mellitus. Arterioscler Thromb Vasc Biol.
2017;37:191–204. doi: 10.1161/ATVBAHA.116.306256.
	66.	 Hansson GK, Libby P, Tabas I. Inflammation and plaque vulnerability. J In-
tern Med. 2015;278:483–493. doi: 10.1111/joim.12406.
	67.	 Quillard T, Araújo HA, Franck G, Shvartz E, Sukhova G, Libby P. TLR2 and
neutrophils potentiate endothelial stress, apoptosis and detachment: im-
plications for superficial erosion. Eur Heart J. 2015;36:1394–1404. doi:
10.1093/eurheartj/ehv044.
	68.	 Franck G, Mawson T, Sausen G, Salinas M, Masson GS, Cole A, Beltrami-
Moreira M, Chatzizisis Y, Quillard T, Tesmenitsky Y, Shvartz E, Sukhova GK,
Swirski FK, Nahrendorf M, Aikawa E, Croce KJ, Libby P. Flow perturbation
mediates neutrophil recruitment and potentiates endothelial injury via
TLR2 in mice: implications for superficial erosion. Circ Res. 2017;121:31–
42. doi: 10.1161/CIRCRESAHA.117.310694.
	69.	 Kolodgie FD, Burke AP, Wight TN, Virmani R. The accumulation of specific
types of proteoglycans in eroded plaques: a role in coronary thrombosis in
the absence of rupture. Curr Opin Lipidol. 2004;15:575–582.
	70.	 Day AJ. The structure and regulation of hyaluronan-binding proteins. Bio-
chem Soc Trans. 1999;27:115–121.
	71.	 Butler LM, Rainger GE, Nash GB. A role for the endothelial glycosami-
noglycan hyaluronan in neutrophil recruitment by endothelial cells cul-
tured for prolonged periods. Exp Cell Res. 2009;315:3433–3441. doi:
10.1016/j.yexcr.2009.08.012.
	72.	Döring Y, Soehnlein O, Weber C. Neutrophil extracellular traps in ath-
erosclerosis and atherothrombosis. Circ Res. 2017;120:736–743. doi:
10.1161/CIRCRESAHA.116.309692.
	73.	 Libby P. Triglycerides on the rise: should we swap seats on the seesaw? Eur
Heart J. 2015;36:774–776. doi: 10.1093/eurheartj/ehu500.
	74.	 Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzenböck A,
Simon D, Laimer D, Bangert C, Kammerlander A, Mascherbauer J, Winter
MP, Distelmaier K, Adlbrecht C, Preissner KT, Lang IM. Coronary neutro-
phil extracellular trap burden and deoxyribonuclease activity in ST-eleva-
tion acute coronary syndrome are predictors of ST-segment resolution
and infarct size. Circ Res. 2015;116:1182–1192. doi: 10.1161/CIRCRE-
SAHA.116.304944.
	75.	 Aguirre-Alvarado C, Segura-Cabrera A, Velázquez-Quesada I, Hernández-
Esquivel MA, García-Pérez CA, Guerrero-Rodríguez SL, Ruiz-Moreno AJ,
Rodríguez-Moreno A, Pérez-Tapia SM, Velasco-Velázquez MA. Virtual
screening-driven repositioning of etoposide as CD44 antagonist in
breast cancer cells. Oncotarget. 2016;7:23772–23784. doi: 10.18632/
oncotarget.8180.
	76.	 Jia H, Dai J, Hou J, Xing L, Ma L, Liu H, Xu M, Yao Y, Hu S, Yamamoto E,
Lee H, Zhang S, Yu B, Jang IK. Effective anti-thrombotic therapy without
stenting: intravascular optical coherence tomography-based management
in plaque erosion (the EROSION study). Eur Heart J. 2017;38:792–800.
doi: 10.1093/eurheartj/ehw381.
	77.	 Libby P. Superficial erosion and the precision management of acute coro-
nary syndromes: not one-size-fits-all. Eur Heart J. 2017;38:801–803. doi:
10.1093/eurheartj/ehw599.
	78.	 Beltrame JF, Crea F, Kaski JC, Ogawa H, Ong P, Sechtem U, Shimokawa
H, Bairey Merz CN; Coronary Vasomotion Disorders International Study
Group (COVADIS). The who, what, why, when, how and where of vaso-
spastic angina. Circ J. 2016;80:289–298. doi: 10.1253/circj.CJ-15-1202.
	79.	 Ong P, Athanasiadis A, Borgulya G, Voehringer M, Sechtem U. 3-Year fol-
low-up of patients with coronary artery spasm as cause of acute coronary
syndrome: the CASPAR (Coronary Artery Spasm in Patients With Acute
Coronary Syndrome) study follow-up. J Am Coll Cardiol. 2011;57:147–
152. doi: 10.1016/j.jacc.2010.08.626.
	80.	 Sato K, Kaikita K, Nakayama N, Horio E, Yoshimura H, Ono T, Ohba K,
Tsujita K, Kojima S, Tayama S, Hokimoto S, Matsui K, Sugiyama S, Yamabe
H, Ogawa H. Coronary vasomotor response to intracoronary acetylcholine
injection, clinical features, and long-term prognosis in 873 consecutive pa-
tients with coronary spasm: analysis of a single-center study over 20 years.
J Am Heart Assoc. 2013;2:e000227. doi: 10.1161/JAHA.113.000227.
	81.	 Bertrand ME, LaBlanche JM, Tilmant PY, Thieuleux FA, Delforge MR, Carre
AG, Asseman P, Berzin B, Libersa C, Laurent JM. Frequency of provoked
coronary arterial spasm in 1089 consecutive patients undergoing coronary
arteriography. Circulation. 1982;65:1299–1306.
	82.	 Pristipino C, Beltrame JF, Finocchiaro ML, Hattori R, Fujita M, Mongiardo
R, Cianflone D, Sanna T, Sasayama S, Maseri A. Major racial differences
in coronary constrictor response between Japanese and Caucasians with
recent myocardial infarction. Circulation. 2000;101:1102–1108.
	83.	 Camici PG, Crea F. Coronary microvascular dysfunction. N Engl J Med.
2007;356:830–840. doi: 10.1056/NEJMra061889.
	84.	 Galiuto L, De Caterina AR, Porfidia A, Paraggio L, Barchetta S, Locorotondo
G, Rebuzzi AG, Crea F. Reversible coronary microvascular dysfunction: a
common pathogenetic mechanism in apical ballooning or tako-tsubo syn-
drome. Eur Heart J. 2010;31:1319–1327. doi: 10.1093/eurheartj/ehq039.
	85.	 Templin C, Ghadri JR, Diekmann J, Napp LC, Bataiosu DR, Jaguszewski M,
Cammann VL, Sarcon A, Geyer V, Neumann CA, Seifert B, Hellermann J,
Schwyzer M, Eisenhardt K, Jenewein J, Franke J, Katus HA, Burgdorf C,
Schunkert H, Moeller C, Thiele H, Bauersachs J, Tschöpe C, Schultheiss HP,
Laney CA, Rajan L, Michels G, Pfister R, Ukena C, Böhm M, Erbel R, Cuneo
A, Kuck KH, Jacobshagen C, Hasenfuss G, Karakas M, Koenig W, Rott-
bauer W, Said SM, Braun-Dullaeus RC, Cuculi F, Banning A, Fischer TA, Va-
sankari T, Airaksinen KE, Fijalkowski M, Rynkiewicz A, Pawlak M, Opolski
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,
Ford I, Ruschitzka F, Prasad A, Lüscher TF. Clinical features and outcomes
of takotsubo (stress) cardiomyopathy. N Engl J Med. 2015;373:929–938.
doi: 10.1056/NEJMoa1406761.
	86.	 Arrebola-Moreno AL, Arrebola JP, Moral-Ruiz A, Ramirez-Hernandez JA,
Melgares-Moreno R, Kaski JC. Coronary microvascular spasm triggers
transient ischemic left ventricular diastolic abnormalities in patients with
chest pain and angiographically normal coronary arteries. Atherosclerosis.
2014;236:207–214. doi: 10.1016/j.atherosclerosis.2014.07.009.
	87.	 Kaski JC, Maseri A, Vejar M, Crea F, Hackett D. Spontaneous coronary
artery spasm in variant angina is caused by a local hyperreactivity to a
generalized constrictor stimulus. J Am Coll Cardiol. 1989;14:1456–1463.
	88.	 Lanza GA, Pedrotti P, Pasceri V, Lucente M, Crea F, Maseri A. Autonomic
changes associated with spontaneous coronary spasm in patients with
variant angina. J Am Coll Cardiol. 1996;28:1249–1256. doi: 10.1016/
S0735-1097(96)00309-9.
	89.	 Shimokawa H, Tomoike H, Nabeyama S, Yamamoto H, Araki H, Nakamura
M, Ishii Y, Tanaka K. Coronary artery spasm induced in atherosclerotic
miniature swine. Science. 1983;221:560–562.
	90.	 Ito A, Shimokawa H, Nakaike R, Fukai T, Sakata M, Takayanagi T, Egashira K,
Takeshita A. Role of protein kinase C-mediated pathway in the pathogenesis
of coronary artery spasm in a swine model. Circulation. 1994;90:2425–2431.
	91.	 Shimokawa H, Seto M, Katsumata N, Amano M, Kozai T, Yamawaki T, Ku-
wata K, Kandabashi T, Egashira K, Ikegaki I, Asano T, Kaibuchi K, Takeshita
A. Rho-kinase-mediated pathway induces enhanced myosin light chain
phosphorylations in a swine model of coronary artery spasm. Cardiovasc
Res. 1999;43:1029–1039.
	92.	Shimokawa H. 2014 Williams Harvey Lecture: importance of coro-
nary vasomotion abnormalities: from bench to bedside. Eur Heart J.
2014;35:3180–3193. doi: 10.1093/eurheartj/ehu427.
	93.	 Dai Z, Aoki T, Fukumoto Y, Shimokawa H. Coronary perivascular fibrosis
is associated with impairment of coronary blood flow in patients with
non-ischemic heart failure. J Cardiol. 2012;60:416–421. doi: 10.1016/j.
jjcc.2012.06.009.
	94.	 Kaski JC, Crea F, Meran D, Rodriguez L, Araujo L, Chierchia S, Davies G,
Maseri A. Local coronary supersensitivity to diverse vasoconstrictive stimuli
in patients with variant angina. Circulation. 1986;74:1255–1265.
	95.	 Lanza GA, Careri G, Crea F. Mechanisms of coronary artery spasm. Circulation.
2011;124:1774–1782. doi: 10.1161/CIRCULATIONAHA.111.037283.
	96.	 Abbate A, Hamza M, Cassano AD, Melchior R, Roberts C, Grizzard J, Shah
K, Hastillo A, Kasirajan V, Crea F, Lanza GA, Vetrovec GW. Sympathec-
tomy as a treatment for refractory coronary artery spasm. Int J Cardiol.
2012;161:e7–e9. doi: 10.1016/j.ijcard.2012.03.006.
	97.	 Masumoto A, Mohri M, Shimokawa H, Urakami L, Usui M, Takeshita A.
Suppression of coronary artery spasm by the Rho-kinase inhibitor fasudil
in patients with vasospastic angina. Circulation. 2002;105:1545–1547.
	98.	 King K, Grazette LP, Paltoo DN, McDevitt JT, Sia SK, Barrett PM, Apple
FS, Gurbel PA, Weissleder R, Leeds H, Iturriaga EJ, Rao A, Adhikari B,
Desvigne-Nickens P, Galis ZS, Libby P. Point-of-care technologies for preci-
sion cardiovascular care and clinical research: National Heart, Lung, and
Blood Institute Working Group. JACC Basic Transl Sci. 2016;1:73–86. doi:
10.1016/j.jacbts.2016.01.008.
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
Filippo Crea and Peter Libby
Treatment
Acute Coronary Syndromes: The Way Forward From Mechanisms to Precision
Print ISSN: 0009-7322. Online ISSN: 1524-4539
Copyright © 2017 American Heart Association, Inc. All rights reserved.
is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulation
doi: 10.1161/CIRCULATIONAHA.117.029870
2017;136:1155-1166Circulation.
http://circ.ahajournals.org/content/136/12/1155
World Wide Web at:
The online version of this article, along with updated information and services, is located on the
http://circ.ahajournals.org//subscriptions/
is online at:CirculationInformation about subscribing toSubscriptions:
http://www.lww.com/reprints
Information about reprints can be found online at:Reprints:
document.Permissions and Rights Question and Answerthis process is available in the
click Request Permissions in the middle column of the Web page under Services. Further information about
Office. Once the online version of the published article for which permission is being requested is located,
can be obtained via RightsLink, a service of the Copyright Clearance Center, not the EditorialCirculationin
Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:
byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom

More Related Content

What's hot

Magnani2006 myocarditis
Magnani2006 myocarditisMagnani2006 myocarditis
Magnani2006 myocarditis
tommy8998
 
Calcifilaxia iv
Calcifilaxia ivCalcifilaxia iv
Calcifilaxia iv
HOME
 
Acute cardioemboli
Acute cardioemboliAcute cardioemboli
Acute cardioembolimedic
 
Dr Dake presentation ICCCV nov 2011
Dr Dake presentation ICCCV nov 2011Dr Dake presentation ICCCV nov 2011
Dr Dake presentation ICCCV nov 2011
Sylvie Tuslanes
 
Basic pathogenesis of atherothrombosis
Basic pathogenesis of atherothrombosisBasic pathogenesis of atherothrombosis
Basic pathogenesis of atherothrombosis
Anwar Santoso, MD, PhD
 
SLE and cardiovascular manifestations
SLE  and cardiovascular manifestationsSLE  and cardiovascular manifestations
SLE and cardiovascular manifestations
Sujit Sahu
 
V. Hyper Coagulation State
V. Hyper Coagulation StateV. Hyper Coagulation State
V. Hyper Coagulation State
najmaldin saki
 
Lutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent Thrombosis
Lutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent ThrombosisLutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent Thrombosis
Lutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent ThrombosisSunil E. Saith, MD, MPH
 
management of LV thrombus
management of LV thrombusmanagement of LV thrombus
management of LV thrombus
Astrid Pramudya
 
Cardiac pathology
Cardiac pathologyCardiac pathology
Cardiac pathologySpringer
 
Anti gbm disease
Anti gbm diseaseAnti gbm disease
Anti gbm disease
Ajay Kurian
 
Coronary stent thrombosis
Coronary stent thrombosisCoronary stent thrombosis
Coronary stent thrombosis
Ramachandra Barik
 
Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...
Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...
Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...
M. Luisetto Pharm.D.Spec. Pharmacology
 
Thrombocytopenia in icu:True pearls of wisdom by Dr Nilesh Wasekar
Thrombocytopenia in icu:True pearls of wisdom by Dr Nilesh WasekarThrombocytopenia in icu:True pearls of wisdom by Dr Nilesh Wasekar
Thrombocytopenia in icu:True pearls of wisdom by Dr Nilesh Wasekar
Dr Wasekar Nilesh Wasekar
 
Angiogenesis
AngiogenesisAngiogenesis
Angiogenesis
Creative-Bioarray
 

What's hot (19)

Magnani2006 myocarditis
Magnani2006 myocarditisMagnani2006 myocarditis
Magnani2006 myocarditis
 
Fisiopatologia[21630]
Fisiopatologia[21630]Fisiopatologia[21630]
Fisiopatologia[21630]
 
Calcifilaxia iv
Calcifilaxia ivCalcifilaxia iv
Calcifilaxia iv
 
Acute cardioemboli
Acute cardioemboliAcute cardioemboli
Acute cardioemboli
 
Dr Dake presentation ICCCV nov 2011
Dr Dake presentation ICCCV nov 2011Dr Dake presentation ICCCV nov 2011
Dr Dake presentation ICCCV nov 2011
 
Basic pathogenesis of atherothrombosis
Basic pathogenesis of atherothrombosisBasic pathogenesis of atherothrombosis
Basic pathogenesis of atherothrombosis
 
SLE and cardiovascular manifestations
SLE  and cardiovascular manifestationsSLE  and cardiovascular manifestations
SLE and cardiovascular manifestations
 
V. Hyper Coagulation State
V. Hyper Coagulation StateV. Hyper Coagulation State
V. Hyper Coagulation State
 
Lutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent Thrombosis
Lutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent ThrombosisLutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent Thrombosis
Lutheran Research Fair--CYP2C19 and Prothrombin 2010A with In-Stent Thrombosis
 
Visualization of atherosclerotic vulnerable plaque
Visualization of atherosclerotic vulnerable plaqueVisualization of atherosclerotic vulnerable plaque
Visualization of atherosclerotic vulnerable plaque
 
management of LV thrombus
management of LV thrombusmanagement of LV thrombus
management of LV thrombus
 
Cardiac pathology
Cardiac pathologyCardiac pathology
Cardiac pathology
 
Dic 2
Dic    2Dic    2
Dic 2
 
Anti gbm disease
Anti gbm diseaseAnti gbm disease
Anti gbm disease
 
Coronary stent thrombosis
Coronary stent thrombosisCoronary stent thrombosis
Coronary stent thrombosis
 
Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...
Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...
Luisetto m, luca c, f. a. khan, g. r. mashori. sudden heart pathology a new r...
 
Thrombocytopenia in icu:True pearls of wisdom by Dr Nilesh Wasekar
Thrombocytopenia in icu:True pearls of wisdom by Dr Nilesh WasekarThrombocytopenia in icu:True pearls of wisdom by Dr Nilesh Wasekar
Thrombocytopenia in icu:True pearls of wisdom by Dr Nilesh Wasekar
 
Angiogenesis
AngiogenesisAngiogenesis
Angiogenesis
 
Cid
CidCid
Cid
 

Similar to Acute coronary-syndromes-circulation

Ballon aortic valvuloplasty
Ballon aortic valvuloplastyBallon aortic valvuloplasty
Ballon aortic valvuloplasty
Patricio Matovelle
 
Management of carotid artery atherosclerosis
Management of carotid artery atherosclerosisManagement of carotid artery atherosclerosis
Management of carotid artery atherosclerosis
abhilekh srivastava
 
Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...
Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...
Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...
QUESTJOURNAL
 
Coagulacion diseminada intravascular
Coagulacion diseminada intravascularCoagulacion diseminada intravascular
Coagulacion diseminada intravascularmrpienda
 
06 the anesthesia patient with acute coronary syndrome copia
06 the anesthesia patient with acute coronary syndrome copia06 the anesthesia patient with acute coronary syndrome copia
06 the anesthesia patient with acute coronary syndrome copiaUSACHCHSJ
 
Nanomedicina1
Nanomedicina1Nanomedicina1
Nanomedicina1
joanamatux
 
2020 state of the art—high-sensitivity troponins in acute coronary syndromes
2020 state of the art—high-sensitivity troponins in acute coronary syndromes2020 state of the art—high-sensitivity troponins in acute coronary syndromes
2020 state of the art—high-sensitivity troponins in acute coronary syndromes
BryanMielesM
 
Left main disease and bifurcation percutaneous coronary artery disease treatment
Left main disease and bifurcation percutaneous coronary artery disease treatmentLeft main disease and bifurcation percutaneous coronary artery disease treatment
Left main disease and bifurcation percutaneous coronary artery disease treatment
Ramachandra Barik
 
Coronary Aneurysms: What Every Radiologist Should Know
Coronary Aneurysms: What Every Radiologist Should KnowCoronary Aneurysms: What Every Radiologist Should Know
Coronary Aneurysms: What Every Radiologist Should KnowGarry Choy MD MBA
 
Ccsvi and ms ameds centrum
Ccsvi and ms ameds centrumCcsvi and ms ameds centrum
Ccsvi and ms ameds centrumAmeds Centrum
 
Endovascularbrachytherapy
EndovascularbrachytherapyEndovascularbrachytherapy
Endovascularbrachytherapy
Parag Roy
 
In stent retenosis pathophysiology
In stent retenosis pathophysiologyIn stent retenosis pathophysiology
In stent retenosis pathophysiology
Nilesh Tawade
 
Issues in radiological pathology
Issues in radiological pathologyIssues in radiological pathology
Issues in radiological pathology
Professor Yasser Metwally
 
International Journal of Clinical Cardiology & Research
International Journal of Clinical Cardiology & ResearchInternational Journal of Clinical Cardiology & Research
International Journal of Clinical Cardiology & Research
SciRes Literature LLC. | Open Access Journals
 
Perioperative cardiac assesment and interventions
Perioperative cardiac  assesment and interventionsPerioperative cardiac  assesment and interventions
Perioperative cardiac assesment and interventions
NAJEEB ULLAH SOFI
 
Venus obstructive outflow
Venus obstructive outflowVenus obstructive outflow
Venus obstructive outflow
ifrahjaved
 
Characterizing stable coronary plaques with msct angiography
Characterizing stable coronary plaques with msct angiographyCharacterizing stable coronary plaques with msct angiography
Characterizing stable coronary plaques with msct angiography
adinatasatria
 
A Complex Case Of Polianeurysmatic Disease
A Complex Case Of Polianeurysmatic DiseaseA Complex Case Of Polianeurysmatic Disease
A Complex Case Of Polianeurysmatic Disease
Salvatore Ronsivalle
 
Sickle cell ppt
Sickle cell pptSickle cell ppt
Sickle cell ppt
Wahid altaf Sheeba hakak
 
Spontaneous coronary artery dissection
Spontaneous coronary artery dissectionSpontaneous coronary artery dissection
Spontaneous coronary artery dissection
hospital
 

Similar to Acute coronary-syndromes-circulation (20)

Ballon aortic valvuloplasty
Ballon aortic valvuloplastyBallon aortic valvuloplasty
Ballon aortic valvuloplasty
 
Management of carotid artery atherosclerosis
Management of carotid artery atherosclerosisManagement of carotid artery atherosclerosis
Management of carotid artery atherosclerosis
 
Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...
Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...
Evaluation of Occlusive Diseases in Cervical Arteries in Patients of Acute Is...
 
Coagulacion diseminada intravascular
Coagulacion diseminada intravascularCoagulacion diseminada intravascular
Coagulacion diseminada intravascular
 
06 the anesthesia patient with acute coronary syndrome copia
06 the anesthesia patient with acute coronary syndrome copia06 the anesthesia patient with acute coronary syndrome copia
06 the anesthesia patient with acute coronary syndrome copia
 
Nanomedicina1
Nanomedicina1Nanomedicina1
Nanomedicina1
 
2020 state of the art—high-sensitivity troponins in acute coronary syndromes
2020 state of the art—high-sensitivity troponins in acute coronary syndromes2020 state of the art—high-sensitivity troponins in acute coronary syndromes
2020 state of the art—high-sensitivity troponins in acute coronary syndromes
 
Left main disease and bifurcation percutaneous coronary artery disease treatment
Left main disease and bifurcation percutaneous coronary artery disease treatmentLeft main disease and bifurcation percutaneous coronary artery disease treatment
Left main disease and bifurcation percutaneous coronary artery disease treatment
 
Coronary Aneurysms: What Every Radiologist Should Know
Coronary Aneurysms: What Every Radiologist Should KnowCoronary Aneurysms: What Every Radiologist Should Know
Coronary Aneurysms: What Every Radiologist Should Know
 
Ccsvi and ms ameds centrum
Ccsvi and ms ameds centrumCcsvi and ms ameds centrum
Ccsvi and ms ameds centrum
 
Endovascularbrachytherapy
EndovascularbrachytherapyEndovascularbrachytherapy
Endovascularbrachytherapy
 
In stent retenosis pathophysiology
In stent retenosis pathophysiologyIn stent retenosis pathophysiology
In stent retenosis pathophysiology
 
Issues in radiological pathology
Issues in radiological pathologyIssues in radiological pathology
Issues in radiological pathology
 
International Journal of Clinical Cardiology & Research
International Journal of Clinical Cardiology & ResearchInternational Journal of Clinical Cardiology & Research
International Journal of Clinical Cardiology & Research
 
Perioperative cardiac assesment and interventions
Perioperative cardiac  assesment and interventionsPerioperative cardiac  assesment and interventions
Perioperative cardiac assesment and interventions
 
Venus obstructive outflow
Venus obstructive outflowVenus obstructive outflow
Venus obstructive outflow
 
Characterizing stable coronary plaques with msct angiography
Characterizing stable coronary plaques with msct angiographyCharacterizing stable coronary plaques with msct angiography
Characterizing stable coronary plaques with msct angiography
 
A Complex Case Of Polianeurysmatic Disease
A Complex Case Of Polianeurysmatic DiseaseA Complex Case Of Polianeurysmatic Disease
A Complex Case Of Polianeurysmatic Disease
 
Sickle cell ppt
Sickle cell pptSickle cell ppt
Sickle cell ppt
 
Spontaneous coronary artery dissection
Spontaneous coronary artery dissectionSpontaneous coronary artery dissection
Spontaneous coronary artery dissection
 

Recently uploaded

POST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its managementPOST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its management
touseefaziz1
 
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptxMaxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
kevinkariuki227
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Saeid Safari
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
aljamhori teaching hospital
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Dr KHALID B.M
 
Are There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdfAre There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdf
Little Cross Family Clinic
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
MedicoseAcademics
 
Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...
Sujoy Dasgupta
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Savita Shen $i11
 
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
bkling
 
Prix Galien International 2024 Forum Program
Prix Galien International 2024 Forum ProgramPrix Galien International 2024 Forum Program
Prix Galien International 2024 Forum Program
Levi Shapiro
 
ARTIFICIAL INTELLIGENCE IN HEALTHCARE.pdf
ARTIFICIAL INTELLIGENCE IN  HEALTHCARE.pdfARTIFICIAL INTELLIGENCE IN  HEALTHCARE.pdf
ARTIFICIAL INTELLIGENCE IN HEALTHCARE.pdf
Anujkumaranit
 
Flu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore KarnatakaFlu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore Karnataka
addon Scans
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
Shweta
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
MedicoseAcademics
 
BRACHYTHERAPY OVERVIEW AND APPLICATORS
BRACHYTHERAPY OVERVIEW  AND  APPLICATORSBRACHYTHERAPY OVERVIEW  AND  APPLICATORS
BRACHYTHERAPY OVERVIEW AND APPLICATORS
Krishan Murari
 
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
i3 Health
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
DR SETH JOTHAM
 

Recently uploaded (20)

POST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its managementPOST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its management
 
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptxMaxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
Maxilla, Mandible & Hyoid Bone & Clinical Correlations by Dr. RIG.pptx
 
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
TEST BANK for Operations Management, 14th Edition by William J. Stevenson, Ve...
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
 
Are There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdfAre There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdf
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
 
Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...Couples presenting to the infertility clinic- Do they really have infertility...
Couples presenting to the infertility clinic- Do they really have infertility...
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
 
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
 
Prix Galien International 2024 Forum Program
Prix Galien International 2024 Forum ProgramPrix Galien International 2024 Forum Program
Prix Galien International 2024 Forum Program
 
ARTIFICIAL INTELLIGENCE IN HEALTHCARE.pdf
ARTIFICIAL INTELLIGENCE IN  HEALTHCARE.pdfARTIFICIAL INTELLIGENCE IN  HEALTHCARE.pdf
ARTIFICIAL INTELLIGENCE IN HEALTHCARE.pdf
 
Flu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore KarnatakaFlu Vaccine Alert in Bangalore Karnataka
Flu Vaccine Alert in Bangalore Karnataka
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
 
BRACHYTHERAPY OVERVIEW AND APPLICATORS
BRACHYTHERAPY OVERVIEW  AND  APPLICATORSBRACHYTHERAPY OVERVIEW  AND  APPLICATORS
BRACHYTHERAPY OVERVIEW AND APPLICATORS
 
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
 

Acute coronary-syndromes-circulation

  • 1. STATEOFTHEART Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1155 Filippo Crea, MD Peter Libby, MD ABSTRACT: Well into the 21st century, we still triage acute myocardial infarction on the basis of the presence or absence of ST-segment elevation, a century-old technology. Meanwhile, we have learned a great 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. Acute Coronary Syndromes The Way Forward From Mechanisms to Precision Treatment © 2017 American Heart Association, Inc. Correspondence to: Peter Libby, MD, Brigham and Women’s Hospital, Harvard Medical School, 77 Ave Louis Pasteur, NRB-741-G, Boston, MA 02115. E-mail plibby@ bwh.harvard.edu IN DEPTH Key Words: erosion ◼ inflammation ◼ microvessels ◼ myocardial infarction ◼ plaque ◼ plaque, atherosclerotic ◼ thrombosis byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 2. Crea and Libby September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701156 W illem Einthoven introduced electrocardiogra- phy at the dawn of the 20th century. Well into the 21st century, we still triage acute myocar- dial infarction on the basis of this venerable technology: ST segments up or not. Although this approach remains the appropriate evidence-based strategy today, aiming toward the future goal of more individualized therapy, can we apply a more pathophysiologically based cat- egorization of the acute coronary syndromes (ACS)? In- deed, we have learned much about the mechanisms of ACS since we last reviewed this topic in these pages.1,2 Moreover, the human disease of atherosclerosis has evolved considerably in the interim.3–5 This review aims to work toward a categorization of the ACS that reach- es beyond equating all ACS with plaque rupture or fis- sure, guided by the latest insights into the underlying mechanisms of ACS, and points a way forward toward the eventual clinical application of these considerations. Postmortem studies carried out in the 1980s pro- posed that plaque rupture (also sometimes referred to as fissure) caused most fatal myocardial infarctions.6 These findings led to the notion of the vulnerable or high-risk plaque characterized by a large central lipid core, an abundance of inflammatory cells, a paucity of smooth muscle cells (SMCs), and a thin fibrous cap.7 These observations spawned the widely accepted con- cept that instability of coronary atheromata resulted from fissuring of a thin-capped fibroatheroma caused by a weakening of its collagen structure wrought by inflammatory mechanisms.8 This concept stimulated manifold attempts to de- velop methods to detect the vulnerable plaque, a quest predicated on the postulate that local interventions could preclude plaque thrombosis and possibly prevent ACS. However, attempts to identify vulnerable plaques proved disappointing because of their low predictive value.9 Pathological studies of human coronary arter- 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 as determined by intravascular ultrasound interroga- tion actually cause clinical events during over 3 years of follow-up.11 Thus, most thin-capped fibroatheroma are clinically quite stable, a realization that renders vulnerable plaque a misnomer. Indeed, plaques with thin-capped fibroatheroma morphology as assessed by radiofrequency backscatter data from intravascular ultrasound (virtual histology) commonly convert to an apparently more stable character during a 1-year fol- low-up period.12 Such evolution in plaques may reflect mutability during the natural history of human athero- sclerosis but could also reflect adherence of many of these patients to a contemporary secondary preven- tion regimen, including smoking cessation, statins, and agents that target the renin-angiotensin axis. Moreover, although we have witnessed wide accep- tance of the formerly controversial concept that inflam- mation contributes to atherogenesis,13 inflammation may not drive all transitions from stable atherosclerosis to acute thrombotic events. In a large study, about half of ACS occurred in the presence of normal levels of C-reactive protein (CRP), a marker of inflammation.14 Another recent study using optical coherence tomog- raphy (OCT) demonstrated that among patients with ACS and plaque rupture, two thirds of patients had im- aging evidence of inflammatory cell infiltration in the region of the ruptured fibrous cap but one third did not and the latter had lower levels of CRP.15 In addition, plaque erosion causes up to a third of ACS in the cur- rent era.16,17 Finally, about one fifth of ACS occur in the apparent absence of coronary thrombosis, suggesting that functional alterations beyond thrombus formation can contribute to ACS pathogenesis.18,19 The ensemble of postmortem studies and in vivo studies using intravascular imaging point to 4 patho- logical pathways to ACS. Although these mechanisms may overlap and coexist in some patients, we present them separately for heuristic simplicity: plaque rupture with systemic inflammation, plaque rupture without systemic inflammation, plaque erosion, and plaque without thrombus (Figure 1).20 This review considers the causes of these 4 predomi- nant pathways to ACS. To strive for a more precision approach, we explore the therapeutic implications of these distinct mechanisms that provoke ACS. Moreover, we advocate the future goal to move beyond triage of therapy based merely on electrocardiographic current of injury by harnessing the recent advances in arterial biology and human studies that shed new mechanis- tic light on ACS pathogenesis. Further improvements in the appropriate deployment of technologies and in outcomes should evolve from a more mechanistically based classification of ACS. PLAQUE RUPTURE WITH SYSTEMIC INFLAMMATION Mechanisms Several studies have implicated systemic inflammation in ACS as assessed by the biomarker CRP (Figure 1A).21 Laboratory studies and observations on human plaques point to inflammatory mechanisms as key regulators of the fragility of the fibrous cap and of the thrombogenic potential of the lipid core (Figure 2). Macrophages likely pave the way for the rupture of the fibrous cap of the plaque: When activated, these cells elaborate enzymes that degrade all components of the arterial extracellular matrix. These enzymes include matrix metalloproteinases and certain cathepsins. Multiple mechanisms regulate these matrix-degrading proteinases: transcription, trans- byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 3. ACS Mechanisms Inform Management STATEOFTHEART Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1157 lation, activation of zymogen precursors, and balance with endogenous inhibitors such as the tissue inhibitors of matrix metalloproteinases or the cystatins. Thus, in- creased amounts of activated proteinases or reduced lev- els of their corresponding inhibitors can enhance break- down of the extracellular matrix of plaque.22 Adaptive immunity also appears altered in coronary plaque instability.23 Patients with ACS have an increased population of particularly proinflammatory CD4+ T cells characterized by low cell surface expression of CD28, a costimulatory molecule critically involved in determin- ing the outcome of antigen recognition by T cells.24 ACS also profoundly perturb the numbers of 2 other circulat- ing T-cell subsets: type 17 helper T cells and CD4+ CD25+ regulatory T cells (Tregs ). The net role of interleukin (IL)-17 in atherosclerosis remains controversial, however; some experimental studies in mice support a predominantly proatherogenic function for IL-17, yet type 17 helper T cells activated in plaques promote the formation of thick collagen fibers, which might increase plaque sta- bility.25,26 Tregs normally help to maintain homeostasis of cells involved in adaptive immunity, including antigen- presenting cells and effector T cells. Tregs mediate these modulatory effects by contact-dependent suppression or by releasing anti-inflammatory cytokines such IL- 10 or transforming growth factor-β1.27 Patients with ACS have a reduced number and suppressive function of circulating Tregs compared with patients with stable angina and healthy control subjects.28 The molecular mechanisms responsible for the T-cell dysregulation in ACS remain largely unknown. Recent work highlights the importance of the strength of the stimulation of the T-cell receptor for antigen in the differentiation of helper T-cell subsets. The altered activity observed in ACS of proteins involved in the regulation of upstream T-cell receptor for antigen signaling such as CD31 and protein tyrosine phosphatase N22 may modulate T-cell functions.29,30 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 non–ST-segment–elevation 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. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 4. Crea and Libby September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701158 Therapeutic Implications Several anti-inflammatory treatments might provide benefit in this setting. Colchicine, a time-honored anti-inflammatory drug, reduced major cardiovascular events in a medium-sized, unblinded randomized study in patients with stable ischemic heart disease.31 Two tri- als underway are evaluating colchicine treatment in pa- tients with coronary artery disease, although not in the acute phase.32 A phase III trial is testing the potential beneficial effects of low-dose methotrexate in patients after myocardial infarction or with coronary artery dis- ease and diabetes mellitus or elements of the metabolic syndrome.33 Targeting proinflammatory cytokines offers an- other approach. Because of the potentially detrimen- tal effects on cardiovascular risk profile or the lack of compelling evidence of benefit in experimental ath- erosclerosis, monoclonal antibodies that neutralize tu- mor necrosis factor-α, interferon-γ, or IL-17 may not prove appropriate for treating atherosclerosis. In a small (n=182) phase II study, the IL-1 receptor antagonist anakinra reduced the area under the curve of high- sensitivity CRP in patients in the 7 days after presenta- tion with ACS.34 A large (n10 000) phase III trial with clinical end points that tested a fully humanized anti– IL-1β monoclonal antibody, canakinumab, in patients with previous myocardial infarction and CRP levels 2 mg/L showed a significant 15% reduction in the prima- ry endpoint of MI, stroke, or death, and a 17% fall in a secondary endpoint that included urgent revascular- ization. Indeed, revascularization procedures declined 30% in the group that received the interleukin-1β neu- tralizing antibody canakinumab 150mg subcutaneously every 3 months. This intervention trial substantiates the causal contribution of inflammation to atherosclerotic events.35-37 T-cell activation signaling offers possible targets. A synthetic CD31-derived peptide can suppresses im- munity in vivo by restoring an inhibitory pathway to a CD31-negative T-cell subset.38 Moreover, the develop- ment of a novel series of inhibitors of the phosphatase protein tyrosine phosphatase N22 might contribute to a better understanding of its role in immune dysregula- tion in patients with ACS.39 Therapeutic strategies that 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-γ, interferon-γ; TGFβ, transforming growth factor-β; and TNF, tumor necrosis factor. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 5. ACS Mechanisms Inform Management STATEOFTHEART Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1159 target Treg include administration of low doses of IL-2 and infusion of autologous Tregs that have undergone differentiation and expansion in vitro.40 Various vaccina- tion strategies also can limit experimental atherogen- esis and may act by augmenting the activity of Treg .41 PLAQUE RUPTURE WITHOUT SYSTEMIC INFLAMMATION Mechanisms When plaque rupture occurs in the absence of systemic inflammatory activation, other mechanisms may con- tribute to pathogenesis, including extreme emotional disturbance ranging from external events of short dura- tion such as earthquakes and a beloved team’s loss of a football (soccer) match to acute manifestations of more long-term emotional disturbances (Figure 1B). Intense physical exertion and local mechanical stress at the level of the artery wall, either increased circumferential stress or reduced shear stress, may also predispose to plaque rupture.42 In addition, subclinical inflammation in the microenvironment of the culprit stenosis might com- pound the chain of events leading to coronary instabil- ity, although the triggers for and effectors of such local inflammation may differ from those that operate in patients with systemic inflammation.43 Although many studies have clarified the molecular mechanisms lead- ing to coronary instability in individuals with systemic inflammation (eg, gauged by CRP elevations), patients without systemic inflammation have undergone less extensive investigation. The precise causes of instability remain poorly understood, providing a strong stimulus for further research. The possible relationship between psychological stress and plaque rupture may relate to sympathetic nervous system activation and catecholamine release associated with an increase in heart rate, blood pres- sure, and coronary vasoconstriction favoring plaque dis- ruption and platelet activation, hypercoagulability, and intense coronary microvascular constriction.44 More- over, β3 -adrenergic stimulation can stimulate release from the bone marrow of proinflammatory monocytes that can home to experimental atheromata and amplify local inflammation.45 Although physical or emotional stress per se may not suffice to cause coronary throm- bosis, it might trigger instability in plaques already pre- 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)-1β and IL-18. Large crystals might also cause mechanical disruption of the fibrous cap. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 6. Crea and Libby September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701160 Local changes in the equilibrium between esterified and free cholesterol might promote plaque rupture47 (Fig- ure 3). Cholesterol crystal formation in the lipid core could heighten the risk of plaque rupture and thrombosis and can coactivate the inflammasome, an intracellular multi- meric complex that generates active IL-1β and IL-18.48 OCT has colocalized cholesterol crystals in human coronary arteries with thin-capped plaques.49 The devel- opment of micro-OCT with a 2-μm resolution may shed new light on this potential mechanism of instability by assessing its occurrence in vivo.50 Cholesterol crystals might also theoretically resist plaque rupture by increas- ing the stiffness of the lipid pool, although this mecha- nism probably contributes little to plaque stability.51 The role of calcium mineral as a causal factor in ACS remains controversial. Large collections of calcified tissue do not augment the biomechanical instability of plaques or as- sociate with lesions that provoke ACS.52,53 Small foci of calcification within the atherosclerotic intima, some caus- ing spotty calcification visible on computed tomography, can also introduce biomechanical inhomogeneities that can favor plaque destabilization.3,54,55 Therapeutic Implications Because it is difficult to limit environmental, physical, or emotional triggers, the altered plaque characteristics produced by intensive lipid-lowering treatment might protect against this form of plaque destabilization.56 In particular, statins and ezetimibe may interfere with cho- lesterol crystal formation.57,58 Cyclodextrin can solubilize cholesterol, limits experimental atherogenesis, and thus might provide a pharmacological approach to combat- ting cholesterol crystal accumulation in plaques.59 Enhancement of cholesterol efflux might promote plaque stabilization, but strategies to enhance this process by manipulation of high-density lipoprotein concentra- tions or by administration of apolipoprotein A-1 have gen- erally failed to confer clinical benefit.60 A large outcome trial with anacetrapib produced a modest event reduction, but since this agent not only raises high-density lipopro- tein but also lowers low-density lipoprotein renders it dif- ficult to attribute event reduction to raising high-density lipoprotein.61 In addition, the inhibition of cholesteryl ester hydrolase, an enzyme that converts cholesteryl esters to free cholesterol, might prevent cholesterol crystallization. Inhibition of acetyl-CoA acetyltransferase-1, which in con- trast promotes cholesterol crystallization, increased ath- eroma volume and major cardiovascular events.62 PLAQUE EROSION Mechanisms Current evidence suggests that mechanisms of plaque disruption distinct from macrophage-driven rupture of the fibrous cap of plaque can commonly cause ACS.63 The mechanism of plaque thrombosis described by pa- thologists as superficial erosion appears not to involve inflammation mediated by macrophages, as in the case of fibrous cap fracture (Figure 1C). Superficial erosion complicates lesions with a distinct epidemiology and morphology and involves pathophysiological mecha- nisms that differ from rupture of the fibrous cap. In- deed, neutrophil activation seems to play a pivotal role in thrombosis due to plaque erosion.4 Patients who presented with ACS associated with plaque erosion defined by OCT had higher systemic myeloperoxidase levels compared with patients with plaque rupture. Moreover, in postmortem coronary artery specimens, luminal thrombi superimposed on eroded plaques con- tain many more myeloperoxidase-positive cells than thrombi associated with ruptured plaques64 (Figure 4). Autopsy studies suggest that fatal plaque erosion as- sociates more commonly with hypertriglyceridemia, dia- betes mellitus, female sex, and old age.65 The morphology of lesions that produce thrombosis caused by superficial erosion differs radically from the features associated with fibrous cap rupture. Superficially eroded lesions contain few macrophages or T lymphocytes, inflammatory cells considered pathogenic in plaque rupture. Rather than depleted collagen, the lesions associated with superficial erosion contain abundant proteoglycan and glycosami- noglycans. Instead of a paucity of SMCs, eroded lesions can contain abundant arterial SMCs. Plaques complicat- ed by fibrous cap fracture typically harbor large necrotic, lipid-rich cores. In contrast, the more fibrous lesions asso- ciated with superficial erosion generally lack prominent central lipid cores.66 A loss of intimal endothelial cells de- fines lesions that provoke thrombosis by the mechanism denoted as superficial erosion. Recent work has implicated engagement of the innate immune receptor Toll-like receptor 2 in promoting the susceptibility of endothelial cells to apoptotic stimuli. Hy- aluronic acid, a prominent constituent of the extracellular matrix of eroded lesions, could serve as an endogenous ligand of Toll-like receptor 2, implicated in promoting endothelial cell apoptosis.67 Experiments that have evalu- ated gain and loss of Toll-like receptor 2 function in mice support these observations.68 Indeed, hyaluronan and its receptor, CD44, colocalize along the plaque/thrombus interface in eroded plaque but not in ruptured or stable plaque.69 Accumulation of hyaluronan and expression of CD44 along the plaque/thrombus interface of erod- ed plaques may promote de-endothelization, resulting in CD44-dependent platelet adhesion and subsequent thrombus formation, mediated in part by a direct action of hyaluronan on fibrin polymerization.70,71 We have hypothesized that thrombosis caused by su- perficial erosion occurs in 2 phases. The first hit, mediat- ed, for example, by Toll-like receptor 2, would jeopardize endothelial viability or adherence, leading to a breach in byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 7. ACS Mechanisms Inform Management STATEOFTHEART Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1161 the integrity of the endothelial monolayer overlying the atherosclerotic plaque. The denuded patch on the intimal surface would then attract platelets that could undergo activation by contact with collagen and other compo- nents of the arterial extracellular matrix usually protected 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: 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 lipoprotein–lowering therapies.73 Moreover, therapies that destabilize neutrophil extra- cellular traps such as deoxyribonuclease administration might limit the propagation of thrombi resulting from superficial erosion.74 Receptors such as CD44 expressed by neutrophils may bind hyaluronan and glycosamino- glycan enriched in plaques complicated by erosion and furnish a potential therapeutic target.75 Jia et al76 recently reported a series of patients with ACS with OCT-defined erosion and treated with anti- coagulant/antiplatelet therapy rather that mechanical revascularization. About 80% of the patients showed a 50% reduction of thrombus volume after 1 month. More than a third of patients lacked detectable thrombus after 1 month. This study suggests that pharmacological rather than interventional treatment can effectively man- age ACS caused by superficial erosion, a proposition that merits and requires rigorous testing in outcome trials.77 PLAQUE WITHOUT THROMBUS Mechanisms In patients with ACS without plaque thrombus, a func- tional alteration of coronary circulation likely causes Figure 4. Pathways implicated in arterial thrombosis caused by superficial erosion. Stimuli such as disturbed flow or engagement of innate im- mune receptors such as Toll-like receptor-2 (TLR2) can activate the endothelial cells that line the arterial intima. These cells can undergo cell death, eg, by apoptosis, depicted by the cell with the interrupted membrane and pyknotic nucleus. Injured or dy- ing endothelial cells can desquamate, uncovering the basement membrane. Neutrophils attracted by chemokines produced by activated endothelial cells can congregate on the denuded intima and can in turn degranulate and die, releasing neutro- phil extracellular traps (NETs). These strands of extruded DNA can bind the contents of the neutrophil granules and other proteins, eg, myeloperoxidase or tissue factor. The NETs can constitute a solid-state reactor that generates oxidants such as hypochlorous acid and stimulates coagulation locally. Platelets interacting with the basement membrane can activate, release their granular contents, including chemokines that can recruit further leukocytes, and form the nidus of a white thrombus. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 8. Crea and Libby September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701162 acute ischemia involving large epicardial coronary arteries or the coronary microcirculation (Figure 1D). Epicardial coronary vasospasm may occur in patients in whom coronary angiography does not demonstrate an obstructive atherosclerotic plaque.78 In the CASPAR study (Coronary Artery Spasm in Patients With Acute Coronary Syndrome), coronary angiography failed to show culprit lesions in ≈30% of patients with sus- pected ACS.79 Intracoronary acetylcholine administra- tion elicited coronary spasm in nearly 50% of these patients. Similar findings pertained to a Japanese population.80 Coronary artery spasm also may cause coronary instability in patients with obstructive ath- erosclerosis. Indeed, a classic study by Bertrand et al81 found that ergonovine induced spasm in 20% of pa- tients with recent myocardial infarction and in 40% of patients with unstable angina. In a more recent study, acetylcholine induced spasm in 20% of white patients and in 60% of patients with myocardial infarction within the previous 14 days.82 Japanese people have a higher prevalence of coronary spasm than whites for unknown reasons. Microvascular spasm also can cause myocardial isch- emia.83 This mechanism likely operates in patients with Takotsubo cardiomyopathy,84 which frequently occurs in the absence of obstructive atherosclerosis, although ≈15% of these patients exhibit concomitant obstructive atherosclerosis.85 Microvascular spasm can also contrib- ute to ischemia in the face of angiographically normal- appearing epicardial coronary arteries. Microvascular ischemia often manifests as angina pectoris but can also cause non–ST-segment–elevation ACS as defined by electrocardiographic alterations and biomarker evi- dence of myocyte injury.86 Coronary spasm can also contribute to plaque instability by causing endothelial damage, as shown in an infarction-prone strain of the Watanabe heritable hyperlipidemic rabbit.44 Either macrovascular or microvascular spasm can result from impaired vasodilatation or from vasocon- 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. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 9. ACS Mechanisms Inform Management STATEOFTHEART Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1163 (Figure  5).87,88 Shimokawa et al89 demonstrated SMC hyperreactivity in a coronary segment in pigs after adventitial exposure of a coronary segment to an in- flammatory stimulus (IL-1β). Subsequent experimental and clinical data suggested that an increase in Rho- kinase activity was a major mechanism of SMC hy- perreactivity.90–92 Perivascular fibrosis in the coronary microvasculature can contribute to myocardial isch- emia.93 This observation highlights the potential contri- bution of fixed structural changes in coronary arterioles beyond impaired endothelial vasodilator function as a contributor to myocardial ischemia in the absence of obstructive disease of the epicardial coronary arteries. Therapeutic Implications Several vasoconstrictor substances implicated as trig- gers of epicardial coronary spasm may cause ischemia in the same patient under different conditions.94 Accord- ingly, the vasoactive substances themselves may not re- spond to therapeutic targeting. The current treatment of epicardial spasm uses nonspecific vasodilators such as long-acting nitrates and calcium channel blockers.95 Nitrates and calcium channel antagonists may alleviate ischemia caused by vasospasm. However, we need to identify the molecular alterations responsible for SMC hyperreactivity because many patients do not respond satisfactorily to standard doses of vasodilators.96 Knowl- edge of the postreceptor mechanisms responsible for SMC hyperreactivity and epicardial coronary spasm might enable the development of new therapeutic op- tions for patients who present poor clinical response to nonspecific vasodilator therapy. Fasudil, a Rho kinase inhibitor, reduces the rate of coronary spasm episodes in patients with vasospastic angina.97 Similarly, further efforts are warranted to unravel the molecular mecha- nisms responsible for coronary microvascular dysfunc- tion in Takotsubo cardiomyopathy and in ischemia with normal coronary arteries.20 The observation that Tako­ tsubo cardiomyopathy may have worse outcomes than previously considered and the challenges of managing patients with episodic ischemia caused by microvascu- lar dysfunction render this quest urgent.85 Preventing or reversing perivascular fibrosis in the coronary arterial microvasculature represents another novel target be- yond traditional antiatherosclerotic therapies. CONCLUSIONS AND PERSPECTIVES A dichotomous approach based on the ECG has tradi- tionally dominated our clinical management of ACS: ST-segment–myocardial infarction versus non–ST- segment–myocardial infarction. Our thinking concern- ing the pathophysiological bases of the ACS has also evolved separately and, in parallel, has been dominated by the concept of the vulnerable plaque as a substrate for fibrous cap rupture or of a minority mechanism: superficial erosion. We and others have championed and explored the role of inflammation as a pathway to the ACS. However, we now have a greater appreciation that not all cases of ACS fit this mold. To move forward, we need to remain vigilant not to cling to our cherished no- tions and to remain open to shifts in the nature of the diseases we treat and to new vistas opened by scientific advances. As discussed here, we need to consider more deeply in the current era arterial spasm, rupture with- out inflammation, and distinct aspects of innate im- munity or dyslipidemia as instigators of plaque erosion. The complexity of clinical ACS surpasses our traditional categorization of this condition. We should evolve be- yond the current constrained concepts by expanding our understanding. We have achieved considerable mastery of plaque rupture with inflammation from a mechanistic and therapeutic viewpoint. Control of traditional risk fac- tors such as hypercholesterolemia, hypertension, and smoking addresses the pathophysiology of this mecha- nism of ACS. The other categories that we discuss here may account for an increasing proportion of ACS, now that we are addressing more effectively the traditional risk factors.5 We should remain open to the possibility, even likelihood, that tailored therapeutic approaches may apply to the prevention and treatment of the less well-recognized pathways to acute myocardial ischemia discussed here. We must aim to link the clinical presentation of ACS more tightly to pathophysiological mechanisms. More than a century after the introduction of ECG, we still triage our patients with ACS on the basis of this mature technology. To move forward in the manage- ment of ACS, we should strive for a more personal- ized approach to therapy based on criteria more tightly linked to the diverse pathophysiological mechanisms than ECG repolarization abnormalities. We need to develop and validate soluble and imaging biomarkers that reflect the underlying mechanism that yields acute ischemia. Point-of-care assessment of such biomarkers would help render their use clinically practical in the triage of patients who present with ACS, with the goal of sparing some the need for urgent invasive diagnos- 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 ACS that reaches beyond our current dichotomous ap- proach based on the ECG. If these ventures succeed, we may one day look back on today’s treatment of ACS as oversimplified and rather primitive. We should strive as a community not to be satisfied with our current ap- proach to ACS management but reach to realize the promise of a more personalized precision deployment of treatment strategies, including new ones based on byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 10. Crea and Libby September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701164 greater mechanistic understanding of the diverse un- derlying causes of acute myocardial ischemia. DISCLOSURES Dr Libby reports receiving grants from Novartis and holding scientific advisory board positions for Olatec and Interleukin Genetics (company no longer operating). Dr Crea reports no conflicts. AFFILIATIONS From Department of Cardiovascular and Thoracic Sciences, Catholic University, Rome, Italy (F.C.); and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (P.L.). FOOTNOTES Circulation is available at http://circ.ahajournals.org. REFERENCES 1. Libby P. Molecular bases of the acute coronary syndromes. Circulation. 1995;91:2844–2850. 2. Libby P. Current concepts of the pathogenesis of the acute coronary syn- dromes. Circulation. 2001;104:365–372. 3. Libby P. Mechanisms of acute coronary syndromes and their implications for therapy. N Engl J Med. 2013;368:2004–2013. doi: 10.1056/NEJM- ra1216063. 4. Libby P, Pasterkamp G. Requiem for the “vulnerable plaque.” Eur Heart J. 2015;36:2984–2987. doi: 10.1093/eurheartj/ehv349. 5. Pasterkamp G, den Ruijter HM, Libby P. Temporal shifts in clinical pre- sentation and underlying mechanisms of atherosclerotic disease. Nat Rev Cardiol. 2017;14:21–29. doi: 10.1038/nrcardio.2016.166. 6. Davies MJ, Thomas A. Thrombosis and acute coronary-artery lesions in sudden cardiac ischemic death. N Engl J Med. 1984;310:1137–1140. doi: 10.1056/NEJM198405033101801. 7. Falk E, Shah PK, Fuster V. Coronary plaque disruption. Circulation. 1995;92:657–671. 8. Finn AV, Nakano M, Narula J, Kolodgie FD, Virmani R. Concept of vulnera- ble/unstable plaque. Arterioscler Thromb Vasc Biol. 2010;30:1282–1292. doi: 10.1161/ATVBAHA.108.179739. 9. Motoyama S, Ito H, Sarai M, Kondo T, Kawai H, Nagahara Y, Harigaya H, Kan S, Anno H, Takahashi H, Naruse H, Ishii J, Hecht H, Shaw LJ, Ozaki Y, Narula J. Plaque characterization by coronary computed tomography angiography and the likelihood of acute coronary events in mid-term follow-up. J Am Coll Cardiol. 2015;66:337–346. doi: 10.1016/j.jacc.2015.05.069. 10. Mann J, Davies MJ. Mechanisms of progression in native coronary artery disease: role of healed plaque disruption. Heart. 1999;82:265–268. 11. Stone GW, Maehara A, Lansky AJ, de Bruyne B, Cristea E, Mintz GS, Meh- ran R, McPherson J, Farhat N, Marso SP, Parise H, Templin B, White R, Zhang Z, Serruys PW; PROSPECT Investigators. A prospective natural-his- tory study of coronary atherosclerosis. N Engl J Med. 2011;364:226–235. doi: 10.1056/NEJMoa1002358. 12. Kubo T, Maehara A, Mintz GS, Doi H, Tsujita K, Choi SY, Katoh O, Nasu K, Koenig A, Pieper M, Rogers JH, Wijns W, Böse D, Margolis MP, Moses JW, Stone GW, Leon MB. The dynamic nature of coronary artery lesion mor- phology assessed by serial virtual histology intravascular ultrasound tissue characterization. J Am Coll Cardiol. 2010;55:1590–1597. doi: 10.1016/j. jacc.2009.07.078. 13. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circula- tion. 2002;105:1135–1143. 14. Cristell N, Cianflone D, Durante A, Ammirati E, Vanuzzo D, Banfi M, Calori G, Latib A, Crea F, Marenzi G, De Metrio M, Moretti L, Li H, Uren NG, Hu 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 myocardial infarction in 41% of cases: a multiethnic case-control study. J Am Coll Cardiol. 2011;58:2654–2661. doi: 10.1016/j.jacc.2011.08.055. 15. Scalone G, Niccoli G, Refaat H, Vergallo R, Porto I, Leone AM, Burzotta F, D’Amario D, Liuzzo G, Fracassi F, Trani C, Crea F. Not all plaque ruptures are born equal: an optical coherence tomography study [published online ahead of print December 24, 2016]. Eur Heart J Cardiovasc Imaging. doi: 10.1093/ehjci/jew208. 16. Arbustini E, Dal Bello B, Morbini P, Burke AP, Bocciarelli M, Specchia G, Virmani R. Plaque erosion is a major substrate for coronary thrombosis in acute myocardial infarction. Heart. 1999;82:269–272. 17. Bentzon JF, Otsuka F, Virmani R, Falk E. Mechanisms of plaque forma- tion and rupture. Circ Res. 2014;114:1852–1866. doi: 10.1161/CIRCRE- SAHA.114.302721. 18. Niccoli G, Montone RA, Cataneo L, Cosentino N, Gramegna M, Refaat H, Porto I, Burzotta F, Trani C, Leone AM, Severino A, Crea F. Morpho- logical-biohumoral correlations in acute coronary syndromes: patho- genetic implications. Int J Cardiol. 2014;171:463–466. doi: 10.1016/j. ijcard.2013.12.238. 19. Jia H, Abtahian F, Aguirre AD, Lee S, Chia S, Lowe H, Kato K, Yo- 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 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 patients with acute coronary syndrome by intravascular optical coherence tomography. J Am Coll Cardiol. 2013;62:1748–1758. doi: 10.1016/j. jacc.2013.05.071. 20. Crea F, Liuzzo G. Pathogenesis of acute coronary syndromes. J Am Coll Cardiol. 2013;61:1–11. doi: 10.1016/j.jacc.2012.07.064. 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 protein in severe unstable angina. N Engl J Med. 1994;331:417–424. doi: 10.1056/NEJM199408183310701. 22. Schönbeck U, Mach F, Sukhova GK, Murphy C, Bonnefoy JY, Fabunmi RP, Libby P. Regulation of matrix metalloproteinase expression in human vascular smooth muscle cells by T lymphocytes: a role for CD40 signaling in plaque rupture? Circ Res. 1997;81:448–454. 23. Liuzzo G, Kopecky SL, Frye RL, O’Fallon WM, Maseri A, Goronzy JJ, Wey- and CM. Perturbation of the T-cell repertoire in patients with unstable angina. Circulation. 1999;100:2135–2139. 24. Liuzzo G, Goronzy JJ, Yang H, Kopecky SL, Holmes DR, Frye RL, Weyand CM. Monoclonal T-cell proliferation and plaque instability in acute coro- nary syndromes. Circulation. 2000;101:2883–2888. 25. Chen S, Crother TR, Arditi M. Emerging role of IL-17 in atherosclerosis. J Innate Immun. 2010;2:325–333. doi: 10.1159/000314626. 26. Gisterå A, Robertson AK, Andersson J, Ketelhuth DF, Ovchinnikova O, Nilsson SK, Lundberg AM, Li MO, Flavell RA, Hansson GK. Transforming growth factor-β signaling in T cells promotes stabilization of atheroscle- rotic plaques through an interleukin-17-dependent pathway. Sci Transl Med. 2013;5:196ra100. doi: 10.1126/scitranslmed.3006133. 27. Brusko TM, Putnam AL, Bluestone JA. Human regulatory T cells: role in autoimmune disease and therapeutic opportunities. Immunol Rev. 2008;223:371–390. doi: 10.1111/j.1600-065X.2008.00637.x. 28. Liuzzo G, Montone RA, Gabriele M, Pedicino D, Giglio AF, Trotta F, Galiffa VA, Previtero M, Severino A, Biasucci LM, Crea F. Identification of unique adaptive immune system signature in acute coronary syndromes. Int J Car- diol. 2013;168:564–567. doi: 10.1016/j.ijcard.2013.01.009. 29. Flego D, Severino A, Trotta F, Previtero M, Ucci S, Zara C, Pedicino D, Mas- saro G, Biasucci LM, Liuzzo G, Crea F. Altered CD31 expression and activity in helper T cells of acute coronary syndrome patients. Basic Res Cardiol. 2014;109:448. doi: 10.1007/s00395-014-0448-3. 30. Flego D, Severino A, Trotta F, Previtero M, Ucci S, Zara C, Massaro G, Pedicino D, Biasucci LM, Liuzzo G, Crea F. Increased PTPN22 expression and defective CREB activation impair regulatory T-cell differentiation in non-ST-segment elevation acute coronary syndromes. J Am Coll Cardiol. 2015;65:1175–1186. doi: 10.1016/j.jacc.2015.01.027. 31. Nidorf SM, Eikelboom JW, Budgeon CA, Thompson PL. Low-dose colchi- cine for secondary prevention of cardiovascular disease. J Am Coll Cardiol. 2013;61:404–410. doi: 10.1016/j.jacc.2012.10.027. 32. Tardif J-C, L’Allier P. Colchicine Cardiovascular Outcomes Trial (COL- COT). https://clinicaltrials.gov/ct2/show/NCT02551094. 2017. Accessed June 1, 2017. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 11. ACS Mechanisms Inform Management STATEOFTHEART Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.029870 September 19, 2017 1165 33. Everett BM, Pradhan AD, Solomon DH, Paynter N, Macfadyen J, Zaharris E, Gupta M, Clearfield M, Libby P, Hasan AA, Glynn RJ, Ridker PM. Ra- tionale and design of the Cardiovascular Inflammation Reduction Trial: a test of the inflammatory hypothesis of atherothrombosis. Am Heart J. 2013;166:199–207.e15. doi: 10.1016/j.ahj.2013.03.018. 34. Morton AC, Rothman AM, Greenwood JP, Gunn J, Chase A, Clarke B, Hall AS, Fox K, Foley C, Banya W, Wang D, Flather MD, Crossman DC. The effect of interleukin-1 receptor antagonist therapy on markers of inflam- mation in non-ST elevation acute coronary syndromes: the MRC-ILA Heart Study. Eur Heart J. 2015;36:377–384. doi: 10.1093/eurheartj/ehu272. 35. Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1β inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am Heart J. 2011;162:597–605. doi: 10.1016/j.ahj.2011.06.012. 36. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH, Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z, Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi PRF, Troquay RPT, Libby P, Glynn RJ and Group CT. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease [published on- line ahead of print August 27, 2017]. N Engl J Med. 2017. doi: 10.1056/ NEJMoa1707914. 37. Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P, Glynn RJ, Ridker P, Lorenzatti A, Krum H, Varigos J, Siostrzonek P, Sinnaeve P, Fonseca F, Nico- lau J, Gotcheva N, Genest J, Yong H, Urina-Triana M, Milicic D, Cifkova R, Vettus R, Koenig W, Anker SD, Manolis AJ, Wyss F, Forster T, Sigurdsson A, Pais P, Fucili A, Ogawa H, Shimokawa H, Veze I, Petrauskiene B, Salvador L, Kastelein J, Cornel JH, Klemsdal TO, Medina F, Budaj A, Vida-Simiti L, Koba- lava Z, Otasevic P, Pella D, Lainscak M, Seung K-B, Commerford P, Dellborg M, Donath M, Hwang J-J, Kultursay H, Flather M, Ballantyne C, Bilazarian S, Chang W, East C, Everett B, Forgosh L, Glynn R, Harris B, Libby P, Ligueros M, Thuren T, Bohula E, Charmarthi B, Cheng S, Chou S, Danik J, McMahon G, Maron B, Ning M, Olenchock B, Pande R, Perlstein T, Pradhan A, Rost N, Singhal A, Taqueti V, Wei N, Burris H, Cioffi A, Dalseg AM, Ghosh N, Gralow J, Mayer T, Rugo H, Fowler V, Limaye AP, Cosgrove S, Levine D, Lopes R, Scott J, Thuren T, Ligueros M, Hilkert R, Tamesby G, Mickel C, Manning B, Woelcke J, Tan M, Manfreda S, Ponce T, Kam J, Saini R, Banker K, Salko T, Nandy P, Tawfik R, O'Neil G, Manne S, Jirvankar P, Lal S, Nema D, Jose J, Collins R, Bailey K, Blumenthal R, Colhoun H, Gersh B and Glynn RJ. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, dou- ble-blind, placebo-controlled trial [published online ahead of print August 25, 2017]. The Lancet. doi: 10.1016/S0140-6736(17)32247-X. 38. Fornasa G, Clement M, Groyer E, Gaston AT, Khallou-Laschet J, Morvan M, Guedj K, Kaveri SV, Tedgui A, Michel JB, Nicoletti A, Caligiuri G. A CD31-derived peptide prevents angiotensin II-induced atherosclerosis pro- gression and aneurysm formation. Cardiovasc Res. 2012;94:30–37. doi: 10.1093/cvr/cvs076. 39. Stanford SM, Krishnamurthy D, Falk MD, Messina R, Debnath B, Li S, Liu T, Kazemi R, Dahl R, He Y, Yu X, Chan AC, Zhang ZY, Barrios AM, Woods VL Jr, Neamati N, Bottini N. Discovery of a novel series of inhibitors of lym- phoid tyrosine phosphatase with activity in human T cells. J Med Chem. 2011;54:1640–1654. doi: 10.1021/jm101202j. 40. Putnam AL, Brusko TM, Lee MR, Liu W, Szot GL, Ghosh T, Atkinson MA, Bluestone JA. Expansion of human regulatory T-cells from patients with type 1 diabetes. Diabetes. 2009;58:652–662. doi: 10.2337/db08-1168. 41. Shah PK, Chyu KY, Dimayuga PC, Nilsson J. Vaccine for atherosclerosis. J Am Coll Cardiol. 2014;64:2779–2791. doi: 10.1016/j.jacc.2014.10.018. 42. Koskinas KC, Sukhova GK, Baker AB, Papafaklis MI, Chatzizisis YS, Coskun AU, Quillard T, Jonas M, Maynard C, Antoniadis AP, Shi GP, Libby P, Edel- man ER, Feldman CL, Stone PH. Thin-capped atheromata with reduced collagen content in pigs develop in coronary arterial regions exposed to persistently low endothelial shear stress. Arterioscler Thromb Vasc Biol. 2013;33:1494–1504. doi: 10.1161/ATVBAHA.112.300827. 43. Chatzizisis YS, Baker AB, Sukhova GK, Koskinas KC, Papafaklis MI, Beigel R, Jonas M, Coskun AU, Stone BV, Maynard C, Shi GP, Libby P, Feldman CL, Edelman ER, Stone PH. Augmented expression and activity of extracellular matrix-degrading enzymes in regions of low endothelial shear stress colo- calize with coronary atheromata with thin fibrous caps in pigs. Circulation. 2011;123:621–630. doi: 10.1161/CIRCULATIONAHA.110.970038. 44. Shiomi M, Ishida T, Kobayashi T, Nitta N, Sonoda A, Yamada S, Koike T, Kuniyoshi N, Murata K, Hirata K, Ito T, Libby P. Vasospasm of athero- sclerotic coronary arteries precipitates acute ischemic myocardial damage in myocardial infarction-prone strain of the Watanabe heritable hyperlip- idemic rabbits. Arterioscler Thromb Vasc Biol. 2013;33:2518–2523. doi: 10.1161/ATVBAHA.113.301303. 45. Libby P, Nahrendorf M, Swirski FK. Leukocytes link local and systemic in- flammation in ischemic cardiovascular disease: an expanded “cardiovas- cular continuum”. J Am Coll Cardiol. 2016;67:1091–1103. doi: 10.1016/j. jacc.2015.12.048. 46. Burke AP, Farb A, Malcom GT, Liang Y, Smialek JE, Virmani R. Plaque rup- ture and sudden death related to exertion in men with coronary artery disease. JAMA. 1999;281:921–926. 47. Janoudi A, Shamoun FE, Kalavakunta JK, Abela GS. Cholesterol crystal in- duced arterial inflammation and destabilization of atherosclerotic plaque. Eur Heart J. 2016;37:1959–1967. doi: 10.1093/eurheartj/ehv653. 48. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nuñez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA, Rock KL, Moore KJ, Wright SD, Hornung V, Latz E. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Na- ture. 2010;464:1357–1361. doi: 10.1038/nature08938. 49. Nakamura S, Inami S, Murai K, Takano M, Takano H, Asai K, Yasutake M, Shimizu W, Mizuno K. Relationship between cholesterol crystals and culprit lesion characteristics in patients with stable coronary artery disease: an optical coherence tomography study. Clin Res Cardiol. 2014;103:1015– 1021. doi: 10.1007/s00392-014-0748-5. 50. Liu L, Gardecki JA, Nadkarni SK, Toussaint JD, Yagi Y, Bouma BE, Tearney GJ. Imaging the subcellular structure of human coronary atherosclerosis using micro-optical coherence tomography. Nat Med. 2011;17:1010– 1014. doi: 10.1038/nm.2409. 51. Loree HM, Tobias BJ, Gibson LJ, Kamm RD, Small DM, Lee RT. Mechanical properties of model atherosclerotic lesion lipid pools. Arterioscler Thromb. 1994;14:230–234. 52. Huang H, Virmani R, Younis H, Burke AP, Kamm RD, Lee RT. The impact of calcification on the biomechanical stability of atherosclerotic plaques. Circulation. 2001;103:1051–1056. 53. Virmani R, Joner M, Sakakura K. Recent highlights of ATVB: calcification. Arterioscler Thromb Vasc Biol. 2014;34:1329–1332. doi: 10.1161/ATVBA- HA.114.304000. 54. Ruiz JL, Weinbaum S, Aikawa E, Hutcheson JD. Zooming in on the genesis of atherosclerotic plaque microcalcifications. J Physiol. 2016;594:2915– 2927. doi: 10.1113/JP271339. 55. Camici PG, Rimoldi OE, Gaemperli O, Libby P. Non-invasive anatomic and functional imaging of vascular inflammation and unstable plaque. Eur Heart J. 2012;33:1309–1317. doi: 10.1093/eurheartj/ehs067. 56. Libby P. How does lipid lowering prevent coronary events? New insights from human imaging trials. Eur Heart J. 2015;36:472–474. doi: 10.1093/ eurheartj/ehu510. 57. Abela GS, Vedre A, Janoudi A, Huang R, Durga S, Tamhane U. Effect of statins on cholesterol crystallization and atherosclerotic plaque sta- bilization. Am J Cardiol. 2011;107:1710–1717. doi: 10.1016/j.amj- card.2011.02.336. 58. Patel R, Janoudi A, Vedre A, Aziz K, Tamhane U, Rubinstein J, Abela OG, Berger K, Abela GS. Plaque rupture and thrombosis are reduced by lowering cholesterol levels and crystallization with ezetimibe and are correlated with fluorodeoxyglucose positron emission tomography. Ar- terioscler Thromb Vasc Biol. 2011;31:2007–2014. doi: 10.1161/ATVBA- HA.111.226167. 59. Zimmer S, Grebe A, Bakke SS, Bode N, Halvorsen B, Ulas T, Skjelland M, De Nardo D, Labzin LI, Kerksiek A, Hempel C, Heneka MT, Hawxhurst V, Fitzgerald ML, Trebicka J, Björkhem I, Gustafsson JÅ, Westerterp M, Tall AR, Wright SD, Espevik T, Schultze JL, Nickenig G, Lütjohann D, Latz E. Cyclodextrin promotes atherosclerosis regression via macrophage repro- gramming. Sci Transl Med. 2016;8:333ra50. doi: 10.1126/scitranslmed. aad6100. 60. Mohammadpour AH, Akhlaghi F. Future of cholesteryl ester transfer pro- tein (CETP) inhibitors: a pharmacological perspective. Clin Pharmacokinet. 2013;52:615–626. doi: 10.1007/s40262-013-0071-8. 61. HPS/TIMI55-REVEAL Collaborative Group. Effects of Anacetrapib in Pa- tients with Atherosclerotic Vascular Disease [published online ahead of print August 28, 2017]. N Engl J Med. doi: 10.1056/NEJMoa1706444. 62. Meuwese MC, de Groot E, Duivenvoorden R, Trip MD, Ose L, Maritz FJ, Ba- sart DC, Kastelein JJ, Habib R, Davidson MH, Zwinderman AH, Schwocho LR, Stein EA; CAPTIVATE Investigators. ACAT inhibition and progression of carotid atherosclerosis in patients with familial hypercholesterolemia: the CAPTIVATE randomized trial. JAMA. 2009;301:1131–1139. doi: 10.1001/ jama.301.11.1131. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 12. Crea and Libby September 19, 2017 Circulation. 2017;136:1155–1166. DOI: 10.1161/CIRCULATIONAHA.117.0298701166 63. Lüscher TF. Substrates of acute coronary syndromes: new insights into plaque rupture and erosion. Eur Heart J. 2015;36:1347–1349. doi: 10.1093/eurheartj/ehv149. 64. Ferrante G, Nakano M, Prati F, Niccoli G, Mallus MT, Ramazzotti V, Mon- tone RA, Kolodgie FD, Virmani R, Crea F. High levels of systemic my- eloperoxidase are associated with coronary plaque erosion in patients with acute coronary syndromes: a clinicopathological study. Circulation. 2010;122:2505–2513. doi: 10.1161/CIRCULATIONAHA.110.955302. 65. Yahagi K, Kolodgie FD, Lutter C, Mori H, Romero ME, Finn AV, Virmani R. Pathology of human coronary and carotid artery atherosclerosis and vascular calcification in diabetes mellitus. Arterioscler Thromb Vasc Biol. 2017;37:191–204. doi: 10.1161/ATVBAHA.116.306256. 66. Hansson GK, Libby P, Tabas I. Inflammation and plaque vulnerability. J In- tern Med. 2015;278:483–493. doi: 10.1111/joim.12406. 67. Quillard T, Araújo HA, Franck G, Shvartz E, Sukhova G, Libby P. TLR2 and neutrophils potentiate endothelial stress, apoptosis and detachment: im- plications for superficial erosion. Eur Heart J. 2015;36:1394–1404. doi: 10.1093/eurheartj/ehv044. 68. Franck G, Mawson T, Sausen G, Salinas M, Masson GS, Cole A, Beltrami- Moreira M, Chatzizisis Y, Quillard T, Tesmenitsky Y, Shvartz E, Sukhova GK, Swirski FK, Nahrendorf M, Aikawa E, Croce KJ, Libby P. Flow perturbation mediates neutrophil recruitment and potentiates endothelial injury via TLR2 in mice: implications for superficial erosion. Circ Res. 2017;121:31– 42. doi: 10.1161/CIRCRESAHA.117.310694. 69. Kolodgie FD, Burke AP, Wight TN, Virmani R. The accumulation of specific types of proteoglycans in eroded plaques: a role in coronary thrombosis in the absence of rupture. Curr Opin Lipidol. 2004;15:575–582. 70. Day AJ. The structure and regulation of hyaluronan-binding proteins. Bio- chem Soc Trans. 1999;27:115–121. 71. Butler LM, Rainger GE, Nash GB. A role for the endothelial glycosami- noglycan hyaluronan in neutrophil recruitment by endothelial cells cul- tured for prolonged periods. Exp Cell Res. 2009;315:3433–3441. doi: 10.1016/j.yexcr.2009.08.012. 72. Döring Y, Soehnlein O, Weber C. Neutrophil extracellular traps in ath- erosclerosis and atherothrombosis. Circ Res. 2017;120:736–743. doi: 10.1161/CIRCRESAHA.116.309692. 73. Libby P. Triglycerides on the rise: should we swap seats on the seesaw? Eur Heart J. 2015;36:774–776. doi: 10.1093/eurheartj/ehu500. 74. Mangold A, Alias S, Scherz T, Hofbauer T, Jakowitsch J, Panzenböck A, Simon D, Laimer D, Bangert C, Kammerlander A, Mascherbauer J, Winter MP, Distelmaier K, Adlbrecht C, Preissner KT, Lang IM. Coronary neutro- phil extracellular trap burden and deoxyribonuclease activity in ST-eleva- tion acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ Res. 2015;116:1182–1192. doi: 10.1161/CIRCRE- SAHA.116.304944. 75. Aguirre-Alvarado C, Segura-Cabrera A, Velázquez-Quesada I, Hernández- Esquivel MA, García-Pérez CA, Guerrero-Rodríguez SL, Ruiz-Moreno AJ, Rodríguez-Moreno A, Pérez-Tapia SM, Velasco-Velázquez MA. Virtual screening-driven repositioning of etoposide as CD44 antagonist in breast cancer cells. Oncotarget. 2016;7:23772–23784. doi: 10.18632/ oncotarget.8180. 76. Jia H, Dai J, Hou J, Xing L, Ma L, Liu H, Xu M, Yao Y, Hu S, Yamamoto E, Lee H, Zhang S, Yu B, Jang IK. Effective anti-thrombotic therapy without stenting: intravascular optical coherence tomography-based management in plaque erosion (the EROSION study). Eur Heart J. 2017;38:792–800. doi: 10.1093/eurheartj/ehw381. 77. Libby P. Superficial erosion and the precision management of acute coro- nary syndromes: not one-size-fits-all. Eur Heart J. 2017;38:801–803. doi: 10.1093/eurheartj/ehw599. 78. Beltrame JF, Crea F, Kaski JC, Ogawa H, Ong P, Sechtem U, Shimokawa H, Bairey Merz CN; Coronary Vasomotion Disorders International Study Group (COVADIS). The who, what, why, when, how and where of vaso- spastic angina. Circ J. 2016;80:289–298. doi: 10.1253/circj.CJ-15-1202. 79. Ong P, Athanasiadis A, Borgulya G, Voehringer M, Sechtem U. 3-Year fol- low-up of patients with coronary artery spasm as cause of acute coronary syndrome: the CASPAR (Coronary Artery Spasm in Patients With Acute Coronary Syndrome) study follow-up. J Am Coll Cardiol. 2011;57:147– 152. doi: 10.1016/j.jacc.2010.08.626. 80. Sato K, Kaikita K, Nakayama N, Horio E, Yoshimura H, Ono T, Ohba K, Tsujita K, Kojima S, Tayama S, Hokimoto S, Matsui K, Sugiyama S, Yamabe H, Ogawa H. Coronary vasomotor response to intracoronary acetylcholine injection, clinical features, and long-term prognosis in 873 consecutive pa- tients with coronary spasm: analysis of a single-center study over 20 years. J Am Heart Assoc. 2013;2:e000227. doi: 10.1161/JAHA.113.000227. 81. Bertrand ME, LaBlanche JM, Tilmant PY, Thieuleux FA, Delforge MR, Carre AG, Asseman P, Berzin B, Libersa C, Laurent JM. Frequency of provoked coronary arterial spasm in 1089 consecutive patients undergoing coronary arteriography. Circulation. 1982;65:1299–1306. 82. Pristipino C, Beltrame JF, Finocchiaro ML, Hattori R, Fujita M, Mongiardo R, Cianflone D, Sanna T, Sasayama S, Maseri A. Major racial differences in coronary constrictor response between Japanese and Caucasians with recent myocardial infarction. Circulation. 2000;101:1102–1108. 83. Camici PG, Crea F. Coronary microvascular dysfunction. N Engl J Med. 2007;356:830–840. doi: 10.1056/NEJMra061889. 84. Galiuto L, De Caterina AR, Porfidia A, Paraggio L, Barchetta S, Locorotondo G, Rebuzzi AG, Crea F. Reversible coronary microvascular dysfunction: a common pathogenetic mechanism in apical ballooning or tako-tsubo syn- drome. Eur Heart J. 2010;31:1319–1327. doi: 10.1093/eurheartj/ehq039. 85. Templin C, Ghadri JR, Diekmann J, Napp LC, Bataiosu DR, Jaguszewski M, Cammann VL, Sarcon A, Geyer V, Neumann CA, Seifert B, Hellermann J, Schwyzer M, Eisenhardt K, Jenewein J, Franke J, Katus HA, Burgdorf C, Schunkert H, Moeller C, Thiele H, Bauersachs J, Tschöpe C, Schultheiss HP, Laney CA, Rajan L, Michels G, Pfister R, Ukena C, Böhm M, Erbel R, Cuneo A, Kuck KH, Jacobshagen C, Hasenfuss G, Karakas M, Koenig W, Rott- bauer W, Said SM, Braun-Dullaeus RC, Cuculi F, Banning A, Fischer TA, Va- sankari T, Airaksinen KE, Fijalkowski M, Rynkiewicz A, Pawlak M, Opolski 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, Ford I, Ruschitzka F, Prasad A, Lüscher TF. Clinical features and outcomes of takotsubo (stress) cardiomyopathy. N Engl J Med. 2015;373:929–938. doi: 10.1056/NEJMoa1406761. 86. Arrebola-Moreno AL, Arrebola JP, Moral-Ruiz A, Ramirez-Hernandez JA, Melgares-Moreno R, Kaski JC. Coronary microvascular spasm triggers transient ischemic left ventricular diastolic abnormalities in patients with chest pain and angiographically normal coronary arteries. Atherosclerosis. 2014;236:207–214. doi: 10.1016/j.atherosclerosis.2014.07.009. 87. Kaski JC, Maseri A, Vejar M, Crea F, Hackett D. Spontaneous coronary artery spasm in variant angina is caused by a local hyperreactivity to a generalized constrictor stimulus. J Am Coll Cardiol. 1989;14:1456–1463. 88. Lanza GA, Pedrotti P, Pasceri V, Lucente M, Crea F, Maseri A. Autonomic changes associated with spontaneous coronary spasm in patients with variant angina. J Am Coll Cardiol. 1996;28:1249–1256. doi: 10.1016/ S0735-1097(96)00309-9. 89. Shimokawa H, Tomoike H, Nabeyama S, Yamamoto H, Araki H, Nakamura M, Ishii Y, Tanaka K. Coronary artery spasm induced in atherosclerotic miniature swine. Science. 1983;221:560–562. 90. Ito A, Shimokawa H, Nakaike R, Fukai T, Sakata M, Takayanagi T, Egashira K, Takeshita A. Role of protein kinase C-mediated pathway in the pathogenesis of coronary artery spasm in a swine model. Circulation. 1994;90:2425–2431. 91. Shimokawa H, Seto M, Katsumata N, Amano M, Kozai T, Yamawaki T, Ku- wata K, Kandabashi T, Egashira K, Ikegaki I, Asano T, Kaibuchi K, Takeshita A. Rho-kinase-mediated pathway induces enhanced myosin light chain phosphorylations in a swine model of coronary artery spasm. Cardiovasc Res. 1999;43:1029–1039. 92. Shimokawa H. 2014 Williams Harvey Lecture: importance of coro- nary vasomotion abnormalities: from bench to bedside. Eur Heart J. 2014;35:3180–3193. doi: 10.1093/eurheartj/ehu427. 93. Dai Z, Aoki T, Fukumoto Y, Shimokawa H. Coronary perivascular fibrosis is associated with impairment of coronary blood flow in patients with non-ischemic heart failure. J Cardiol. 2012;60:416–421. doi: 10.1016/j. jjcc.2012.06.009. 94. Kaski JC, Crea F, Meran D, Rodriguez L, Araujo L, Chierchia S, Davies G, Maseri A. Local coronary supersensitivity to diverse vasoconstrictive stimuli in patients with variant angina. Circulation. 1986;74:1255–1265. 95. Lanza GA, Careri G, Crea F. Mechanisms of coronary artery spasm. Circulation. 2011;124:1774–1782. doi: 10.1161/CIRCULATIONAHA.111.037283. 96. Abbate A, Hamza M, Cassano AD, Melchior R, Roberts C, Grizzard J, Shah K, Hastillo A, Kasirajan V, Crea F, Lanza GA, Vetrovec GW. Sympathec- tomy as a treatment for refractory coronary artery spasm. Int J Cardiol. 2012;161:e7–e9. doi: 10.1016/j.ijcard.2012.03.006. 97. Masumoto A, Mohri M, Shimokawa H, Urakami L, Usui M, Takeshita A. Suppression of coronary artery spasm by the Rho-kinase inhibitor fasudil in patients with vasospastic angina. Circulation. 2002;105:1545–1547. 98. King K, Grazette LP, Paltoo DN, McDevitt JT, Sia SK, Barrett PM, Apple FS, Gurbel PA, Weissleder R, Leeds H, Iturriaga EJ, Rao A, Adhikari B, Desvigne-Nickens P, Galis ZS, Libby P. Point-of-care technologies for preci- sion cardiovascular care and clinical research: National Heart, Lung, and Blood Institute Working Group. JACC Basic Transl Sci. 2016;1:73–86. doi: 10.1016/j.jacbts.2016.01.008. byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom
  • 13. Filippo Crea and Peter Libby Treatment Acute Coronary Syndromes: The Way Forward From Mechanisms to Precision Print ISSN: 0009-7322. Online ISSN: 1524-4539 Copyright © 2017 American Heart Association, Inc. All rights reserved. is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulation doi: 10.1161/CIRCULATIONAHA.117.029870 2017;136:1155-1166Circulation. http://circ.ahajournals.org/content/136/12/1155 World Wide Web at: The online version of this article, along with updated information and services, is located on the http://circ.ahajournals.org//subscriptions/ is online at:CirculationInformation about subscribing toSubscriptions: http://www.lww.com/reprints Information about reprints can be found online at:Reprints: document.Permissions and Rights Question and Answerthis process is available in the click Request Permissions in the middle column of the Web page under Services. Further information about Office. Once the online version of the published article for which permission is being requested is located, can be obtained via RightsLink, a service of the Copyright Clearance Center, not the EditorialCirculationin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions: byguestonSeptember18,2017http://circ.ahajournals.org/Downloadedfrom