Sei sulla pagina 1di 15

The n e w e ng l a n d j o u r na l of m e dic i n e

review article

Mechanisms of Disease

Myocardial Reperfusion Injury


Derek M. Yellon, D.Sc., and Derek J. Hausenloy, Ph.D.

C
oronary heart disease is the leading cause of death world- From the Hatter Cardiovascular Institute,
wide, and 3.8 million men and 3.4 million women die of the disease each University College London Hospital and
Medical School, London. Address reprint
year. After an acute myocardial infarction, early and successful myocardial requests to Dr. Yellon at the Hatter Car-
reperfusion with the use of thrombolytic therapy or primary percutaneous coronary diovascular Institute, University College
intervention (PCI) is the most effective strategy for reducing the size of a myocar­ London Hospital and Medical School, 67
Chenies Mews, London WC1E 6HX, Unit-
dial infarct and improving the clinical outcome. The process of restoring blood ed Kingdom, or at hatter-institute@ucl.
flow to the ischemic myocardium, however, can induce injury. This phenomenon, ac.uk.
termed myocardial reperfusion injury, can paradoxically reduce the beneficial ef­
N Engl J Med 2007;357:1121-35.
fects of myocardial reperfusion. Copyright © 2007 Massachusetts Medical Society.
The potentially detrimental aspect of myocardial reperfusion injury, termed lethal
reperfusion injury, is defined as myocardial injury caused by the restoration of coro­
nary blood flow after an ischemic episode. The injury culminates in the death of
cardiac myocytes that were viable immediately before myocardial reperfusion.1 This
form of myocardial injury, which by itself can induce cardiomyocyte death and in­
crease infarct size (Fig. 1), may in part explain why, despite optimal myocardial re­
perfusion, the rate of death after an acute myocardial infarction approaches 10%,2 and
the incidence of cardiac failure after an acute myocardial infarction is almost 25%.
Studies in animal models of acute myocardial infarction suggest that lethal re­
perfusion injury accounts for up to 50% of the final size of a myocardial infarct, and
in these models a number of strategies have been shown to ameliorate lethal reper­
fusion injury. Yet, the translation of these beneficial effects into the clinical setting
has been disappointing.3 Nevertheless, recent demonstrations of the benefit of ische­
mic postconditioning,4 in which myocardial reperfusion in patients with acute myo­
cardial infarction who are undergoing PCI is interrupted with short-lived episodes of
myocardial ischemia,5-7 have regenerated interest in the reperfusion phase as a target
for cardioprotection. The identification of the reperfusion injury salvage kinase (RISK)
pathway 8 and the mitochondrial permeability transition pore (PTP)9,10 as new tar­
gets for cardioprotection has also intensified research in this area. These new de­
velopments should lead to strategies that improve clinical outcomes in acute myo­
cardial infarction and reduce the risk of heart failure after myocardial infarction.11

M yo c a r di a l R eper f usion Inj ur y a nd Cel l De ath

Myocardial reperfusion injury was first postulated in 1960 by Jennings et al.12 in


their description of the histologic features of reperfused ischemic canine myocar­
dium. They reported cell swelling, contracture of myofibrils, disruption of the sarco­
lemma, and the appearance of intramitochondrial calcium phosphate particles. The
injury to the heart during myocardial reperfusion causes four types of cardiac dys­
function. The first type is myocardial stunning, a term denoting the “mechanical
dysfunction that persists after reperfusion despite the absence of irreversible dam­
age and despite restoration of normal or near-normal coronary flow.”13 The myocar­

n engl j med 357;11  www.nejm.org  september 13, 2007 1121

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

was sustained during the ischemic period.18 The


Myocardial ischemia in absence uncertainty relates to the inability to accurately as­
70 of reperfusion
Infarct size, 70%
sess in situ the progress of necrosis during the
transition from myocardial ischemia to reperfu­
60
sion.1 As a result, the most convincing means of
showing the existence of lethal reperfusion injury
50
as a distinct mediator of cardiomyocyte death is
to show that the size of a myocardial infarct can
Infarct Size (%)

40
be reduced by an intervention used at the begin­
Myocardial ischemia with reperfusion ning of myocardial reperfusion.1,19
30 Reperfusion reduces infarct size by 40%
Part of the remaining 30% infarct is due

20
to lethal reperfusion injury and P o ten t i a l Medi at or s of L e th a l
is therefore preventable
R eper f usion Inj ur y
10 Oxygen Paradox
Myocardial ischemia with reperfusion
5 and cardioprotection Experimental studies have established that the re­
0 Preventing lethal reperfusion injury reduces perfusion of ischemic myocardium generates oxi­
infarct size by a further 25%, realizing the
full benefits of reperfusion dative stress, which itself can mediate myocardial
injury20 (Fig. 2). Oxidative stress is part of the oxy­
Figure 1. Contribution of Lethal Reperfusion Injury to Final Myocardial gen paradox,21 in which the reoxygenation of isch­
Infarct Size. ICM AUTHOR: Yellon RETAKE 1st emic myocardium generates a degree of myocar­
2nd
This hypothetical
FIGURE: 1 of 3
REG Fscheme shows the large reduction in myocardial 3rd infarct
dial injury that greatly exceeds the injury induced
size obtained CASE
by early and successful myocardial reperfusion Revised after a sus- by ischemia alone.21 The role of oxidative stress
Line 4-C
tained episodeEMail
of acute myocardial
ARTIST: ts ischemia. The full benefits
SIZE of myocardi- in lethal reperfusion injury is clouded by the incon­
are not realized becauseH/T
al reperfusionEnon of the H/T
presence of 22p3
lethal reperfusion
Combo clusive results of animal and clinical studies22-52
injury, which diminishes the magnitude of the reduction in infarct size elic-
ited by myocardialFigure
AUTHOR, PLEASE NOTE:
reperfusion. of cardioprotection by antioxidant reperfusion
has been This concept
redrawn is has
and type revealed by the further re-
been reset.
duction in myocardial infarct Please
size obtained by preventing lethal reperfusion
check carefully. therapy (Table 1).
injury with the administration of a cardioprotective intervention at the be- Oxidative stress during myocardial reperfusion
ginning of myocardial
JOB: 35711 reperfusion. Infarcted myocardium is depicted in
ISSUE: 09-13-07 also reduces the bioavailability of the intracellular
pink, and the viable, at-risk myocardium is stained red. Infarct size is ex-
signaling molecule, nitric oxide, thereby removing
pressed as a percentage of the volume of myocardium at risk for infarction.
its cardioprotective effects. These effects include
the inhibition of neutrophil accumulation, inacti­
dium usually recovers from this reversible form vation of superoxide radicals, and improvement of
of injury after several days or weeks. The second coronary blood flow.53 Nitric oxide reperfusion
type of cardiac dysfunction, the no-reflow phenom­ therapy to increase nitric oxide levels can reduce
enon, was originally defined as the “inability to the size of a myocardial infarct in animals,54 but
reperfuse a previously ischemic region.”14 It refers clinical studies of the antianginal nitric oxide do­
to the impedance of microvascular blood flow en­ nor nicorandil have reported benefit only in terms
countered during opening of the infarct-related of improved myocardial reperfusion; results in
coronary artery.15 The third type of cardiac dys­ terms of clinical outcomes after an acute myocar­
function, reperfusion arrhythmias, is potentially dial infarction are mixed (Table 1).47-49
harmful, but effective treatments are available.16
The last type is lethal reperfusion injury. There Calcium Paradox
are several comprehensive reviews of myocardial At the time of myocardial reperfusion, there is an
stunning,17 the no-reflow phenomenon,15 and re­ abrupt increase in intracellular Ca2+, which is sec­
perfusion arrhythmias.16 ondary to sarcolemmal-membrane damage and
The concept of lethal reperfusion injury as an oxidative stress–induced dysfunction of the sarco­
independent mediator of cardiomyocyte death, plasmic reticulum. These two forms of injury over­
distinct from ischemic injury, has been hotly de­ whelm the normal mechanisms that regulate Ca2+
bated; some researchers have suggested that re­ in the cardiomyocyte; this phenomenon is termed
perfusion only exacerbates the cellular injury that the calcium paradox1 (Fig. 2). The result is intra­

1122 n engl j med 357;11  www.nejm.org  september 13, 2007

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

cellular and mitochondrial Ca2+ overload, and this Experimental studies have shown reductions in
excess of Ca2+ induces cardiomyocyte death by infarct size of up to 50% with several interventions
causing hypercontracture of the heart cells and aimed at neutrophils during myocardial reper­
mitochondrial PTP opening1 (Fig. 2). Attenuating fusion. These interventions include leukocyte-
intracellular Ca2+ overload with pharmacologic an­ depleted blood63; antibodies against the cell-adhe­
tagonists of the sarcolemmal Ca2+ ion channel, sion molecules P-selectin,64 CD11 and CD18,65 and
the mitochondrial Ca2+ uniporter, or the sodium– the intercellular adhesion molecule 166; and phar­
hydrogen exchanger decreases myocardial infarct macologic inhibitors of complement activation.67
size by up to 50% in experimental studies.55-57 However, the corresponding clinical studies have
However, the results of the corresponding clini­ not shown any meaningful cardioprotective effect
cal studies have been negative.27,29 That inhibition of such interventions (Table 1).32-38
of sodium–hydrogen exchange at the time of PCI After inconclusive experimental studies,68,69
does not protect the myocardium during an acute clinical studies of the antiinflammatory agent
myocardial infarction is consistent with the results aden­osine as an adjunct to PCI have shown an
of experimental studies in which the beneficial 11% reduction in the size of myocardial infarcts,
effects of inhibiting sodium–hydrogen exchange but benefits in terms of clinical outcomes were
were shown to occur during myocardial ischemia limited to patients presenting within 3 hours af­
and not reperfusion (Table 1).29,58 MCC-135, the ter the onset of symptoms (Table 1).39,40
first in a new class of agents that reduce intra­
cellular Ca2+ loading by inhibiting the sodium– Metabolic Modulation
hydrogen exchanger and promoting Ca2+ uptake Several experimental and numerous clinical stud­
by the sarcoplasmic reticulum, has also not influ­ ies have examined the cardioprotective potential of
enced infarct size when given during reperfusion therapy with glucose, insulin, and potassium ad­
(Table 1).30 ministered as an adjunct to myocardial reperfu­
sion.70,71 These studies have been conducted on the
pH Paradox premise that ischemic myocardium benefits more
The rapid restoration of physiologic pH during from metabolizing glucose than from fatty acids.72
myocardial reperfusion, which follows the wash­ A recent very large, randomized, controlled study
out of lactic acid and the activation of the sodium– from several centers reported no cardioprotective
hydrogen exchanger and the sodium–bicarbonate benefit from therapy with glucose, insulin, and po­
symporter, contributes to lethal reperfusion injury tassium as an adjunct to myocardial reperfusion
(Fig. 2). This phenomenon is termed the pH par­ in patients with acute myocardial infarction (Ta­
adox.59 In neonatal rat cardiomyocytes, experimen­ ble 1).41 The delay in initiating this therapy, the
tal studies have shown that reoxygenation with prolonged period of myocardial ischemia, and high
acidic buffer is cardioprotective60; this effect may and potentially damaging glucose levels have all
be mediated by the inhibition of mitochondrial PTP been cited as reasons for the lack of cardioprotec­
opening.61 However, in clinical studies, delaying tion.71 The effect of therapy with glucose, insulin,
the restoration of physiologic pH during myocar­ and potassium administered in the ambulance to
dial reperfusion using sodium–hydrogen exchange patients with acute myocardial infarction before
inhibition did not protect the heart (Table 1).29,58 myocardial reperfusion has occurred is being in­
vestigated in the Immediate Metabolic Myocardial
Inflammation Enhancement During Initial Assessment and Treat­
After an acute myocardial infarction, the release of ment in Emergency Care (IMMEDIATE) trial.42
chemoattractants draws neutrophils into the in­
farct zone during the first 6 hours of myocardial Magnesium Therapy
reperfusion, and during the next 24 hours they Experimental studies have reported that intrave­
migrate into the myocardial tissue. This process is nous magnesium administered during myocardial
facilitated by cell-adhesion molecules. These neu­ reperfusion can reduce myocardial infarct size, but
trophils cause vascular plugging and release deg­ the mechanism of this effect is unclear.73 Initial
radative enzymes and reactive oxygen species clinical studies of adjunctive reperfusion therapy
(Fig. 2).62 with magnesium in patients with acute myocardial

n engl j med 357;11  www.nejm.org  september 13, 2007 1123

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

infarction were inconclusive; the timing of its


Figure 2. Major Mediators of Lethal Reperfusion Injury.
administration was not sufficiently controlled.43,44
During myocardial reperfusion, the acute ischemic
However, subsequent trials with magnesium ad­ myocardium is subjected to several abrupt biochemical
ministered immediately before PCI have also not and metabolic changes, which compound the changes
shown cardioprotection (Table 1).45,46 generated during the period of myocardial ischemia.
These changes include mitochondrial reenergization
Therapeutic Hypothermia (purple), the generation of reactive oxygen species
(ROS) (orange), intracellular Ca2+ overload (green), the
Mild hypothermia (33 to 35°C) has been reported rapid restoration of physiologic pH (blue), and inflam-
to benefit patients surviving a cardiac arrest.74 Ex­ mation (red), all of which interact with each other to
perimental studies have shown a 10% reduction in mediate cardiomyocyte death through the opening of
myocardial infarct size for every 1°C decrease in the mitochondrial permeability transition pore (PTP)
and the induction of cardiomyocyte hypercontracture.
body temperature75; mild hypothermia reduces
During myocardial reperfusion, ROS are generated by
myocardial infarct size in human-sized pigs.76 xanthine oxidase (mainly from endothelial cells) and
However, initial clinical studies of therapeutic hy­ the re-energized electron transport chain in the cardio-
pothermia in patients with acute myocardial in­ myocyte mitochondria. Several hours later, a further
farction who are undergoing primary PCI have source of ROS is NADPH oxidase (mainly from neutro-
phils). ROS mediate myocardial injury by inducing
not shown any beneficial effects (Table 1).50-52
mitochondrial PTP opening, acting as neutrophil che-
moattractants, mediating dysfunction of the sarcoplas-
Mitochondrial PTP mic reticulum and contributing to intracellular Ca2+
The mitochondrial PTP is a nonselective channel overload, damaging the cell membrane by lipid peroxi-
of the inner mitochondrial membrane. Opening dation, inducing enzyme denaturation, and causing di-
rect oxidative damage to DNA. During myocardial re-
the channel collapses the mitochondrial membrane
perfusion, the already Ca2+ -overloaded cardiomyocyte
potential and uncouples oxidative phosphorylation, is subjected to a further influx of Ca2+ through a dam-
resulting in ATP depletion and cell death.9 Dur­ aged sarcolemmal membrane, ROS-mediated dysfunc-
ing myocardial ischemia, the mitochondrial PTP tion of the sarcoplasmic reticulum, and reverse func-
remains closed, only to open within the first few tion of the Na+ –Ca2+ exchanger. The generation of ATP
by the reenergized electron transport chain in the set-
minutes after myocardial reperfusion in response
ting of intracellular Ca2+ overload induces cardiomyo-
to mitochondrial Ca2+ overload, oxidative stress, cyte death by hypercontracture, a process that is facili-
restoration of a physiologic pH, and ATP deple­ tated by the rapid restoration of physiologic pH during
tion.61,77 Therefore, the mitochondrial PTP is a myocardial reperfusion. Furthermore, the restoration
critical determinant of lethal reperfusion injury, of the mitochondrial membrane potential drives the
entry of Ca2+ into mitochondria that, in conjunction
and as such it is an important new target for car­
with the loss of the inhibitory effect of the acidic pH on
dioprotection. the mitochondrial PTP and the generation of ROS, act
in concert to mediate the opening of the mitochondrial
PTP. This opening induces cardiomyocyte death by un-
Ta rge t ing L e th a l coupling oxidative phosphorylation and inducing mito-
R eper f usion Inj ur y chondrial swelling. During myocardial reperfusion, the
rapid washout of lactic acid together with the function
Experimental studies have shown that interven­ of the Na+ –H+ and Na+ –HCO3 transporters mediate
tions during myocardial reperfusion can reduce the rapid restoration of physiologic pH, facilitating mi-
myocardial infarct size by up to 50%, suggesting tochondrial PTP opening and cardiomyocyte hypercon-
that lethal reperfusion injury contributes to up to tracture. Several hours after the onset of myocardial re-
perfusion, neutrophils accumulate in the infarcted
half of the final myocardial infarct size. However,
myocardial tissue in response to the release of the che-
the disappointing attempts to translate the bene­ moattractants (ROS, cytokines, and the activated com-
ficial effects that were shown in animal models plement). The up-regulated cell-adhesion molecules
into clinical practice have raised the question of P-selectin, CD18 and CD11, and intracellular adhesion
whether such infarct models are relevant to myo­ molecule 1 (ICAM-1) then facilitate the migration of
neutrophils into the myocardial tissue, where they
cardial infarction in people.3 Several reasons have
mediate cardiomyocyte death by causing vascular
been proposed for the disparity between findings plugging, releasing degradative enzymes, and gener-
in animals and in patients (Table 2).3,79 In the clin­ ating ROS.
ical setting, the varying degrees of ischemia in an

1124 n engl j med 357;11  www.nejm.org  september 13, 2007

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

Blood vessel

Chemoattractants
Endothelial cell Reactive oxygen species Washout of
Cytokines lactic acid
Activated complement
Neutrophil

Vascular plugging
Degradative enzymes

NADPH
oxidase

Xanthine Cell-adhesion
oxidase molecules
P-selectin
CD18 and CD11 Na+
ICAM-1 H+

Membrane Ca2+ overload


peroxidation
Ca2+
Na+ Na+ HCO3
pH
Reactive correction
oxygen species
Sarcoplasmic
reticulum
Opening of the
mitochondrial
PTP
Mitochondria
reenergized
Cardiomyocyte

Cardiomyocyte
hypercontracture Myofibrils

Lethal reperfusion
injury

acute myocardial infarction can cause the loss of the interventions examined so far, many may have
COLOR FIGURE

innate cardioprotective adaptations such as ische­Draft


been
4 of questionable
08/23/07 benefit in preclinical studies
mic preconditioning and postconditioning within
Author or were given to patients at a dose and schedule that
Yellon
different regions of the ischemic myocardium. Fig # had
2 not been validated in studies in animals.
Title
This heterogeneity could contribute to the incon­
ME
As a way forward, and in agreement with the
clusive results of clinical studies. Furthermore,
DE of working group of the National Heart, Lung, and
Artist SBL
AUTHOR PLEASE NOTE:
Figure has been redrawn and type has been reset
Please check carefully

Issue date
n engl j med 357;11  www.nejm.org  september 13, 2007 1125

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Table 1. Previous Attempts to Reduce Lethal Reperfusion Injury in Patients with Acute Myocardial Infarction.*

1126
No. of Period of Timing of
Cardioprotective Strategy and Trial Patients Ischemia Intervention Details of Study Results
hr
Antioxidants
Flaherty et al.22 120 ≤4 (92% of Before PCI Intravenous bolus of superoxide dis- No difference in recovery of LVEF 4–6 wk after PCI
patients) mutase (10 mg/kg of body weight)
followed by a 60-min infusion
of 0.2 mg/kg/min
Downey23 (EMIP-FR) 19,725 ≤6 (83% of ≤15 min after throm- Intravenous infusion of trimetazidine No difference in 35-day mortality
patients) bolysis
Guan et al.25 38 4.5 Before PCI Oral allopurinol Improved LVEF and less oxidative stress
Tsujita et al.26 101 3.5 Before PCI Intravenous edaravone Reduced infarct size, less oxidative stress and reperfu-
sion arrhythmias, improved short-term clinical
outcomes
Reduction of intracellular
The

Ca2+ overload and Na+–H+


exchange inhibitors
Boden et al.27 874 ≤6 (85% of After thrombolysis Oral diltiazem 36–96 hr after onset No effect on death, nonfatal myocardial infarction, or
patients) of infarct symptoms recurrent ischemia but reduction in nonfatal cardi-
ac events, including myocardial revascularization
Théroux et al.28 3,439 3 Before PCI Na+–H+ exchange inhibitor No effect on infarct size or clinical outcomes
cariporide
Zeymer et al.29 1,389 3 During thrombolysis, Na+–H+ exchange inhibitor No effect on infarct size or clinical outcomes
before PCI eniporide
Bär et al.30 387 3.5 Before PCI Intravenous MCC-135 No effect on infarct size of LVEF measured on SPECT
at either 7 days or 30 days
n e w e ng l a n d j o u r na l

Jang et al.31 Before PCI Intravenous MCC-135 Still recruiting


of

Antiinflammatory agent
Baran et al.32 394 3.5 Before or during Anti-CD18 antibody No effect on infarct size, coronary blood flow, or ST-
thrombolysis segment resolution
Faxon et al.33 420 3.8 Before PCI Anti-CD11 and anti-CD18 antibody No effect on infarct size measured on SPECT at 5–9

n engl j med 357;11  www.nejm.org  september 13, 2007


days and no effect on TIMI flow or clinical events
m e dic i n e

Copyright © 2007 Massachusetts Medical Society. All rights reserved.


Tanguay et al.34 During thrombolysis P-selectin antagonist No effect on infarct size measured on SPECT or LVEF
at 30 days or on ST-segment resolution or clinical

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


outcomes
Mertens et al.35 88 ≤6 During thrombolysis P-selectin antagonist Prematurely discontinued but no effect on myocardial
blood flow, LVEF, or ST-segment resolution
Mahaffey et al.36 943 2.7 During thrombolysis Pexelizumab (Alexion) (an anti-C5 No difference in CK-MB–measured infarct size or 90-
complement antibody) day composite end point of death, cardiac failure,
or stroke
Granger et al.37 960 3.2 Before PCI Pexelizumab No difference in CK-MB–measured infarct size or 90-
day composite end point of death, cardiac failure,
or stroke
Armstrong et al.38 5,745 3.2 Before PCI Pexelizumab No difference in 30-day mortality or 90-day composite
end point of death or cardiac failure
Adenosine
Ross et al.39 and Kloner et al.40 2,118 3.3 15 min after PCI Intravenous adenosine An 11% reduction in infarct size but no effect on clini-
cal outcomes; however, subgroup analysis re-
vealed improved clinical outcomes in patients re-
ceiving adenosine ≤3 hr after onset of chest pain
Metabolic modulation (glucose,
insulin, and potassium)
Mehta et al.41 20,201 3.9 Both before and after Intravenous glucose, insulin, and No effect on mortality, cardiac arrest, cardiogenic
reperfusion potassium given during throm- shock, or reinfarction at 30 days
bo­lysis or PCI
Beshansky and Selker 42 15,450 Before reperfusion Intravenous glucose, insulin, and Currently recruiting, expected completion in August
potassium given in ambulance 2009
Magnesium
Woods et al.43 2,316 During thrombolysis Intravenous magnesium Reduced mortality and cardiac failure with magne-
sium treatment
ISIS-444 4,319 During thrombolysis Intravenous magnesium No effect on mortality
Santoro et al.45 150 3.3 Before PCI Intravenous magnesium No effect on infarct zone wall-motion score or LVEF
Antman et al.46 6,213 3.8 Before PCI or before Intravenous magnesium No effect on 30-day mortality
or during throm­
bolysis
Nicorandil
Ono et al.47 58 5.6 Before PCI Intravenous nicorandil Improved microcirculation and clinical outcomes in
short term
Mechanisms of Disease

Ishii et al.48 360 4.8 Before PCI Intravenous nicorandil Improved myocardial reperfusion and fewer deaths
and less cardiac failure after 2.4-yr follow-up
Kitakaze et al.49 545 Before PCI Intravenous nicorandil No effect on mortality, infarct size, LVEF, or myocardi-
al reperfusion

n engl j med 357;11  www.nejm.org  september 13, 2007


Therapeutic hypothermia
Dixon et al.50 42 3.5 Before PCI Endovascular cooling to 34.7°C Nonsignificant reduction in adverse cardiac events
for first 3 hr of reperfusion and infarct size
O’Neill51 400 <6 Before PCI Endovascular cooling to 34.7°C No difference in adverse cardiac events and infarct

Copyright © 2007 Massachusetts Medical Society. All rights reserved.


for first 3 hr of reperfusion size, although patients with anterior acute myo-
cardial infarction who are sufficiently cooled be-

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


fore PCI may benefit
Ly et al.52 12 3 During PCI Noninvasive surface cooling to 34.5°C Safe and feasible

* PCI denotes percutaneous coronary intervention, LVEF left ventricular ejection fraction, SPECT single-photon-emission computed tomography, TIMI Thrombolysis in Myocardial
Infarction, and CK-MB creatine kinase MB isoform.

1127
The n e w e ng l a n d j o u r na l of m e dic i n e

Blood Institute (NHLBI) charged with investigat­ phil accumulation,88 and delaying the restoration
ing the recurring issue of the lack of clinical trans­ of neutral pH.89 Furthermore, ischemic postcon­
lation of preclinical findings,3 only agents that ditioning activates the RISK pathway 8 and inhib­
have been conclusively shown to be cardioprotec­ its the opening of the mitochondrial PTP 9 —
tive in experiments in animals by multiple inves­ both important ways of protecting against lethal
tigators should be investigated in the clinical reperfusion injury. Ischemic preconditioning, a
setting.78 phenomenon in which the size of a myocardial
infarct is reduced by initiating episodes of tran­
sient myocardial ischemia and reperfusion be­
Ne w S t r ategie s for Pr e v en t ing
L e th a l R eper f usion Inj ur y fore the sustained ischemic episode, appears to
inhibit lethal reperfusion injury through the
Targeting individual mediators of lethal reperfu­ same mechanisms as ischemic postcondition­
sion injury has produced discrepant findings in ing.90
studies in animals, and clinical studies that use Several small studies have used ischemic
this strategy have not been successful. A more ef­ postconditioning in patients with acute myocar­
fective approach may be to target more than one dial infarction who are undergoing PCI with a
mediator at a time (Fig. 3). The recently described protocol that has reduced myocardial infarct
interventional strategy of ischemic postcondition­ size by 36% and improved myocardial reperfu­
ing, which by its nature targets several mediators sion (Table 3).5-7 The effect of ischemic postcon­
of lethal reperfusion injury, has been shown to ditioning on clinical outcomes remains to be
reduce myocardial injury in patients with acute determined, however.85,86
myocardial infarction who are undergoing PCI.5 A closely related strategy for preventing lethal
These findings, along with a number of preclini­ reperfusion injury is to initiate, at the time of
cal studies,8,19,80 have not only re-ignited interest myocardial reperfusion, transient episodes of
in the myocardial reperfusion phase as a target for ischemia and reperfusion in a tissue or an organ
cardioprotection, but they also have provided con­ remote from the heart. This phenomenon is
firmatory evidence of the existence of lethal re­ termed remote ischemic postconditioning.91 Pre­
perfusion injury in humans (Table 3).85,86 Further­ liminary clinical trials are under way to deter­
more, the RISK pathway 8 and the mitochondrial mine whether transient upper-limb ischemia can
PTP 9 are emerging as new targets for preventing reduce myocardial injury in patients with acute
lethal reperfusion injury (Fig. 3). myocardial infarction who are undergoing PCI.
Given the invasive nature of the ischemic post­
Ischemic Postconditioning conditioning protocol and its restriction to pa­
In 2003, Zhao et al.4 showed that after a 45-min­ tients with acute myocardial infarction who are
ute episode of sustained myocardial ischemia, the undergoing PCI, the use of pharmacologic agents
interruption of myocardial reperfusion with three that recruit the signal-transduction pathway ac­
30-second cycles of myocardial ischemia and re­ tivated by ischemic postconditioning may be a
perfusion could reduce the myocardial infarct size more effective strategy.
in dogs from 47% to 11%.4 They named this form
of cardioprotection ischemic postconditioning, a Targeting the RISK Pathway
term that highlights the myocardial reperfusion The RISK pathway 92 refers to a group of protein
phase as a target of cardioprotection, although in kinases that, when specifically activated during
reality it constitutes another variant of modified myocardial reperfusion, confer cardioprotection
myocardial reperfusion.87 by preventing lethal reperfusion injury 8,19; in a
The mechanism of ischemic postconditioning– sense, the RISK pathway mediates a form of pro­
induced protection is not fully understood, but the grammed cell survival. There is extensive pre­
procedure has been shown to target the important clinical evidence that activation of the RISK path­
mediators of lethal reperfusion injury by reducing way by pharmacologic agents8,80 or by mechanical
oxidative stress, decreasing intracellular Ca2+ over­ interventions such as ischemic preconditioning
load, improving endothelial function, attenuating or postconditioning90 reduces myocardial infarct
apoptotic cardiomyocyte death, reducing neutro­ size by up to 50%. The cardioprotection has been

1128 n engl j med 357;11  www.nejm.org  september 13, 2007

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

Table 2. Major Differences between Animal Models and Clinical Studies of Patients with Acute Myocardial Infarction.*

Characteristic Animal Models Clinical Studies Comments


Subjects Most studies use a homogeneous Studies use heterogeneous, middle- Encourage the use of older animals
group of healthy, relatively aged patient populations with with coexisting illnesses such as
young animals free of coexist- coexisting illnesses such as dia- diabetes, hyperlipidemia, athero-
ing illnesses. betes, hypertension, and dyslip- sclerosis, and hypertension to en-
idemia, all of which may influ- sure cardioprotection is possible
ence cardioprotection. in these settings.
Medication In most studies, the animals are Patients may be taking different Ensure that patients are not receiving
receiving no other medication. medications that may influence medication that could interfere
cardioprotection. with cardioprotection.
Period of acute myocar- Beneficial effects with cardiopro- Most patients present with longer Consider selecting certain patient
dial ischemia tection are observed after rela- periods of ischemia, ranging groups such as those presenting
tively short periods of isch- from 3 to 12 hr. Both the dura- early (<3 hr) after symptom onset
emia, ranging from 30 to 60 tion and severity of ischemia vary or those with an anterior myocardi-
min. The animals are subjected between patients within the al infarction. Alternatively, use more
to the same duration and se- same study; these factors may clinically relevant animal models
verity of ischemia. influence cardioprotection. such as a human-sized pig subject-
ed to a long period of ischemia.
Reperfusion time Most studies assess cardioprotec- Much longer periods of reperfusion Encourage the use of a longer period
tion after relatively short peri- occur in patients, permitting of reperfusion in studies in ani-
ods of reperfusion, ranging time for the effects of infarct mals.
from 120 min to 3 days. healing and left ventricular re-
modeling to take place.
Infarction model In most studies, acute coronary An acute myocardial infarction is an Consider using more clinically relevant
occlusion is mechanically in- acute inflammatory condition. In animal models such as animals
duced in healthy coronary ar- most patients with this condi- with atherosclerotic hearts.
teries. tion, acute coronary occlusion is
due to thrombus formation at a
site of a ruptured coronary ath-
erosclerotic plaque.
Intervention Many of the interventions admin- If interventions have not shown con- In the clinical setting, use only inter-
istered at the time of myocardi- clusive cardioprotection in exper- ventions rigorously shown in ex-
al reperfusion have not shown imental studies, they are also un- perimental studies to be conclu-
conclusive cardioprotection. likely to be cardioprotective in sively cardioprotective. A potential
the clinical setting. approach would be the use of the
intervention in a multicenter, ran-
domized, controlled study in the
animal model.†
Timing of intervention The timing of the intervention rela- The timing of the intervention rela- Consider selecting certain patient
tive to the period of ischemia tive to the period of ischemia groups, such as those presenting
and the onset of myocardial re- and the onset of myocardial re- after a specific time. In clinical
perfusion is similar in all ani- perfusion varies between pa- studies, ensure that the interven-
mals. tients. The timing of the inter- tion is administered before myo-
vention should be guided by the cardial reperfusion.
studies in animals.
Infarct size Varies from 30% to 60% of the Infarct sizes of 13% to 16% ex- Encourage the use of more accurate
total volume of myocardium at pressed as a percentage of left measurement of infarct size using
risk, providing a greater scope ventricular volume (using delayed-enhancement cardiac mag-
for cardioprotection. SPECT) appear to be the normal netic resonance imaging, which can
range, which may limit the scope express infarct size as a percentage
for cardioprotection. of the ischemic risk area.
End points for cardio- Most studies use recovery of left The clinically relevant end points are Consider more robust end points in
protection ventricular function or myocar- outcomes such as short-term studies in animals, such as long-
dial infarct size as the mea- and long-term effects on illness term effects on left ventricular
sured end points. and death. function and death.

* SPECT denotes single-photon-emission computed tomography.


† Data are from Bolli et al.3 and Baxter et al.78

n engl j med 357;11  www.nejm.org  september 13, 2007 1129

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Acute myocardial Cardioprotective Myocardial Less lethal


infarction strategy reperfusion reperfusion injury

Ischemic postconditioning
Thrombolytic
Pharmacologic activation
therapy
of the RISK pathway

Pharmacologic inhibition
of mitochondrial PTP opening
Percutaneous
Combined pharmacologic coronary intervention
approach

Figure 3. New Cardioprotective Strategies for Reducing Lethal Reperfusion Injury.


For patients with an acute myocardial infarction, ischemic postconditioning or pharmacologic agents that activate the reperfusion injury
salvage kinase (RISK) pathway or inhibit the opening of the mitochondrial permeability transition pore (PTP), or a multitargeted phar-
macologic approach before or during the immediate onset of myocardial
C O L O R F I G Ureperfusion,
RE may attenuate lethal reperfusion injury and reduce
the final myocardial infarct size. Draft 3 08/09/07
Author Yellon
Fig # 3
Title
attributed to inhibition of mitochondrial
ME PTP open­ KAI-9803, an inhibitor of protein kinase delta,
ing,93 improved uptake of CaDE 2+ in the sarcoplasmic during myocardial reperfusion (Table 3).81
Artist SBL
reticulum,94 and the recruitment ofAUTHOR antiapoptotic
PLEASE NOTE:
pathways.19 Targeting the Mitochondrial PTP
Figure has been redrawn and type has been reset
Please check carefully

Pharmacologic agents such as


Issue date glucagon-like Proof-of-concept clinical studies are in progress to
peptide 1,95 erythropoietin,96 atorvastatin,97 and determine whether pharmacologic suppression of
atrial natriuretic peptide,98 all of which reduce in­ mitochondrial PTP opening by an intravenous bo­
farct size by activating the RISK pathway, are being lus of cyclosporine, administered immediately be­
examined in proof-of-concept studies in patients fore PCI, confers cardioprotection during an acute
with acute myocardial infarction who are under­ myocardial infarction (Table 3). Pharmacologic in­
going PCI (Table 3).49,82,83 A recent clinical study hibition of mitochondrial PTP opening during
has shown that high-dose atorvastatin given to myocardial reperfusion with the use of cyclospo­
patients with a non–ST-elevation myocardial in­ rine or sanglifehrin A reduces myocardial infarct
farction at the time of urgent PCI reduces myo­ size in studies in animals by up to 50%, suggesting
cardial injury during PCI.84 that mitochondrial PTP opening may contribute to
Protein kinase C is another potential prosur­ half of the final infarct size.10,100 Furthermore,
vival protein kinase implicated in cardioprotec­ mice lacking cyclophilin D (a key component of the
tion; in animal infarct models, activation of the mitochondrial PTP) have been reported to sustain
cardioprotective protein kinase C epsilon isoform smaller myocardial infarcts than control ani­
or inhibition of the pro-injurious protein kinase mals.101 Pharmacologic inhibition of mitochon­
C delta isoform reduces myocardial infarct size drial PTP in human atrial trabeculae subjected to
when administered during myocardial reperfu­ simulated ischemia–reperfusion injury is also car­
sion.99 A preliminary clinical study has reported dioprotective.102 Studies are under way to investi­
reduced myocardial infarct size in patients under­ gate the mitochondrial PTP as a target for cardio­
going primary PCI who were given intracoronary protection in the clinical setting. However, more

1130 n engl j med 357;11  www.nejm.org  september 13, 2007

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

Table 3. New Cardioprotective Strategies for Reducing Lethal Reperfusion Injury in Patients with Acute Myocardial Infarction.*

Cardioprotective No. of Period of Timing of


Strategy and Source Patients Ischemia Intervention Details of Study Clinical End Points
hr
Ischemic postconditioning
Staat et al.5 30 5.5 During PCI Four 60-sec low-pressure in- Reduced infarct size by 36%
flations and deflations of and improved myocardial
coronary-angioplasty bal- reperfusion
loon immediately after
stent deployment
Laskey6 17 5.7 During PCI One 90-sec inflation and de- Improved ST-segment reso-
flation of coronary-angio- lution and coronary blood
plasty balloon immediate- flow
ly after stent deployment
Ma et al.7 94 7 During PCI Three 30-sec low-pressure in- Reduced infarct size, im-
flations and deflations of proved wall-motion score
coronary-angioplasty bal- index, increased myocar-
loon immediately after dial reperfusion, and im-
stent deployment proved endothelial func-
tion
Atrial natriuretic peptide
Kitakaze et al.49 569 Before PCI Intravenous infusion Reduced infarct size by 15%,
improved LVEF by 5%,
and improved myocardial
reperfusion, but no effect
on mortality; reduced
composite end point of
cardiac death and cardiac
failure
Protein kinase C–delta
inhibitor (KAI-9803)
Roe81 150 Before PCI Intracoronary bolus of KAI- Reduced infarct size and im-
9803 proved ST-segment reso-
lution
Glucagon-like peptide 1
Nikolaidis et al.82 21 6.3 3 hr after PCI Intravenous glucagon-like Improved LVEF from 29%
peptide 1 given to pa- to 39%
tients with poor LVEF
Darbepoetin alfa
(a long-acting eryth-
ropoietin analogue)
Lipsic et al.83 22 3.3 Before PCI Intravenous bolus of darbe- Mobilized endothelial progen-
poetin alfa itor cells but no effect on
left ventricular function
Remote ischemic postcondi- Before PCI Remote ischemic postcondi- In progress
tioning tioning using transient
upper-limb ischemia
Atorvastatin84 171 Before PCI High-dose atorvastatin ad- Reduced myocardial injury
ministered 12 hr before during PCI
PCI
Mitochondrial PTP inhibi- Before PCI Intravenous bolus of cyclo- In progress
tion sporine

* LVEF denotes left ventricular ejection fraction, PCI percutaneous coronary intervention, and PTP permeability transition pore.

n engl j med 357;11  www.nejm.org  september 13, 2007 1131

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

specific and safer inhibitors of mitochondrial PTP itself, however, can induce injury to the myocar­
opening need to be developed in order to take ad­ dium, thereby reducing the beneficial effects of
vantage of this strategy. myocardial reperfusion. The cardiomyocyte death
associated with the irreversible, lethal form of myo­
Wavefront of Reperfusion Injury cardial reperfusion injury diminishes the infarct-
There is emerging experimental evidence that myo­ reducing effects of myocardial reperfusion by in­
cardial infarct size can increase with the duration dependently inducing cardiomyocyte death. For
of myocardial reperfusion, suggesting a potential this reason, lethal reperfusion injury would be ex­
wavefront of myocardial reperfusion injury medi­ pected to adversely affect clinical outcomes after
ated by apoptosis and the inflammation-induced an acute myocardial infarction, and it may contrib­
death of cardiomyocytes.103 These findings raise ute to the mortality despite early and successful
the possibility of reducing myocardial infarct size reperfusion.
by intervening late in myocardial reperfusion with Until recently, the efficacy that has been shown
the use of antiapoptotic and antiinflammatory for most cardioprotective agents in animal mod­
agents.104 els has been difficult to confirm in clinical trials.
This new strategy and the strategies described There is, however, general agreement that ischemic
above for preventing lethal reperfusion injury are preconditioning and postconditioning are cardio­
important because they protect cardiomyocytes by protective not only in animal hearts but also in
means of a mechanism that is effective in all ex­ human hearts. The increasing understanding of
perimental studies; this mechanism also under­ the mechanism of the protection, particularly with
lies the cardioprotective phenomena of ischemic regard to the RISK pathway and the inhibition of
preconditioning and ischemic postconditioning. mitochondrial PTP opening, has led to the devel­
Large clinical trials will be required to ensure that opment of new pharmacologic interventions to
these new cardioprotective strategies improve clin­ invoke the mechanism at the time of reperfusion
ical outcomes in patients with acute myocardial (Fig. 3). These pathways, which can target all of
infarction. Such new cardioprotective strategies the known mediators of lethal reperfusion injury,
may also benefit patients who sustain acute myo­ have already been shown to reduce lethal reperfu­
cardial ischemia–reperfusion injury during coro­ sion injury in small-scale trials of patients with
nary-artery bypass grafting or cardiac-transplant acute myocardial infarction who are undergoing
surgery and patients surviving a cardiac arrest. PCI.5,7 These clinical results have regenerated in­
terest in the reperfusion phase as a target for car­
C onclusions dioprotection. Preliminary clinical data indicate
that these new cardioprotective strategies confer
For patients presenting with an acute myocardial a benefit to patients with acute myocardial infarc­
infarction, early and successful myocardial reper­ tion over and above that provided by myocardial
fusion by means of thrombolytic therapy or pri­ reperfusion alone, but they remain to be con­
mary PCI is the most effective interventional strat­ firmed in large-scale clinical studies.
Supported by the British Heart Foundation.
egy for reducing infarct size and improving clinical No potential conflict of interest relevant to this article was re­
outcomes. The process of myocardial reperfusion ported.

References
1. Piper HM, Garcia-Dorado D, Ovize M. 4. Zhao ZQ, Corvera JS, Halkos ME, et al. myocardial infarction: a pilot study. Cath­
A fresh look at reperfusion injury. Cardio­ Inhibition of myocardial injury by ischemic eter Cardiovasc Interv 2005;65:361-7.
vasc Res 1998;38:291-300. postconditioning during reperfusion: com­ 7. Ma X, Zhang X, Li C, Luo M. Effect of
2. Keeley EC, Boura JA, Grines CL. Pri­ parison with ischemic preconditioning. Am postconditioning on coronary blood flow
mary angioplasty versus intravenous throm­ J Physiol Heart Circ Physiol 2003;285: velocity and endothelial function and LV
bolytic therapy for acute myocardial infarc­ H579-H588. [Erratum, Am J Physiol Heart recovery after myocardial infarction. J In­
tion: a quantitative review of 23 randomised Circ Physiol 2004;286:H477.] terv Cardiol 2006;19:367-75.
trials. Lancet 2003;361:13-20. 5. Staat P, Rioufol G, Piot C, et al. Post­ 8. Hausenloy DJ, Yellon DM. New direc­
3. Bolli R, Becker L, Gross G, Mentzer R Jr, conditioning the human heart. Circula­ tions for protecting the heart against isch­
Balshaw D, Lathrop DA. Myocardial protec­ tion 2005;112:2143-8. aemia-reperfusion injury: targeting the
tion at a crossroads: the need for translation 6. Laskey WK. Brief repetitive balloon oc­ Reperfusion Injury Salvage Kinase (RISK)-
into clinical therapy. Circ Res 2004;95:125- clusions enhance reperfusion during per­ pathway. Cardiovasc Res 2004;61:448-60.
34. cutaneous coronary intervention for acute 9. Idem. The mitochondrial permeability

1132 n engl j med 357;11  www.nejm.org  september 13, 2007

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

transition pore: its fundamental role in with and without thrombolytic therapy: et al. Efficacy of a novel P-selectin antago­
mediating cell death during ischaemia a double-blind, placebo-controlled, ran­ nist, rPSGL-Ig for reperfusion therapy in
and reperfusion. J Mol Cell Cardiol 2003; domized trial. Eur Heart J 2000;21:1537- acute myocardial infarction: the RAPSODY
35:339-41. 46. trial. J Am Coll Cardiol 2003;41:Suppl:
10. Hausenloy DJ, Duchen MR, Yellon 25. Guan W, Osanai T, Kamada T, et al. 404A. abstract.
DM. Inhibiting mitochondrial permeabil­ Effect of allopurinol pretreatment on free 35. Mertens P, Maes A, Nuyts J, et al. Re­
ity transition pore opening at reperfusion radical generation after primary coronary combinant P-selectin glycoprotein ligand-
protects against ischaemia-reperfusion in­ angioplasty for acute myocardial infarc­ immunoglobulin, a P-selectin antagonist,
jury. Cardiovasc Res 2003;60:617-25. tion. J Cardiovasc Pharmacol 2003;41:699- as an adjunct to thrombolysis in acute
11. Yellon DM, Hausenloy DJ. Realizing 705. myocardial infarction: the P-Selectin An­
the clinical potential of ischemic precon­ 26. Tsujita K, Shimomura H, Kaikita K, et tagonist Limiting Myonecrosis (PSALM)
ditioning and postconditioning. Nat Clin al. Long-term efficacy of edaravone in pa­ trial. Am Heart J 2006;152:125.e1-125.e8.
Pract Cardiovasc Med 2005;2:568-75. tients with acute myocardial infarction. 36. Mahaffey KW, Granger CB, Nicolau
12. Jennings RB, Sommers HM, Smyth Circ J 2006;70:832-7. JC, et al. Effect of pexelizumab, an anti-
GA, Flack HA, Linn H. Myocardial necro­ 27. Boden WE, van Gilst WH, Schelde­ C5 complement antibody, as adjunctive
sis induced by temporary occlusion of a waert RG, et al. Diltiazem in acute myocar­ therapy to fibrinolysis in acute myocardi­
coronary artery in the dog. Arch Pathol dial infarction treated with thrombolytic al infarction: the COMPlement inhibition
1960;70:68-78. agents: a randomised placebo-controlled in myocardial infarction treated with
13. Braunwald E, Kloner RA. The stunned trial. Lancet 2000;355:1751-6. thromboLYtics (COMPLY) trial. Circulation
myocardium: prolonged, postischemic ven­ 28. Théroux P, Chaitman BR, Danchin N, 2003;108:1176-83.
tricular dysfunction. Circulation 1982;66: et al. Inhibition of the sodium-hydrogen 37. Granger CB, Mahaffey KW, Weaver
1146-9. exchanger with cariporide to prevent myo­ WD, et al. Pexelizumab, an anti-C5 com­
14. Krug A, Du Mesnil de Rochemont R, cardial infarction in high-risk ischemic sit­ plement antibody, as adjunctive therapy to
Korb G. Blood supply of the myocardium uations: main results of the GUARDIAN primary percutaneous coronary interven­
after temporary coronary occlusion. Circ trial. Circulation 2000;102:3032-8. tion in acute myocardial infarction: the
Res 1966;19:57-62. 29. Zeymer U, Suryapranata H, Monassier COMplement inhibition in Myocardial in­
15. Ito H. No-reflow phenomenon and JP, et al. The Na(+)/H(+) exchange inhibi­ farction treated with Angioplasty (COMMA)
prognosis in patients with acute myocar­ tor eniporide as an adjunct to early reper­ trial. Circulation 2003;108:1184-90.
dial infarction. Nat Clin Pract Cardiovasc fusion therapy for acute myocardial in­ 38. Armstrong PW, Granger CB, Adams
Med 2006;3:499-506. farction: results of the Evaluation of the PX, et al. Pexelizumab for acute ST-eleva­
16. Manning AS, Hearse DJ. Reperfusion- Safety and Cardioprotective Effects of tion myocardial infarction in patients un­
induced arrhythmias: mechanisms and pre­ Eniporide in Acute Myocardial Infarction dergoing primary percutaneous coronary
vention. J Mol Cell Cardiol 1984;16:497- (ESCAMI) trial. J Am Coll Cardiol 2001; intervention: a randomized controlled tri­
518. 38:1644-50. al. JAMA 2007;297:43-51.
17. Bolli R, Marbán E. Molecular and cel­ 30. Bär FW, Tzivoni D, Dirksen MT, et al. 39. Ross AM, Gibbons RJ, Stone GW,
lular mechanisms of myocardial stun­ Results of the first clinical study of ad­ Kloner RA, Alexander RW. A randomized,
ning. Physiol Rev 1999;79:609-34. junctive CAldaret (MCC-135) in patients double-blinded, placebo-controlled multi­
18. Kloner RA. Does reperfusion injury undergoing primary percutaneous coro­ center trial of adenosine as an adjunct to
exist in humans? J Am Coll Cardiol 1993; nary intervention for ST-Elevation Myo­ reperfusion in the treatment of acute myo­
21:537-45. cardial Infarction: the randomized multi­ cardial infarction (AMISTAD-II). J Am Coll
19. Yellon DM, Baxter GF. Reperfusion centre CASTEMI study. Eur Heart J 2006; Cardiol 2005;45:1775-80.
injury revisited: is there a role for growth 27:2516-23. 40. Kloner RA, Forman MB, Gibbons RJ,
factor signaling in limiting lethal reper­ 31. Jang IK, Pettigrew V, Picard MH, et al. Ross AM, Alexander RW, Stone GW. Im­
fusion injury? Trends Cardiovasc Med 1999; A randomized, double-blind, placebo-con­ pact of time to therapy and reperfusion
9:245-9. trolled study of the safety and efficacy of modality on the efficacy of adenosine in
20. Zweier JL. Measurement of superox­ intravenous MCC-135 as an adjunct to pri­ acute myocardial infarction: the AMIS­
ide-derived free radicals in the reperfused mary percutaneous coronary intervention TAD-2 trial. Eur Heart J 2006;27:2400-5.
heart: evidence for a free radical mecha­ in patients with acute myocardial infarc­ 41. Mehta SR, Yusuf S, Diaz R, et al. Ef­
nism of reperfusion injury. J Biol Chem tion: rationale and design of the Evalua­ fect of glucose-insulin-potassium infu­
1988;263:1353-7. tion of MCC-135 for Left Ventricular Salvage sion on mortality in patients with acute
21. Hearse DJ, Humphrey SM, Chain EB. in Acute MI (EVOLVE) study. J Thromb ST-segment elevation myocardial infarc­
Abrupt reoxygenation of the anoxic potas­ Thrombolysis 2005;20:147-53. tion: the CREATE-ECLA randomized con­
sium-arrested perfused rat heart: a study 32. Baran KW, Nguyen M, McKendall GR, trolled trial. JAMA 2005;293:437-46.
of myocardial enzyme release. J Mol Cell et al. Double-blind, randomized trial of 42. Beshansky J, Selker H. Immediate Met­
Cardiol 1973;5:395-407. an anti-CD18 antibody in conjunction abolic Myocardial Enhancement During
22. Flaherty JT, Pitt B, Gruber JW, et al. with recombinant tissue plasminogen ac­ Initial Assessment and Treatment in Emer­
Recombinant human superoxide dis­ tivator for acute myocardial infarction: gency Care (IMMEDIATE) clinical study.
mutase (h-SOD) fails to improve recovery Limitation of Myocardial Infarction fol­ (Accessed August 20, 2007, at http://www.
of ventricular function in patients under­ lowing Thrombolysis in Acute Myocardial immediatetrial.com.)
going coronary angioplasty for acute myo­ Infarction (LIMIT AMI) study. Circulation 43. Woods KL, Fletcher S, Roffe C, Haider
cardial infarction. Circulation 1994;89: 2001;104:2778-83. Y. Intravenous magnesium sulphate in sus­
1982-91. 33. Faxon DP, Gibbons RJ, Chronos NA, pected acute myocardial infarction: results
23. Downey JM. Free radicals and their Gurbel PA, Sheehan F. The effect of block­ of the second Leicester Intravenous Mag­
involvement during long-term myocar­ ade of the CD11/CD18 integrin receptor on nesium Intervention Trial (LIMIT-2). Lan­
dial ischemia and reperfusion. Annu Rev infarct size in patients with acute myocar­ cet 1992;339:1553-8.
Physiol 1990;52:487-504. dial infarction treated with direct angio­ 44. ISIS-4 (Fourth International Study of
24. Effect of 48-h intravenous trimetazi­ plasty: the results of the HALT-MI study. J Infarct Survival) Collaborative Group.
dine on short- and long-term outcomes of Am Coll Cardiol 2002;40:1199-204. ISIS-4: a randomised factorial trial assess­
patients with acute myocardial infarction, 34. Tanguay JF, Krucoff MW, Gibbons RJ, ing early oral captopril, oral mononitrate,

n engl j med 357;11  www.nejm.org  september 13, 2007 1133

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

and intravenous magnesium sulphate in 56. Carry MM, Mrak RE, Murphy ML, al. Reduction of reperfusion injury in the
58,050 patients with suspected acute myo­ Peng CF, Straub KD, Fody EP. Reperfusion canine preparation by intracoronary ade­
cardial infarction. Lancet 1995;345:669- injury in ischemic myocardium: protec­ nosine: importance of the endothelium
85. tive effects of ruthenium red and of nitro­ and the no-reflow phenomenon. Circula­
45. Santoro GM, Antoniucci D, Bolognese prusside. Am J Cardiovasc Pathol 1989; tion 1987;76:1135-45.
L, et al. A randomized study of intrave­ 2:335-44. 69. Goto M, Miura T, Iliodoromitis EK, et
nous magnesium in acute myocardial in­ 57. Gumina RJ, Buerger E, Eickmeier C, al. Adenosine infusion during early reper­
farction treated with direct coronary an­ Moore J, Daemmgen J, Gross GJ. Inhibi­ fusion failed to limit myocardial infarct
gioplasty. Am Heart J 2000;140:891-7. tion of the Na(+)/H(+) exchanger confers size in a collateral deficient species. Car­
46. Antman E, Cooper H, McKinlay S, et greater cardioprotection against 90 min­ diovasc Res 1991;25:943-9.
al. Early administration of intravenous utes of myocardial ischemia than isch­ 70. Jonassen AK, Sack MN, Mjos OD, Yel­
magnesium to high-risk patients with emic preconditioning in dogs. Circulation lon DM. Myocardial protection by insulin
acute myocardial infarction in the Magne­ 1999;100:2519-26. at reperfusion requires early administra­
sium in Coronaries (MAGIC) Trial: a ran­ 58. Avkiran M, Marber MS. Na(+)/H(+) ex­ tion and is mediated via Akt and p70s6
domised controlled trial. Lancet 2002;360: change inhibitors for cardioprotective ther­ kinase cell-survival signaling. Circ Res
1189-96. apy: progress, problems and prospects. 2001;89:1191-8.
47. Ono H, Osanai T, Ishizaka H, et al. J Am Coll Cardiol 2002;39:747-53. 71. Apstein CS, Opie LH. A challenge to
Nicorandil improves cardiac function and 59. Lemasters JJ, Bond JM, Chacon E, et the metabolic approach to myocardial is­
clinical outcome in patients with acute al. The pH paradox in ischemia-reperfu­ chae­mia. Eur Heart J 2005;26:956-9.
myocardial infarction undergoing primary sion injury to cardiac myocytes. EXS 1996; 72. Opie LH. The glucose hypothesis: re­
percutaneous coronary intervention: role 76:99-114. lation to acute myocardial ischemia. J Mol
of inhibitory effect on reactive oxygen spe­ 60. Bond JM, Herman B, Lemasters JJ. Cell Cardiol 1970;1:107-15.
cies formation. Am Heart J 2004;148:E15. Protection by acidotic pH against anoxia/ 73. Christensen CW, Rieder MA, Silver­
48. Ishii H, Ichimiya S, Kanashiro M, et reoxygenation injury to rat neonatal cardiac stein EL, Gencheff NE. Magnesium sul­
al. Impact of a single intravenous admin­ myocytes. Biochem Biophys Res Commun fate reduces myocardial infarct size when
istration of nicorandil before reperfusion 1991;179:798-803. administered before but not after coro­
in patients with ST-segment-elevation myo­ 61. Kim JS, Jin Y, Lemasters JJ. Reactive nary reperfusion in a canine model. Cir­
cardial infarction. Circulation 2005;112: oxygen species, but not Ca2+ overloading, culation 1995;92:2617-21.
1284-8. trigger pH- and mitochondrial permeabil­ 74. Hypothermia after Cardiac Arrest Study
49. Kitakaze M, Asakura M, Shintani Y, et ity transition-dependent death of adult rat Group. Mild therapeutic hypothermia to
al. Large-scale trial using atrial natriuret­ myocytes after ischemia-reperfusion. Am improve the neurologic outcome after car­
ic peptide or nicorandil as an adjunct to J Physiol Heart Circ Physiol 2006;290: diac arrest. N Engl J Med 2002;346:549-56.
percutanoeus coronary intervention for ST- H2024-H2034. [Erratum, N Engl J Med 2002;346:1756.]
segment elevation acute myocardial in­ 62. Vinten-Johansen J. Involvement of 75. Chien GL, Wolff RA, Davis RF, Van
farction. Presented at the American Heart neutrophils in the pathogenesis of lethal Winkle DM. “Normothermic range” tem­
Association Scientific Sessions, Chicago, myocardial reperfusion injury. Cardiovasc perature affects myocardial infarct size.
November 14, 2006. Res 2004;61:481-97. Cardiovasc Res 1994;28:1014-7.
50. Dixon SR, Whitbourn RJ, Dae MW, et 63. Litt MR, Jeremy RW, Weisman HF, 76. Dae MW, Gao DW, Sessler DI, Chair K,
al. Induction of mild systemic hypother­ Winkelstein JA, Becker LC. Neutrophil de­ Stillson CA. Effect of endovascular cool­
mia with endovascular cooling during pletion limited to reperfusion reduces ing on myocardial temperature, infarct
primary percutaneous coronary interven­ myocardial infarct size after 90 minutes of size, and cardiac output in human-sized
tion for acute myocardial infarction. J Am ischemia: evidence for neutrophil-mediat­ pigs. Am J Physiol Heart Circ Physiol
Coll Cardiol 2002;40:1928-34. ed reperfusion injury. Circulation 1989;80: 2002;282:H1584-H1591.
51. O’Neill W. Cooling as an adjunct to 1816-27. 77. Griffiths EJ, Halestrap AP. Mitochon­
primary PCI for myocardial infarction. 64. Hayward R, Campbell B, Shin YK, drial non-specific pores remain closed
Presented at the Transcatheter Cardiovas­ Scalia R, Lefer AM. Recombinant soluble during cardiac ischaemia, but open upon
cular Therapeutics Conference, Washing­ P-selectin glycoprotein ligand-1 protects reperfusion. Biochem J 1995;307:93-8.
ton, DC, September 18, 2003. against myocardial ischemic reperfusion 78. Baxter GF, Hale SL, Miki T, et al. Ad­
52. Ly HQ, Denault A, Dupuis J, et al. A injury in cats. Cardiovasc Res 1999;41:65- enosine A1 Agonist at Reperfusion Trial
pilot study: the Noninvasive Surface Cool­ 76. (AART): results of a three-center, blinded,
ing Thermoregulatory System for Mild Hy­ 65. Ma XL, Tsao PS, Lefer AM. Antibody randomized, controlled experimental in­
pothermia Induction in Acute Myocardial to CD-18 exerts endothelial and cardiac farct study. Cardiovasc Drugs Ther 2000;
Infarction (the NICAMI Study). Am Heart protective effects in myocardial ischemia 14:607-14.
J 2005;150:933. and reperfusion. J Clin Invest 1991;88: 79. Dirksen MT, Laarman GJ, Simoons
53. Zweier JL, Talukder MA. The role of 1237-43. ML, Duncker DJGM. Reperfusion injury
oxidants and free radicals in reperfusion 66. Zhao ZQ, Lefer DJ, Sato H, Hart KK, in humans: a review of clinical trials on
injury. Cardiovasc Res 2006;70:181-90. Jefforda PR, Vinten-Johansen J. Monoclo­ reperfusion injury inhibitory strategies.
54. Weyrich AS, Ma XL, Lefer AM. The nal antibody to ICAM-1 preserves post­ Cardiovasc Res 2007;74:343-55.
role of L-arginine in ameliorating reper­ ischemic blood flow and reduces infarct 80. Hausenloy DJ, Yellon DM. Reperfu­
fusion injury after myocardial ischemia in size after ischemia-reperfusion in rabbit. sion Injury Salvage Kinase signaling: taking
the cat. Circulation 1992;86:279-88. J Leukoc Biol 1997;62:292-300. a RISK for cardioprotection. Heart Fail Rev
55. Klein HH, Pich S, Lindert S, Neben­ 67. Vakeva AP, Agah A, Rollins SA, Matis 2007;12:217-34.
dahl K, Warneke G, Kreuzer H. Treatment LA, Li L, Stahl GL. Myocardial infarction 81. Roe M. Inhibition of d-protein kinase
of reperfusion injury with intracoronary and apoptosis after myocardial ischemia C to ameliorate reperfusion injury during
calcium channel antagonists and reduced and reperfusion: role of the terminal primary percutaneous coronary interven­
coronary free calcium concentration in complement components and inhibition tion for acute ST-elevation myocardial in­
regionally ischemic, reperfused porcine by anti-C5 therapy. Circulation 1998;97: farction: results from the DELTA MI trial.
hearts. J Am Coll Cardiol 1989;13:1395- 2259-67. J Am Coll Cardiol 2007;49:215A. abstract.
401. 68. Olafsson B, Forman MB, Puett DW, et 82. Nikolaidis LA, Mankad S, Sokos GG,

1134 n engl j med 357;11  www.nejm.org  september 13, 2007

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

et al. Effects of glucagon-like peptide-1 in preconditioning and postconditioning. tects the myocardium by up-regulating a
patients with acute myocardial infarction Trends Cardiovasc Med 2005;15:69-75. pro-survival pathway. J Am Coll Cardiol
and left ventricular dysfunction after suc­ 91. Kerendi F, Kin H, Halkos ME, et al. 2003;41:508-15.
cessful reperfusion. Circulation 2004;109: Remote postconditioning: brief renal 98. Yang XM, Philipp S, Downey JM, Co­
962-5. ischemia and reperfusion applied before hen MV. Atrial natriuretic peptide admin­
83. Lipsic E, van der Meer P, Voors AA, et coronary artery reperfusion reduces myo­ istered just prior to reperfusion limits in­
al. A single bolus of a long-acting erythro­ cardial infarct size via endogenous activa­ farction in rabbit hearts. Basic Res Cardiol
poietin analogue darbepoetin alfa in pa­ tion of adenosine receptors. Basic Res Car­ 2006;101:311-8.
tients with acute myocardial infarction: diol 2005;100:404-12. 99. Inagaki K, Churchill E, Mochly-Rosen
a randomized feasibility and safety study. 92. Zhao ZQ, Morris CD, Budde JM, et al. D. Epsilon protein kinase C as a potential
Cardiovasc Drugs Ther 2006;20:135-41. Inhibition of myocardial apoptosis reduc­ therapeutic target for the ischemic heart.
84. Patti G, Pasceri V, Colonna G, et al. es infarct size and improves regional con­ Cardiovasc Res 2006;70:222-30.
Atorvastatin pretreatment improves out­ tractile dysfunction during reperfusion. 100. Hausenloy DJ, Maddock HL, Baxter
comes in patients with acute coronary Cardiovasc Res 2003;59:132-42. GF, Yellon DM. Inhibiting mitochondrial
syndromes undergoing early percutane­ 93. Davidson SM, Hausenloy D, Duchen permeability transition pore opening: a new
ous coronary intervention: results of the MR, Yellon DM. Signalling via the Reper­ paradigm for myocardial preconditioning?
ARMYDA-ACS randomized trial. J Am fusion Injury Signalling Kinase (RISK) Cardiovasc Res 2002;55:534-43.
Coll Cardiol 2007;49:1272-8. pathway links closure of the mitochon­ 101. Baines CP, Kaiser RA, Purcell NH, et
85. Vinten-Johansen J, Yellon DM, Opie drial permeability transition pore to cardio­ al. Loss of cyclophilin D reveals a critical
LH. Postconditioning: a simple, clinically protection. Int J Biochem Cell Biol 2006; role for mitochondrial permeability tran­
applicable procedure to improve revascu­ 38:414-9. sition in cell death. Nature 2005;434:658-
larization in acute myocardial infarction. 94. Abdallah Y, Gkatzoflia A, Gligorievski 62.
Circulation 2005;112:2085-8. D, et al. Insulin protects cardiomyocytes 102. Shanmuganathan S, Hausenloy DJ,
86. Yellon DM, Opie LH. Postcondition­ against reoxygenation-induced hypercon­ Duchen MR, Yellon DM. Mitochondrial
ing for protection of the infarcting heart. tracture by a survival pathway targeting permeability transition pore as a target
Lancet 2006;367:456-8. SR Ca2+ storage. Cardiovasc Res 2006;70: for cardioprotection in the human heart.
87. Heusch G. Postconditioning: old wine 346-53. Am J Physiol Heart Circ Physiol 2005;289:
in a new bottle? J Am Coll Cardiol 2004; 95. Bose AK, Mocanu MM, Carr RD, H237-H242.
44:1111-2. Brand CL, Yellon DM. Glucagon-like pep­ 103. Zhao ZQ, Nakamura M, Wang NP, et
88. Zhao ZQ, Vinten-Johansen J. Postcondi­ tide 1 can directly protect the heart against al. Dynamic progression of contractile
tioning: reduction of reperfusion-induced ischemia/reperfusion injury. Diabetes 2005; and endothelial dysfunction and infarct
injury. Cardiovasc Res 2006;70:200-11. 54:146-51. extension in the late phase of reperfusion.
89. Cohen MV, Yang XM, Downey JM. The 96. Bullard AJ, Govewalla P, Yellon DM. J Surg Res 2000;94:133-44.
pH hypothesis of postconditioning: stac­ Erythropoietin protects the myocardium 104. Doukas J, Wrasidlo W, Noronha G,
cato reperfusion reintroduces oxygen and against reperfusion injury in vitro and et al. Phosphoinositide 3-kinase gamma/
perpetuates myocardial acidosis. Circula­ in vivo. Basic Res Cardiol 2005;100:397- delta inhibition limits infarct size after
tion 2007;115:1895-903. 403. myocardial ischemia/reperfusion injury.
90. Hausenloy DJ, Tsang A, Yellon DM. 97. Bell RM, Yellon DM. Atorvastatin, ad­ Proc Natl Acad Sci U S A 2006;103:19866-
The reperfusion injury salvage kinase path­ ministered at the onset of reperfusion, 71.
way: a common target for both ischemic and independent of lipid lowering, pro­ Copyright © 2007 Massachusetts Medical Society.

posting presentations at medical meetings on the internet


Posting an audio recording of an oral presentation at a medical meeting on the
Internet, with selected slides from the presentation, will not be considered prior
publication. This will allow students and physicians who are unable to attend the
meeting to hear the presentation and view the slides. If there are any questions
about this policy, authors should feel free to call the Journal’s Editorial Offices.

n engl j med 357;11  www.nejm.org  september 13, 2007 1135

Downloaded from www.nejm.org at UNIVERSIDAD DE CHILE on August 5, 2009 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.

Potrebbero piacerti anche