Bulluck 2018 Cardiovascular Magnetic Resonance in Acute ST-Segment-Elevation Myocardial Infarction

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Cardiovascular Magnetic Resonance in Acute

ST-Segment–Elevation Myocardial Infarction


Heerajnarain Bulluck, Rohan Dharmakumar, Andrew E. Arai, Colin Berry, and Derek J. Hausenloy
2018, 137 (18), 1949-1964• DOI: 10.1161/CIRCULATIONAHA.117.030693 • Publication Date (Web): 18 Jun 2018
Downloaded from www.ahajournals.org on March 3, 2020

More About This Article

Additional resources and features associated with this article are available within the HTML version:

• Supporting Information
• Access to high resolution figures
• Links to articles and content related to this article
• Copyright permission to reproduce figures and/or text from this article
Downloaded from http://ahajournals.org by on March 3, 2020

is published by the AHA Journals.


Circulation

IN DEPTH
Cardiovascular Magnetic Resonance in Acute
ST-Segment–Elevation Myocardial Infarction
Recent Advances, Controversies, and Future Directions

ABSTRACT: Although mortality after ST-segment elevation myocardial Heerajnarain Bulluck,


infarction (MI) is on the decline, the number of patients developing heart MBBS, PhD
failure as a result of MI is on the rise. Apart from timely reperfusion Rohan Dharmakumar, PhD
by primary percutaneous coronary intervention, there is currently no Andrew E. Arai, MD
established therapy for reducing MI size. Thus, new cardioprotective Colin Berry, MBCHB, PhD
therapies are required to improve clinical outcomes after ST-segment– Derek J. Hausenloy, MD,
elevation MI. Cardiovascular magnetic resonance has emerged as an PhD
important imaging modality for assessing the efficacy of novel therapies
for reducing MI size and preventing subsequent adverse left ventricular
remodeling. The recent availability of multiparametric mapping
cardiovascular magnetic resonance imaging has provided new insights
into the pathophysiology underlying myocardial edema, microvascular
Downloaded from http://ahajournals.org by on March 3, 2020

obstruction, intramyocardial hemorrhage, and changes in the remote


myocardial interstitial space after ST-segment–elevation MI. In this article,
we provide an overview of the recent advances in cardiovascular magnetic
resonance imaging in reperfused patients with ST-segment–elevation
MI, discuss the controversies surrounding its use, and explore future
applications of cardiovascular magnetic resonance in this setting.

I
mprovements in the treatment of patients with acute ST-segment–elevation myo-
cardial infarction (STEMI) have led to a decline in mortality over the past 4 decades,1
with 1-year cardiac mortality in patients with STEMI plateauing at ≈8%.2 However,
morbidity caused by post–myocardial infarction (MI) heart failure remains significant
and is on the rise.3 The process of reperfusion itself can paradoxically induce further
myocardial injury and cardiomyocyte death, a phenomenon called myocardial reper-
fusion injury.4 There is currently no effective therapy for reducing myocardial reperfu-
sion injury, despite a wealth of research in this field. This has been attributed partly
to undertaking clinical studies despite the lack of reproducible and robust preclinical
data, and the design of the clinical cardioprotection studies in terms of patient selec-
tion and inappropriate timing and mode of delivery of the cardioprotective agent.5
Thus, the search continues for a novel and effective therapy for reducing MI size and
preventing heart failure that can be administered as an adjunct to primary percuta-
neous coronary intervention (PPCI) after STEMI. Cardiovascular magnetic resonance Key Words: hemorrhage ◼ magnetic
(CMR) has emerged as an important imaging modality for assessing the cardiopro- resonance imaging ◼ percutaneous
coronary intervention ◼ ST elevation
tective efficacy of novel therapies for reducing MI size and preventing adverse left myocardial infarction
ventricle (LV) remodeling in reperfused patients with STEMI.6,7 It is currently the gold
© 2018 American Heart Association, Inc.
standard imaging modality for quantifying MI size8 and is able to detect small suben-
docardial MI (as little as 1 g)9 with good accuracy.10 http://circ.ahajournals.org

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1949


Bulluck et al The Role of CMR in STEMI

With the recent availability of native T1, T2, T2*, and compromised, extravasation of red blood cells into the
postcontrast T1 mapping (to derive extracellular vol- myocardium can occur, which is called intramyocardial
STATE OF THE ART

ume fraction [ECV] mapping),11 more in-depth insights hemorrhage.14 The breakdown products of hemoglobin
can be obtained into the pathophysiological processes within the myocardium can be detected as a hypoin-
such as the evolution of myocardial edema in the first tense core38 within the MI zone on T2* imaging or T2*
week after STEMI,12,13 the chronic manifestation of in- mapping (Figures 1 and 2). Of the 2 approaches, T2*
tramyocardial hemorrhage, its prognostic significance mapping has been shown to have greater sensitivity
over microvascular obstruction14,15 and changes in the for detecting intramyocardial hemorrhage after STEMI
remote myocardial interstitial space in those developing compared with T2 mapping (Table).21,39
adverse LV remodeling16,17 after STEMI. In this article, A small study previously showed that the hypoin-
we provide an overview of some of the recent advances tense core on the T1 or T2 maps could provide an alter-
in CMR imaging in reperfused patients with STEMI, dis- native method to detect intramyocardial hemorrhage
cuss the recent controversies surrounding its use, and in cases when T2* is not available or not interpretable
explore future applications of CMR in this setting. (Figures 1 and 2).38 Further studies are needed to con-
firm these findings.

BASIC CONCEPTS
Area at Risk
In this section, we provide a brief description of some
of the basic concepts concerning the use of CMR in The area at risk refers to the territory supplied by the
STEMI. The Table provides an explanation of the more infarct-related artery that would have infarcted after
technical terms used throughout this article. Figure 1A STEMI if reperfusion had not taken place to salvage
also provides an illustration of the various components viable myocardium. The area at risk includes both the
of the myocardium that can be interrogated by CMR. reversibly-injured myocardium (also referred to as the
salvaged myocardium) and the infarcted myocardium.
Figure 3 shows an example of the edema-based area at
MI Size risk by T1 and T2 mapping and the corresponding MI
MI size refers to the infarcted myocardium and is conven- size on late gadolinium enhancement in a patient pre-
tionally quantified by late gadolinium enhancement (Table senting with an acute inferior STEMI treated by PPCI.
Downloaded from http://ahajournals.org by on March 3, 2020

and Figure 1B).36 The gadolinium chelate cannot cross the A number of CMR approaches to delineating the
intact cell membranes.36 After acute myocardial necrosis, area at risk after STEMI have been described (Table).
the cell membranes are ruptured, allowing the contrast However, they all have their limitations,40 and although
agent to enter these cells. In the chronic setting, there is each technique has been validated against histology
also expansion of the extracellular space caused by col- in the preclinical setting,23,26,29,30,32 there is currently no
lagen deposits and the relatively low residual amount of consensus on which CMR method should be used to
intact cardiomyocytes in the region of the infarct. There- quantify the area at risk in the clinical setting. So far, no
fore, after gadolinium chelate administration, the con- studies have compared these techniques head to head
trast redistributes itself from the vascular compartment to in the same patient cohort. Once the area at risk and
the interstitial space, and at pseudoequilibrium, a higher MI size are known, the myocardial salvage index can be
concentration of contrast is distributed in areas of acute calculated as described in the Table.
or chronic MI than in normal myocardium9 (Table). The myocardial salvage index is considered a more
sensitive measure for assessing the efficacy of novel car-
Microvascular Obstruction and dioprotective therapies compared with MI size alone34
because it normalizes the MI size reduction to the area
Intramyocardial Hemorrhage at risk, the latter of which varies from patient to pa-
Microvascular obstruction refers to the inability to re- tient. Therefore, the myocardial salvage index may help
perfuse the coronary microcirculation in a previously to reduce the sample size for clinical cardioprotection
ischemic region, despite opening of the epicardial ves- studies compared with using MI size alone.35
sel.37 Microvascular obstruction can be identified as
a dark hypointense core within the areas of hyperen-
hancement on either early gadolinium enhancement RECENT ADVANCES IN THE ROLE OF
(referred to as early microvascular obstruction) or con- CMR IN REPERFUSED PATIENTS WITH
ventional late gadolinium enhancement (referred to as
late microvascular obstruction) sequences, defined in STEMI
the Table and illustrated in Figure 1B. In this section, we elaborate on how CMR has improved
If the coronary microvasculature injury after STEMI our understanding of the relationship between micro-
is especially severe and the integrity of the vessels is vascular obstruction and intramyocardial hemorrhage,

1950 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693


Bulluck et al The Role of CMR in STEMI

Table. Technical Descriptions of the Various Components of the Myocardium Assessed by CMR in STEMI

STATE OF THE ART


Components Technical Description
Native T1, postcontrast T1, This is the time constant to recover 63% of its longitudinal magnetization (spin-lattice relaxation) and is expressed in
and ECV milliseconds. It is referred to as native T1 when performed before the administration of gadolinium chelate.11
It is recommended that postcontrast T1 be performed at least 10 min after contrast administration.11
Once the native and postcontrast T1 of the blood and myocardium and the hematocrit are known, the ECV can be calculated
with this formula11:
ECV (%)=(100−hematocrit)×(1/postcontrast T1myocardium)−(1/native T1myocardium)
(1/postcontrast T1blood)−(1/native T1blood)
T2 This refers to the constant representing the decay of transverse magnetization (spin-spin relaxation) to 37% of its initial
equilibrium value and is expressed in milliseconds.11
T2* This is the time constant representing the decay of transverse magnetization in the presence of local magnetic field
inhomogeneities and is expressed in milliseconds.11
Parametric mapping This is a process whereby an anatomic map is generated with each pixel representing a specific magnetic tissue property
(T1, T2, or T2* or ECV) derived from the spatially corresponding voxel of a set of coregistered magnetic resonance source
images.11
MI size This refers to the mass or volume of infarcted myocardium and is conventionally quantified by late gadolinium enhancement.
After 10 min, pseudoequilibrium of gadolinium chelate concentration between blood and well-perfused myocardium
develops. During this time, a higher concentration of contrast is distributed in areas of acute or chronic MI (significantly
shortening T1) than in normal myocardium. Therefore, with appropriate nulling of the remote myocardium with inversion
recovery T1-weighted sequences, the necrotic or scarred areas appear bright.6,9
Early microvascular obstruction This can be identified as areas of dark core within the MI zone with first-pass perfusion or EGE imaging performed with a
fixed, high TI (eg, 440 to 500 ms at 1.5 T) acquired at 1–4 min after contrast injection.
Late microvascular obstruction This can be identified as a dark core within the areas of hyperenhancement on conventional late gadolinium enhancement
sequences acquired >10 min after contrast injection.
Intramyocardial hemorrhage This can be identified with T2* -weighted imaging or T2* mapping.
From brain imaging data, the degradation of the extravasated erythrocytes to oxyhemoglobin, deoxyhemoglobin, and
methemoglobin (strongly paramagnetic) is dynamic and exhibits different magnetic properties at various stages, as previously
described by Bradley.18 Breakdown of the erythrocyte membrane eventually leads to ferritin and hemosiderin deposits within
the macrophages, and the iron-degradation products can be detected with T2* imaging.
Most studies have used a cutoff value for T2*of <20 ms14,19 to detect intramyocardial hemorrhage, but a threshold-based
Downloaded from http://ahajournals.org by on March 3, 2020

method of 2 SDs below the mean remote myocardial T2* can also be used.20 In centers where T2* imaging is not available,
many studies have also used T2-weighted imaging, but its diagnostic performance is less robust than T2*-weighted imaging.21
Area at risk This refers to the territory supplied by the infarct-related artery and includes both the reversibly-injured myocardial (the
salvaged myocardium) and the infarcted myocardium. The area at risk can be indirectly assessed by CMR with late gadolinium
enhancement imaging (to derive the endocardial surface area),22 T2-weighted imaging,23,24 T2 mapping,25,26 native T1
mapping,26,27 EGE imaging,28,29 precontrast SSFP cine,30 and postcontrast SSFP cine imaging.31,32
T1 mapping and T2 mapping are currently considered the most robust of the CMR techniques because they have a better
contrast-to-noise ratio than the T2-weighted/T1-weighted/cine imaging techniques, they are less prone to blood pool and
motion artifacts, and they are less likely to have signal dropout resulting from surface coil inhomogeneities.25,27,33
Myocardial salvage and Myocardial salvage can be calculated by subtracting the MI size from the area at risk.
myocardial salvage index
The myocardial salvage index refers to the ratio of the myocardial salvage to the area at risk.34

Remote myocardium This is usually defined as a myocardial segment 180° from the infarcted territory.35

CMR indicates cardiovascular magnetic resonance; ECV, extracellular volume fraction; EGE, early gadolinium enhancement; MI, myocardial infarction; SSFP, steady-
state free precession; and STEMI, ST-segment–elevation myocardial infarction.

as well as the role of MI size, intramyocardial hemor- hancement imaging. Bulluck and colleagues41 recently
rhage, residual myocardial iron, and the remote myo- showed that postcontrast T1 mapping can accurately
cardium in the development of adverse LV remodeling. quantify acute MI size in a small group of patients with
Finally, we explore the prognostic significance of these STEMI, obviating the need to perform late gadolinium
CMR-derived indexes in reperfused patients with STEMI. enhancement imaging. Recent studies have also investi-
gated whether native T1 mapping (precontrast) can as-
T1 Mapping for the Quantification of MI sess MI size. Kali and colleagues42 showed that chronic
Size MI (median of 13.6 years after MI) could be detected
Postcontrast T1 mapping has recently emerged as a with native T1 mapping at 3 T in a small cohort of 25
promising technique for the quantification of MI size patients. Liu and colleagues43 showed that native T1
as an alternative to conventional late gadolinium en- mapping at 3 T could also identify acute MI size. How-

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1951


Bulluck et al The Role of CMR in STEMI
STATE OF THE ART
Downloaded from http://ahajournals.org by on March 3, 2020

Figure 1. A, Schematic representation of the different components of the myocardium in a patient with reper-
fused ST-segment–elevation myocardial infarction (STEMI) and (B) methods for delineating the area at risk, MI
size, microvascular obstruction, and intramyocardial hemorrhage by CMR.
A, The various components of the myocardium in a reperfused patient with STEMI that can be assessed by cardiovas-
cular magnetic resonance (CMR), namely the area at risk, salvaged myocardium, the area of hyperenhancement, and
the hypointense core on late gadolinium enhancement (LGE) images and the remote myocardium. The MI size includes
both the areas of hyperenhancement and the hypointense core on LGE images. The hypointense core on LGE represents
microvascular obstruction (MVO), and a proportion of patients would also have intramyocardial hemorrhage (IMH) within
these areas (areas of low T2* on T2*-weighted images or mapping). The area at risk is a combination of both the MI size
and the salvaged myocardium. B, The CMR-based methods and their timing during CMR acquisition for delineating the
area at risk, MI size, MVO (early and late), and IMH. The most robust method for demarcating the area at risk currently
is T1 and T2 mapping. LGE is the gold standard for detecting MI size and late MVO. T2* mapping is currently the gold
standard to detect IMH. The table provides an indication of the abilities of each sequence to detect each components of
an MI by STEMI graded as follows:  indicates robust, ie, shown by several studies; , possible, ie, shown by some
studies; and , theoretically possible and/or shown by 1 to 2 studies. EGE indicates early gadolinium enhancement; and
ESA, endocardial surface area.

ever, only 58 short-axis T1 maps without microvascular ed. Garg and colleagues44 showed that an ECV value
obstruction were analyzed in that study.43 These find- of ≥0.46 could predict chronic MI size from the acute
ings are of great interest because one could potentially ECV maps. However, Bland-Altman analysis showed a
perform a comprehensive CMR study in STEMI without bias of 1.9% and wide limits of agreement of ±10.5%.
the need of contrast agent, and this approach would Therefore, further studies are needed to build on these
significantly shorten the scan time and make CMR findings.45 However, as it stands, ECV mapping may
available to a wider range of patients. complement late gadolinium enhancement imaging to
Whether ECV on an acute CMR scan can estimate assess MI size and to predict wall motion recovery at
final MI size after STEMI has recently been investigat- follow-up, as recently shown by 2 small studies.16,46

1952 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693


Bulluck et al The Role of CMR in STEMI

STATE OF THE ART


Figure 2. Infarct core of a patient with microvascular obstruction and intramyocardial hemorrhage.
This figure shows an example of a patient with an acute anterior ST-segment–elevation myocardial infarction treated by pri-
mary percutaneous coronary intervention. The cardiovascular magnetic resonance images on day 4 showing the infarct core on
the late gadolinium enhancement (LGE) images represent microvascular obstruction, and the T2*, T1, and T2 maps represent
intramyocardial hemorrhage (arrows).

Despite the promises of T1 mapping in STEMI for and intramyocardial hemorrhage in ≈35% to 40% of
measuring MI size, the published studies have been reperfused patients with STEMI. However, the detec-
small and from single-centers. Larger studies are there- tion of microvascular obstruction and intramyocardial
fore required to confirm these findings using different hemorrhage is dependent on the CMR techniques, the
field strengths, mapping sequences, and vendors be- timing of imaging, and the definitions used.
fore they can be more widely adopted.

Acute MI Size and Subsequent Adverse


Relationship Between Microvascular LV Remodeling
Obstruction and Intramyocardial Conventional theory assumes that the larger the MI, the
Hemorrhage higher the LV wall stress, and the LV therefore dilates to
Downloaded from http://ahajournals.org by on March 3, 2020

Serial CMR imaging of patients with STEMI in the first maintain the stroke volume as a compensatory mecha-
few days after PPCI has provided evidence that the in- nism (via the Frank-Starling principle). However, LV dila-
cidence and extent of microvascular obstruction and tion, as described by the Laplace relationship, leads to
intramyocardial hemorrhage vary with time. The extent further wall stress and begets more LV dilatation in the
of late microvascular obstruction has been reported to absence of compensatory LV hypertrophy. Westman and
peak at 4 to 12 hours, to remain stable the first 2 days, colleagues50 have recently proposed that there is an im-
and to reduce in size by day 10 after STEMI.14 Further- perfect association between MI size and adverse LV re-
more, in those patients with microvascular obstruction modeling. They showed in 122 patients with STEMI that
persisting at 1 week, Ørn and colleagues47 showed that 15% of those with an MI <18.5% developed adverse LV
they were more likely to develop adverse LV remodeling remodeling and that 40% of those with an MI ≥18.5%
compared with those with microvascular obstruction at developed adverse LV remodeling. However, Westman
day 2 only. On the other hand, the detection of intra- and colleagues50 used a definition of >10-mL/m2 increase
myocardial hemorrhage has been shown to peak at day in indexed LV end-diastolic volume in their study. A more
3 and to reduce in incidence by day 10 after PPCI.12 appropriate definition for adverse LV remodeling after
Hamirani and colleagues39 showed that intramyo- STEMI by CMR may be a cutoff value of a 12% change
cardial hemorrhage (detected by either T2-weighted in LV end-diastolic volume.51 This study also showed an
imaging or T2* imaging) was more strongly associated imperfect association between acute MI size and adverse
with late microvascular obstruction (R=0.89–0.93) than LV remodeling and between the presence of microvas-
with early microvascular obstruction (R=0.30). Using cular obstruction and adverse LV remodeling.51 Some
the more robust method of T2* mapping for the detec- patients with a large MI and microvascular obstruction
tion of intramyocardial hemorrhage, Carrick and col- developed reverse LV remodeling, whereas other pa-
leagues14 showed in 286 patients that all patients with tients with a small MI and no microvascular obstruction
intramyocardial hemorrhage also had late microvascu- also developed adverse LV remodeling.51 As alluded to by
lar obstruction. Westman and colleagues,50 the development of adverse
From these recent studies,14,39,48,49 it appears that LV remodeling is complex and multifactorial, and an ex-
early microvascular obstruction occurs in ≈60% to cessive inflammatory response together with MI size may
65%, late microvascular obstruction in ≈50% to 55%, play an important role after STEMI.

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1953


Bulluck et al The Role of CMR in STEMI
STATE OF THE ART

Figure 3. Delineation of the edema-based area at risk by T1 and T2 mapping and myocardial infarction (MI)
size by late gadolinium enhancement (LGE) that can be used to calculate myocardial salvage and myocardial
salvage index.
Top, An example of the edema-based area at risk by T1 and T2 mapping (indicated by black arrows) and the corresponding
Downloaded from http://ahajournals.org by on March 3, 2020

MI size on LGE (red arrow) in a patient with an inferior ST-segment–elevation MI treated by primary percutaneous coronary
intervention, with the cardiovascular magnetic resonance (CMR) images acquired on day 3 after reperfusion. Bottom, The
corresponding postprocess images using a threshold-based method to delineate the area at risk (threshold of 2 SDs) and MI
size (threshold of 5 SDs) as the highlighted areas with the areas of hypointense core included as part of the area at risk and MI
size. The myocardial salvage (area at risk subtract the MI size) and myocardial salvage index (myocardial salvage as a ratio of
the area at risk) can be calculated retrospectively from a single CMR scan.

Residual Myocardial Iron and Adverse LV In a small cohort of patients with STEMI, Bulluck and
Remodeling colleagues15 recently found that intramyocardial hem-
orrhage and subsequent residual myocardial iron at
There are emerging data that intramyocardial hemor- follow-up were associated with persistently elevated T2
rhage at the time of PPCI leads to residual myocardial values in the surrounding infarct tissue and with adverse
iron during the convalescent phase, and it may be a LV remodeling.15 Carberry and colleagues55 have recent-
source of prolonged inflammation and have an im- ly confirmed these findings in a cohort of 203 patients
pact on adverse LV remodeling.52 Kali and colleagues52 with STEMI. They showed that 36% of their cohort had
showed in a small cohort of 15 patients with STEMI intramyocardial hemorrhage by T2* mapping and 59%
and in 20 canines (with histological validation) that of those had residual myocardial iron at 6 months. Re-
intramyocardial hemorrhage resulted in residual myo- sidual myocardial iron was associated with adverse LV
cardial iron within the MI zone, and this provided a remodeling at 6 months and worse clinical outcomes
source of prolonged inflammatory burden in the chron- after a median follow-up of 4 years.55 Figure 4A shows
ic phase. These findings were recently confirmed in a an example of a patient with an acute inferior STEMI
canine model of MI, and the extent of residual iron was treated by PPCI and with late microvascular obstruction
strongly correlated with markers of inflammation and and intramyocardial hemorrhage despite restoration
adverse LV remodeling.53 Roghi and colleagues54 found of normal flow in the epicardial coronary artery dur-
higher levels of non–transferrin-bound iron in 7 of 15 ing PPCI. At follow-up, there was residual myocardial
patients with STEMI with microvascular obstruction and iron and persistently elevated T2 values in the areas sur-
postulated that intramyocardial hemorrhage could be a rounding the residual iron, which may represent persis-
source of cardiotoxicity in these patients. tent myocardial inflammation.

1954 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693


Bulluck et al The Role of CMR in STEMI

STATE OF THE ART


Figure 4. A, Paired acute and follow-up T2* and T2 maps and (B) paired acute and follow-up automated extracellu-
lar volume fraction (ECV) maps of a patient with an inferior ST-segment–elevation myocardial infarction (STEMI).
Shown is an example of the paired acute and follow-up (6 months) late gadolinium enhancement (LGE) and automated ECV
maps and T2* and T2 maps of a patient with an inferior STEMI, treated by primary percutaneous coronary intervention with
restoration of normal flow in the infarct-related epicardial coronary artery. The arrows show the area of microvascular obstruc-
tion with intramyocardial hemorrhage. The corresponding ECV maps show the area of microvascular obstruction where con-
trast fails to penetrate the areas of microvascular obstruction and appears as pseudonormal myocardium on the acute scan. At
follow-up, the microvascular obstruction has resolved, and the space previously occupied by the microvascular obstruction has
very high ECV (white areas within the MI zone corresponding to the upper limit of the look-up color scale). The white regions
of interest on the ECV maps (black arrows) are representative areas of the remote myocardium that can be used to assess
changes in the extracellular matrix. There were residual myocardial iron at follow-up and persistently elevated T2 in the areas
surrounding the residual iron (blue arrows on the follow-up images).
Downloaded from http://ahajournals.org by on March 3, 2020

The Role of the Remote Myocardium and verse events remains to be tested in future, adequately
Clinical Outcomes powered studies. Figure 4B shows an example of the
paired acute and follow-up (6 months) late gadolinium
Whether changes in the extracellular matrix in the re- enhancement and automated ECV maps with represen-
mote myocardium (Table) in patients with STEMI treated tative regions of interest of the remote myocardium of a
by PPCI are associated with adverse LV remodeling has patient with an anterior STEMI treated by PPCI.
been a topic of ongoing research.16,17,56–58 Using auto- Carrick and colleagues57 demonstrated that higher
mated ECV maps, Bulluck and colleagues16 have shown native T1 of the remote myocardium of 267 reperfused
in a small cohort of 40 patients with STEMI that ECV in patients with STEMI on the acute CMR scan was associ-
the remote myocardium was acutely elevated and that ated with changes in LV end-diastolic volume from base-
this elevation in ECV persisted in those who developed line to 6 months, and it was independently associated
adverse LV remodeling at 5 months, suggesting that re- with adverse cardiac events after a median follow-up of
mote compensatory changes in the extracellular matrix 845 days. Most recently, in a similar number of patients,
occurred in this subset of patients. The increase in acute Reinstadler and colleagues56 also showed that the re-
ECV could have been due to an increase in the intravas- mote native myocardial T1 was independently associated
cular compartment as a compensatory mechanism. At with adverse events after adjusting for clinical risk factors
follow-up, the increase in ECV likely represented diffused and other CMR variables after a follow-up of 6 months.
interstitial fibrosis as part of the remodeling process. Therefore, T1 mapping has the potential to comple-
Garg and colleagues58 recently confirmed these findings ment clinical and other CMR-derived parameters to im-
in a cohort of 50 patients with STEMI. Furthermore, they prove the risk stratification of patients with STEMI and
also showed that remote segments with ECV expansion warrants further investigation in a multicenter setting.
were associated with impaired wall thickening. In a larg-
er cohort of patients (n=131), Carberry and colleagues17
showed that the change in ECV of the remote myocardi- Acute MI Size and Clinical Outcomes
um was a multivariable associated with the change in LV Morbidity and mortality after STEMI are closely relat-
end-diastolic volume at 6 months. Whether this change ed to acute MI size. A recent meta-analysis of 2632
in ECV could independently predict those at risk of ad- patients showed that MI size measured by CMR or

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1955


Bulluck et al The Role of CMR in STEMI

single-photon emission computed tomography with- Therefore, on the basis of the current evidence dis-
in a month after PPCI from 10 randomized controlled cussed so far, the prognosis worsens with a larger MI.
STATE OF THE ART

trials was strongly associated with 1-year hospitaliza- Patients with a larger MI are also more likely to have
tion for heart failure and all-cause mortality.59 For ev- microvascular obstruction and intramyocardial hemor-
ery 5% increase in MI size, there was a 20% increase rhage. The prognosis is worse for patients with STEMI
in the relative hazard ratio for 1-year hospitalization with microvascular obstruction compared with those
for heart failure and all-cause mortality. The event without microvascular obstruction61 and is worst for
rate was 1.2% in those with an MI ≤8% of the LV, those with microvascular obstruction and intramyocar-
and there was a stepwise increase to 2.5% in those dial hemorrhage.14
with an MI >8% to ≤17.9% of the LV, 5.6% in those Currently, there are no effective methods to detect
with an MI >17.9% to 29.8% of the LV, and 8.8% those at risk of microvascular obstruction and intra-
in those with an MI >29.8% of the LV. This study myocardial hemorrhage at the time of PPCI. Amier and
adds to the growing body of evidence that acute MI colleagues62 recently showed that anterior STEMI and
size is prognostic and therefore remains a valid surro- the use of glycoprotein IIb/IIIa inhibitors were associ-
gate end point for clinical trials. However, this study ated with the development of intramyocardial hemor-
also highlights the fact that one of the limitations rhage. However, this was a retrospective post hoc anal-
of using CMR in randomized controlled trials is that ysis, and intramyocardial hemorrhage was identified
there is an element of selection bias because only with T2-weighted imaging instead of the more robust
those patients fit enough to tolerate a CMR study T2*-weighted imaging, and their findings need to be
would enter these randomized controlled trials, and confirmed in future studies. Furthermore, there are no
the overall event rates were 2.2% and 2.6% for all- established therapies to prevent or minimize the bur-
cause mortality and hospitalization for heart failure den of microvascular obstruction and intramyocardial
at 1 year, respectively, much lower than those report- hemorrhage in the clinical setting,4 although promising
ed from clinical registries.2,3 results are emerging in the preclinical setting.63

Microvascular Obstruction, Intramyocardial


Hemorrhage, and Clinical Outcomes CURRENT CONTROVERSIES IN CMR
IMAGING OF PATIENTS WITH STEMI
Downloaded from http://ahajournals.org by on March 3, 2020

Both microvascular obstruction and intramyocardial


hemorrhage are associated with larger MI size, adverse Is Myocardial Edema Confined to the
LV remodeling, and worse clinical outcomes.39,48 In a Infarct Zone, or Does It Extend Into the
meta-analysis of >1000 patients, Van Kranenburg and
Salvaged Myocardium?
colleagues48 showed that the presence of microvascu-
lar obstruction was an independent predictor of ma- In the 1980s, Higgins and colleagues64 were the first
jor adverse clinical outcome at 2 years in patients with to study myocardial T2 CMR in a canine model of MI.
STEMI, whereas MI size was not independently associ- Both T2 and T1 were shown to change similarly in the
ated with adverse events. There was a graded reduc- setting of an acute MI. The observed changes were
tion in event-free survival in patients with an MI <25% theoretically consistent with myocardial edema and
of the LV with and without microvascular obstruction correlated with the measurements of myocardial water
and those with an MI >25% of the LV with and without content estimated by wet weight to dry weight ratios.
microvascular obstruction. The prognostic value of mi- The changes in myocardial T2 have been attributed to
crovascular obstruction over MI size for mortality and a combination of an increase in absolute tissue water,
hospitalization for heart failure was recently confirmed the movement of water from the extracellular to the in-
by de Waha and colleagues49 in a pooled analysis of tracellular compartment, and the conversion of protein-
patient-level data from 7 randomized controlled trials bound water to free water.65
(n=1688) at 1 year and by Symons and colleagues60 Not all experimental and clinical studies have sup-
in a longitudinal study of 810 patients after a median ported the concept that the region of hyperintensity on
follow-up of 5.5 years. T2-weighted images delineates the area at risk. Aletras
Early studies showed that intramyocardial hemor- and colleagues25 and Tilak and colleagues66 validated
rhage, detected by T2-weighted images, was closely the edema-based area at risk using T2-weighted im-
related to the development of adverse LV remodeling ages against microspheres in reperfused and nonre-
and worse clinical outcomes.39 Most recently, using perfused canine MI. Ubachs and colleagues67 showed
T2* mapping, Carrick and colleagues14 showed that that the area at risk derived from T2-weighted imaging
intramyocardial hemorrhage was more closely associ- matched that obtained from single-photon emission
ated with adverse clinical outcomes than microvascular computed tomography in animals both with and with-
obstruction. out late gadolinium enhancement.

1956 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693


Bulluck et al The Role of CMR in STEMI

In contrast, Kim and colleagues68 reported that T2- myocardial edema within the first few days of an MI
weighted imaging did not depict the area at risk in is dynamic.

STATE OF THE ART


anesthetized dogs that were subjected to different du- Fernández-Jiménez and colleagues73 studied the in-
rations of coronary occlusion. MI size by late gadolinium terplay between myocardial water content (using desic-
enhancement was compared with the gold standard cation) and T2 relaxation times in a porcine model of
method of triphenyltetrazolium chloride staining, and reperfused MI. They reported a “bimodal” pattern of
area at risk by T2-weighted imaging was compared with edema, appearing at 2 hours, resolving by 24 hours,
the gold standard method of fluorescent microspheres and then reappearing on day 4 and peaking at day 7.
4 days after reperfusion. The T2-weighted hyperinten- The first wave of edema was attributed to myocardial
sity correlated and matched in shape better with the reperfusion injury, and the second wave of edema was
MI size than with the area at risk, and these findings attributed to inflammation during the tissue-healing
were confirmed in a small group of patients with acute phase.74 However, they did not take into account the
MI. Although the findings from the canine model of MI presence of microvascular obstruction and intramyocar-
may have resulted from the presence of well-developed dial hemorrhage, which may have interfered with their
collaterals in that species, the T2-weighted sequence in T2 measurements, and they had a small sample size of
that study was different from that used so far by other 5 pigs at each time point. Furthermore, the same ani-
studies.23,66 In the clinical setting, using hybrid positron mals were not serially scanned. Most recently, Fernán-
emission tomography/CMR imaging in reperfused pa- dez-Jiménez and colleagues13 showed that the bimodal
tients with STEMI, Bulluck and colleagues69 recently edematous response of the infarct-related territory also
showed that the area of reduced 18F-fluorodeoxyglu- was present in humans. However, they used the less
cose uptake was significantly larger than the MI size robust T2-weighted imaging modality instead for both
and closely matched the area at risk delineated by T2 quantifying the extent of the edema-based area at risk
mapping on the acute scan, supporting the notion that and for detecting intramyocardial hemorrhage. It was
T2-mapping delineates both the reversibly and the irre- not clear how many of their 14 patients had intramyo-
versibly injured myocardium within the area at risk.69 In cardial hemorrhage in that cohort. Furthermore, on a
addition, the areas of reduced 18F-fluorodeoxyglucose patient-level basis (their supplementary material Figures
uptake within the salvaged myocardium in the area at 3 and 4), not all patients displayed a bimodal response
risk were no longer present on a follow-up scan several for the intensity and extent of edema in the infarct-
months later.69 Hammer-Hansen and colleagues70 have related territory.
Downloaded from http://ahajournals.org by on March 3, 2020

provided further insights into this topic using a canine In contrast to these studies, in a cohort of 30 patients
model of MI. They showed that T2 values were elevat- with serial imaging, Carrick and colleagues12 found that
ed in both the MI zone and salvaged myocardium, and the extent of myocardial edema was maximal at day 3
they were both significantly higher than the T2 values and decreased by day 10 after PPCI, suggesting a uni-
in the remote myocardium. modal peak in myocardial edema in the first few days
Therefore, the majority of the current literature sup- after reperfusion. In patients with STEMI with intramyo-
ports the notion that edema, assessed by CMR with T2 cardial hemorrhage, they observed a bimodal pattern
mapping, occurs in both the irreversibly and reversibly in T2 and T2* values within the MI core, whereas in
injured myocardium during the first week of a reper- patients without intramyocardial hemorrhage, only a
fused STEMI in the clinical setting.25,27,69,71 unimodal pattern in T2 and T2* values was observed,
suggesting that the presence of intramyocardial hem-
Dynamic Changes in the Edema-Based orrhage may have been responsible for the apparent
bimodal edema pattern in T2 and T2* values.
Area at Risk During the First Week in Nordlund and colleagues31 recently provided some
Reperfused Patients With STEMI evidence that there was no bimodal pattern of edema
It was initially believed that edema is stable in the first in the clinical setting. They combined data from 3 stud-
week of an MI.71,72 However, both of these studies per- ies involving 215 patients and showed that there were
formed T2-weighted imaging only on days 1 and 7, no differences in size, quality of the T2-weighted im-
and the peak in the edema-based area at risk shown ages, and the ability to detect the culprit territory when
by Carrick and colleagues12 on day 3 was therefore patients having a CMR study on day 1 to 6 onward
not identified by the previous 2 studies.71 Emerging were compared.
evidence suggests that the extent of myocardial ede- Given the dynamic changes in the extent of edema
ma is dynamic. Hammer-Hansen and colleagues70 have and acute MI size over the first few days after STEMI, it
shown in a canine model of reperfused MI that at 2 is important that future clinical cardioprotection studies
hours T2 values were higher in both the infarcted and define the time window for performing the acute CMR
salvaged myocardium compared with those who had scan to optimize the quantification of both acute MI
the scan at 48 hours, highlighting that the extent of size and area at risk. There is currently no consensus on

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1957


Bulluck et al The Role of CMR in STEMI

the optimal timing for performing the acute CMR scan, phenomenon in a porcine model of acute MI. At day
although 3 to 5 days6 and 4 to 7 days13 after PPCI have 1, they found higher ECV values in the peri-infarct zone
STATE OF THE ART

been proposed. that could have been the result of severe edema or a
mixture of infarcted and salvaged myocardium in that
area that contributed to the overestimation of MI size.
Cardioprotective Therapies and the By 7 days, the ECV values in the peri-infarct zone de-
Edema-Based Area at Risk in Reperfused creased to the same level as the rest of the salvaged
Patients With STEMI myocardium, such that MI size assessed by late gado-
The utility of T2-weighted imaging for quantifying the linium enhancement matched that by histology.
area at risk has been called into question because cer- Acquiring late gadolinium enhancement too early
tain cardioprotective therapies have been shown to (<8 minutes after contrast administration) can result
reduce not only MI size but also the extent of myocar- in an overestimation of MI size,82 and acquiring late
dial edema measured by T2-weighted and T2-mapping gadolinium enhancement at 25 minutes for acute MI
imaging. Ischemic postconditioning75 and remote isch- size has been shown to be a better predictor of LV re-
emic conditioning using transient arm or leg ischemia covery.83 However, waiting 25 minutes after contrast
and reperfusion76 have been shown to reduce both MI administration may be difficult in the clinical setting be-
size and the extent of edema delineated by T2-mapping cause of time constraints and would affect workflow.
and T2-weighted imaging, leading to an underestima- Therefore, late gadolinium enhancement images are
tion of the area at risk by these techniques. However, currently acquired 10 to 15 minutes after contrast6 as
these were small studies of reperfused patients with a compromise.
STEMI, and they were not adequately powered to as- Several studies have already shown a significant re-
sess that end point. Intuitively, if a cardioprotective gression in MI size (by 30%–35%) occurring between
therapy can reduce MI size, it should be expected to the acute CMR scan and the chronic phase.47,84,85 Sever-
al factors are responsible for this observation. First, the
also limit the extent of myocardial edema, which itself is
compositions of the different tissues being identified
the result of myocardial ischemia and reperfusion injury.
by late gadolinium enhancement in the acute (myocar-
However, in a recently published large study of 696
dial necrosis compounded with edema, intramyocar-
patients with STEMI by Eitel and colleagues,77 ischemic
dial hemorrhage, and microvascular obstruction) and
postconditioning or a combination of remote ischemic
Downloaded from http://ahajournals.org by on March 3, 2020

chronic (focal replacement fibrosis) phase are different.


conditioning and ischemic postconditioning did not re-
The regression of MI size between the acute, subacute
duce the extent of myocardial edema compared with
and chronic phases therefore represents the gradual
the control arm. Furthermore, drugs such as metopro-
resolution of edema, intramyocardial hemorrhage, and
lol78 and exenatide79 that were effective at increasing
microvascular obstruction and the gradual replacement
the myocardial salvage index did not reduce the ex-
of the necrotic cardiomyocytes with focal scars.71,85
tent of myocardial edema. One potential explanation
As it stands, the optimal timing of CMR for MI size is
for this discrepancy could be that those therapies that
not well established, but late gadolinium enhancement
are potent enough to reduce MI size in reperfused pa-
appears to stabilize by 10 days after the index event.12
tients with STEMI are capable of reducing the extent of
In most centers, patients with STEMI with an uncom-
myocardial edema. In contrast, those therapies that are
plicated inpatient stay are discharged within the first
less potent, do not reduce MI size, and only increase
week. Logistically, this would mean that patients would
myocardial salvage do not affect the extent of myocar-
have to come back for the CMR scan, and this may lead
dial edema. Either way, these findings may bring into
to a proportion of patients dropping out.6 Efforts are
question the use of edema-based area at risk measured
underway to attempt to standardize the acquisition of
by CMR to assess myocardial salvage in clinical cardio-
CMR for clinical cardioprotection studies.6
protection studies.

The Optimal Timing of Late Gadolinium FUTURE DIRECTIONS


Enhancement Imaging for Acute and The Need to Standardize the Use of CMR
Chronic MI Size Quantification in Reperfused Patients With STEMI
Acute MI size has been shown to be dynamic and to Both acute and chronic MI sizes by CMR have been
decrease significantly in size between days 1 and 7 after shown to be stronger predictors of clinical outcomes
STEMI.80 In a recent study by Carrick and colleagues,12 compared with LV ejection fraction or LV volumes.86,87
MI size was shown to be similar between days 1 and 3 Therefore, because of the robustness and high repro-
and reduced by day 10. Jablonowski and colleagues81 ducibility of CMR to quantify MI size,10,36,86–88 both
recently provided some mechanistic insights into this acute MI size and chronic MI size are increasingly being

1958 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693


Bulluck et al The Role of CMR in STEMI

used as surrogate end points for randomized controlled such as T1 mapping19 and T2* mapping14 to provide ad-
trials assessing the effectiveness of new cardioprotec- equate prognostic information over postcontrast imag-

STATE OF THE ART


tive therapies and can reduce the sample size required.6 ing modalities and to be applied to all patients, regard-
However, acute MI size is influenced by a number of less of renal function, warrants further investigation.
factors, including the timing of the CMR scan, the dose
of contrast used, the time elapsed after contrast admin-
istration and acquisition of late gadolinium enhance-
CMR as a Tool to Risk Stratify and
ment imaging, and the method used for the quantifica- Guide Management in Patients With
tion of MI size.6,89 Therefore, there is an urgent need STEMI at Risk of Developing Adverse LV
for the CMR community to come up with a consensus Remodeling and Subsequent Heart Failure
statement to standardize the acquisition of late gado- A number of cardioprotective therapies have had prom-
linium enhancement imaging and the quantification of ising results in the experimental setting for reducing MI
MI size in future studies.6 This would no doubt help to size and preventing adverse LV remodeling but have
strengthen the robustness of this technique and would failed to be translated into the clinical setting.93 A more
facilitate prospective or retrospective collaborative re- targeted approach using CMR to identify those most at
search with merging of CMR data from different cen- risk (eg, according to their acute MI size or according to
ters around the world. the presence or absence of microvascular obstruction)
may improve the translation of those promising experi-
mental results in the clinical setting to prevent adverse
Toward Improving the Tolerability and
LV remodeling and reduce the onset of heart failure in
Accessibility of CMR in Reperfused these patients. Stiermaier and colleagues94 and Pon-
Patients With STEMI tone and colleagues95 most recently proposed CMR risk
There have already been some advances in free-breath- scores to identify those at high risk of adverse events.
ing and motion-corrected T290 and T2* mapping91 and Their approach would be ideal to identify high-risk pa-
late gadolinium enhancement imaging,92 negating the tients, but the CMR indexes and cutoff values included
need for breath-holding and helping to accelerate the in each study differed, and further validation work is
acquisition of these sequences. Other developments in required to build on their findings. Furthermore, CMR
real-time cine imaging for LV ejection fraction and fu- could identify those with intramyocardial hemorrhage,
Downloaded from http://ahajournals.org by on March 3, 2020

ture refinements in CMR fingerprinting techniques (for and they could be targeted with anti-inflammatory
robust and fast acquisition of simultaneous T1 and T2 agents or chelation therapy15,96 to minimize the cardio-
mapping data per slice within 1 breath-hold) will fur- toxic effects of the residual iron (which has been shown
ther reduce scan time and will be highly desirable in the to occur in two third of patients with intramyocardial
acute STEMI setting. These developments would make hemorrhage from a pooled analysis of 4 studies)55 dur-
CMR, which is currently confined to those fit enough ing the convalescent phase of an acute STEMI. This
to lie inside the scanner for at least 30 to 45 minutes, warrants further investigation.
accessible to a wider number of patients. Furthermore,
reducing the duration of the scan to 20 to 30 minutes CMR as a Tool for Risk Stratification
could also potentially reduce the cost of a scan and
improve the participation of more centers in STEMI-
of Patients With STEMI at Risk of
related CMR research. However, most of these new de- Arrhythmic Events
velopments are still in the validation phase and are not The current guidelines recommend an implantable car-
available to all CMR vendors and platforms. dioverter-defibrillator for primary prevention in those
The use of gadolinium chelates is not recom- with symptomatic heart failure and LV ejection frac-
mended in those with a glomerular filtration rate <30 tion <35% 40 days after the index event.97 However,
mL·min−1·1.73 −1m−2, to minimize the risk of nephrogen- mortality within the first 30 days after MI has been
ic systemic fibrosis, and constitutes a contraindication shown to be the highest98 but is believed to be pre-
in CMR-based clinical cardioprotection studies. Further- dominantly the result of MI or myocardial rupture.99
more, the US Food and Drug Administration has recent- Despite that, a quarter of those deaths are still caused
ly issued a new warning about gadolinium remaining in by ventricular arrhythmias,99 and there currently is no
a patient’s brainstem for months to years after receiv- established tool to risk stratify these patients in the
ing these drugs. However, gadolinium retention has not acute phase of an MI. Acute MI size (cutoff of 23.5
been directly linked to adverse events in patients with g/m2 or 31% of the LV) in combination with LV ejec-
normal renal function, and the current benefits of us- tion fraction (cutoff of 36%) by CMR within the first
ing gadolinium chelates outweigh any potential risks. week after an MI has been shown to predict adverse
Therefore, the potential for noncontrast CMR imaging arrhythmic cardiac events at 2 years.100 Other studies

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1959


Bulluck et al The Role of CMR in STEMI

have shown that CMR in the chronic phase could also Sources of Funding
be used to predict sudden cardiac death with the use This work was supported in part by grants from the British Heart Founda-
STATE OF THE ART

of the peri-infarct zone extent101 or chronic MI size102 tion (FS/10/039/28270), National Institute for Health Research University Col-
lege London Hospitals Biomedical Research Center, Duke-National University
in combination with LV ejection fraction. Furthermore, Singapore Medical School, Singapore Ministry of Health’s National Medical
residual myocardial iron as a consequence of intramyo- Research Council under its Clinician Scientist-Senior Investigator scheme
cardial hemorrhage has also been shown to be proar- (NMRC/CSA-SI/0011/2017) and Collaborative Center Grant scheme (NMRC/
CGAug16C006), Singapore Ministry of Education Academic Research Fund Tier
rhythmic.103 Although all of these were relatively small 2 (MOE2016-T2-2–021), and EU-CARDIOPROTECTION CA16225 Cooperation
and single-center studies, CMR has shown promise to in Science and Technology Action to Prof Hausenloy; in part by grants from
improve the risk stratification of patients with STEMI the Intramural Research Program of the National Heart, Lung, and Blood Insti-
tute, National Institutes of Health to Dr Arai; in part by grants from extramural
at risk of ventricular arrhythmias. With the availability funding from the National Heart, Lung, and Blood Institute, National Institutes
of inline, automated ECV mapping104 and free-breath- of Health (R01 HL136578 and R01 HL133407) to Dr Dharmakumar; and in
ing T2* mapping,91 characterization of the infarct size, part by grants from the British Heart Foundation Center of Research Excel-
lence Award (RE/13/5/30177), BHF grants (FS/15/54/31639, PG/11/2/28474,
peri-infarct zone, and infarct core may become more PG/14/64/31043), Medical Research Scotland, and the Chief Scientist Office
objective, and CMR would play an invaluable role in and a Senior Fellowship from the Scottish Funding Council to Dr Berry.
identifying those most likely to benefit from a primary
prevention implantable cardioverter-defibrillator in the Disclosures
near future. None.

CONCLUSIONS REFERENCES
1. Nabel EG, Braunwald E. A tale of coronary artery disease and myocardial
Over the last few years, CMR scans performed in the infarction. N Engl J Med. 2012;366:54–63. doi: 10.1056/NEJMra1112570.
acute phase of reperfused STEMI have improved our 2. Pedersen F, Butrymovich V, Kelbæk H, Wachtell K, Helqvist S, Kastrup J,
Holmvang L, Clemmensen P, Engstrøm T, Grande P, Saunamäki K, Jør-
understanding of the changes occurring in the infarct-
gensen E. Short- and long-term cause of death in patients treated with
ed, salvaged, and remote myocardium and have provid- primary PCI for STEMI. J Am Coll Cardiol. 2014;64:2101–2108. doi:
ed insights into the impact of MI size, microvascular ob- 10.1016/j.jacc.2014.08.037.
3. Ezekowitz JA, Kaul P, Bakal JA, Armstrong PW, Welsh RC, McAlister FA.
struction, and intramyocardial hemorrhage on clinical Declining in-hospital mortality and increasing heart failure incidence
outcomes. There are ongoing controversies surround- in elderly patients with first myocardial infarction. J Am Coll Cardiol.
ing the use of CMR to delineate the area at risk and 2009;53:13–20. doi: 10.1016/j.jacc.2008.08.067.
Downloaded from http://ahajournals.org by on March 3, 2020

4. Bulluck H, Foin N, Tan JW, Low AF, Sezer M, Hausenloy DJ. Invasive assess-
the dynamic changes in myocardial edema in the first ment of the coronary microcirculation in reperfused ST-segment-elevation
few days after PPCI. However, more work is ongoing, myocardial infarction patients: where do we stand? Circ Cardiovasc Interv.
and CMR holds promise to become accessible to more 2017;10:e004373.
5. Bulluck H, Yellon DM, Hausenloy DJ. Reducing myocardial infarct size:
patients and to be used as a tool to risk stratify patients, challenges and future opportunities. Heart. 2016;102:341–348. doi:
to guide treatment, and to improve clinical outcomes 10.1136/heartjnl-2015-307855.
after PPCI in the near future. 6. Bulluck H, Hammond-Haley M, Weinmann S, Martinez-Macias R, Haus-
enloy DJ. Myocardial infarct size by CMR in clinical cardioprotection stud-
ies: insights from randomized controlled trials. JACC Cardiovasc Imaging.
2017;10:230–240. doi: 10.1016/j.jcmg.2017.01.008.
ARTICLE INFORMATION 7. Ibanez B, James S, Agewall S, Antunes MJ, Bucciarelli-Ducci C, Bueno H,
Caforio ALP, Crea F, Goudevenos JA, Halvorsen S, Hindricks G, Kastrati
Correspondence
A, Lenzen MJ, Prescott E, Roffi M, Valgimigli M, Varenhorst C, Vranckx P,
Derek J. Hausenloy, MD, PhD, Cardiovascular & Metabolic Diseases Program, Widimský P; ESC Scientific Document Group. 2017 ESC guidelines for the
Duke–NUS Graduate Medical School Singapore, 8 College Rd, Singapore, Sin- management of acute myocardial infarction in patients presenting with
gapore 169857. E-mail derek.hausenloy@duke-nus.edu.sg ST-segment elevation: the Task Force for the management of acute myo-
cardial infarction in patients presenting with ST-segment elevation of the
European Society of Cardiology (ESC). Eur Heart J. 2018;39:119–177. doi:
Affiliations
10.1093/eurheartj/ehx393.
Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University 8. Schulz-Menger J, Bluemke DA, Bremerich J, Flamm SD, Fogel MA, Friedrich
College London, United Kingdom (H.B., D.J.H.). Royal Papworth Hospital, Cam- MG, Kim RJ, von Knobelsdorff-Brenkenhoff F, Kramer CM, Pennell DJ, Plein
bridge, United Kingdom (H.B.). Biomedical Imaging Research Institute and S, Nagel E. Standardized image interpretation and post processing in car-
Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (R.D.). Depart- diovascular magnetic resonance: Society for Cardiovascular Magnetic Reso-
ment of Medicine, David Geffen School of Medicine, University of California, nance (SCMR) board of trustees task force on standardized post processing.
Los Angeles (R.D.). Laboratory for Advanced Cardiovascular Imaging, National J Cardiovasc Magn Reson. 2013;15:35. doi: 10.1186/1532-429X-15-35.
Heart, Lung, and Blood Institute, National Institutes of Health, Department of 9. Kim HW, Farzaneh-Far A, Kim RJ. Cardiovascular magnetic resonance in
Health and Human Services, Bethesda, MD (A.E.A.). British Heart Foundation patients with myocardial infarction: current and emerging applications. J
Glasgow Cardiovascular Research Center, Institute of Cardiovascular and Medi- Am Coll Cardiol. 2009;55:1–16. doi: 10.1016/j.jacc.2009.06.059.
cal Sciences, University of Glasgow, United Kingdom (C.B.). National Institute 10. Wagner A, Mahrholdt H, Holly TA, Elliott MD, Regenfus M, Parker M,
of Health Research University College London Hospitals Biomedical Research Klocke FJ, Bonow RO, Kim RJ, Judd RM. Contrast-enhanced MRI and
Centre, United Kingdom (D.J.H.). Barts Heart Centre, St. Bartholomew’s Hospi- routine single photon emission computed tomography (SPECT) perfusion
tal, London, United Kingdom (D.J.H.). National Heart Research Institute Singa- imaging for detection of subendocardial myocardial infarcts: an imaging
pore, National Heart Centre Singapore, Singapore (D.J.H.). Cardiovascular and study. Lancet. 2003;361:374–379. doi: 10.1016/S0140-6736(03)12389-6.
Metabolic Disorders Program, Duke–National University of Singapore, Singa- 11. Messroghli DR, Moon JC, Ferreira VM, Grosse-Wortmann L, He T, Kell-
pore (D.J.H.). Yong Loo Lin School of Medicine, National University Singapore, man P, Mascherbauer J, Nezafat R, Salerno M, Schelbert EB, Taylor AJ,
Singapore (D.J.H.). Thompson R, Ugander M, van Heeswijk RB, Friedrich MG. Clinical recom-

1960 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693


Bulluck et al The Role of CMR in STEMI

mendations for cardiovascular magnetic resonance mapping of T1, T2, 25. Verhaert D, Thavendiranathan P, Giri S, Mihai G, Rajagopalan S, Sim-
T2* and extracellular volume: a consensus statement by the Society for onetti OP, Raman SV. Direct T2 quantification of myocardial edema in

STATE OF THE ART


Cardiovascular Magnetic Resonance (SCMR) endorsed by the European acute ischemic injury. JACC Cardiovasc Imaging. 2011;4:269–278. doi:
Association for Cardiovascular Imaging (EACVI). J Cardiovasc Magn Re- 10.1016/j.jcmg.2010.09.023.
son. 2017;19:75. doi: 10.1186/s12968-017-0389-8. 26. Ugander M, Bagi PS, Oki AJ, Chen B, Hsu LY, Aletras AH, Shah S,
12. Carrick D, Haig C, Ahmed N, Rauhalammi S, Clerfond G, Carberry J, Mor- Greiser A, Kellman P, Arai AE. Myocardial edema as detected by pre-
di I, McEntegart M, Petrie MC, Eteiba H, Hood S, Watkins S, Lindsay MM, contrast T1 and T2 CMR delineates area at risk associated with acute
Mahrous A, Welsh P, Sattar N, Ford I, Oldroyd KG, Radjenovic A, Berry C. myocardial infarction. JACC Cardiovasc Imaging. 2012;5:596–603. doi:
Temporal evolution of myocardial hemorrhage and edema in patients af- 10.1016/j.jcmg.2012.01.016.
ter acute ST-segment elevation myocardial infarction: pathophysiological 27. Bulluck H, White SK, Rosmini S, Bhuva A, Treibel TA, Fontana M, Abdel-
insights and clinical implications. J Am Heart Assoc. 2016;5:e002834. Gadir A, Herrey A, Manisty C, Wan SM, Groves A, Menezes L, Moon
13. Fernández-Jiménez R, Barreiro-Pérez M, Martin-García A, Sánchez- JC, Hausenloy DJ. T1 mapping and T2 mapping at 3T for quantifying
González J, Agüero J, Galán-Arriola C, García-Prieto J, Díaz-Pelaez E, Vara the area at risk in reperfused STEMI patients. J Cardiovasc Magn Reson.
P, Martinez I, Zamarro I, Garde B, Sanz J, Fuster V, Sánchez PL, Ibanez B. Dy- 2015;17:73. doi: 10.1186/s12968-015-0173-6.
namic edematous response of the human heart to myocardial infarction: 28. Matsumoto H, Matsuda T, Miyamoto K, Shimada T, Mikuri M, Hiraoka
implications for assessing myocardial area at risk and salvage. Circulation. Y. Peri-infarct zone on early contrast-enhanced CMR imaging in patients
2017;136:1288–1300. doi: 10.1161/CIRCULATIONAHA.116.025582. with acute myocardial infarction. JACC Cardiovasc Imaging. 2011;4:610–
14. Carrick D, Haig C, Ahmed N, McEntegart M, Petrie MC, Eteiba H, 618. doi: 10.1016/j.jcmg.2011.03.015.
Hood S, Watkins S, Lindsay MM, Davie A, Mahrous A, Mordi I, Rau- 29. Hammer-Hansen S, Leung SW, Hsu LY, Wilson JR, Taylor J, Greve AM,
halammi S, Sattar N, Welsh P, Radjenovic A, Ford I, Oldroyd KG, Berry Thune JJ, Køber L, Kellman P, Arai AE. Early gadolinium enhancement for
C. Myocardial hemorrhage after acute reperfused ST-segment-elevation determination of area at risk: a preclinical validation study. JACC Cardio-
myocardial infarction: relation to microvascular obstruction and prog- vasc Imaging. 2017;10:130–139. doi: 10.1016/j.jcmg.2016.04.009.
nostic significance. Circ Cardiovasc Imaging. 2016;9:e004148. doi: 30. Kumar A, Beohar N, Arumana JM, Larose E, Li D, Friedrich MG, Dhar-
10.1161/CIRCIMAGING.115.004148. makumar R. CMR imaging of edema in myocardial infarction using
15. Bulluck H, Rosmini S, Abdel-Gadir A, White SK, Bhuva AN, Treibel TA, cine balanced steady-state free precession. JACC Cardiovasc Imaging.
Fontana M, Ramlall M, Hamarneh A, Sirker A, Herrey AS, Manisty C, 2011;4:1265–1273. doi: 10.1016/j.jcmg.2011.04.024.
Yellon DM, Kellman P, Moon JC, Hausenloy DJ. Residual myocardial iron 31. Nordlund D, Klug G, Heiberg E, Koul S, Larsen TH, Hoffmann P, Met-
following intramyocardial hemorrhage during the convalescent phase of zler B, Erlinge D, Atar D, Aletras AH, Carlsson M, Engblom H, Arheden
reperfused ST-segment-elevation myocardial infarction and adverse left H. Multi-vendor, multicentre comparison of contrast-enhanced SSFP and
T2-STIR CMR for determining myocardium at risk in ST-elevation myocar-
ventricular remodeling. Circ Cardiovasc Imaging. 2016;9:e004940.
dial infarction. Eur Heart J Cardiovasc Imaging. 2016;17:744–753. doi:
16. Bulluck H, Rosmini S, Abdel-Gadir A, White SK, Bhuva AN, Treibel TA,
10.1093/ehjci/jew027.
Fontana M, Gonzalez-Lopez E, Reant P, Ramlall M, Hamarneh A, Sirker
32. Nordlund D, Kanski M, Jablonowski R, Koul S, Erlinge D, Carlsson M,
A, Herrey AS, Manisty C, Yellon DM, Kellman P, Moon JC, Hausenloy DJ.
Engblom H, Aletras AH, Arheden H. Experimental validation of contrast-
Automated extracellular volume fraction mapping provides insights into
enhanced SSFP cine CMR for quantification of myocardium at risk in
the pathophysiology of left ventricular remodeling post-reperfused ST-
acute myocardial infarction. J Cardiovasc Magn Reson. 2017;19:12. doi:
elevation myocardial infarction. J Am Heart Assoc. 2016;5:e003555.
10.1186/s12968-017-0325-y.
17. Carberry J, Carrick D, Haig C, Rauhalammi SM, Ahmed N, Mordi I, McEn-
33. Dall’Armellina E, Piechnik SK, Ferreira VM, Si QL, Robson MD, Fran-
Downloaded from http://ahajournals.org by on March 3, 2020

tegart M, Petrie MC, Eteiba H, Hood S, Watkins S, Lindsay M, Davie A,


cis JM, Cuculi F, Kharbanda RK, Banning AP, Choudhury RP, Karamit-
Mahrous A, Ford I, Sattar N, Welsh P, Radjenovic A, Oldroyd KG, Berry C.
sos TD, Neubauer S. Cardiovascular magnetic resonance by non
Remote zone extracellular volume and left ventricular remodeling in sur-
contrast T1-mapping allows assessment of severity of injury in acute
vivors of ST-elevation myocardial infarction. Hypertension. 2016;68:385–
myocardial infarction. J Cardiovasc Magn Reson. 2012;14:15. doi:
391. doi: 10.1161/HYPERTENSIONAHA.116.07222.
10.1186/1532-429X-14-15.
18. Bradley WG Jr. MR appearance of hemorrhage in the brain. Radiology.
34. Bøtker HE, Kaltoft AK, Pedersen SF, Kim WY. Measuring myocardial sal-
1993;189:15–26. doi: 10.1148/radiology.189.1.8372185.
vage. Cardiovasc Res. 2012;94:266–275. doi: 10.1093/cvr/cvs081.
19. Carrick D, Haig C, Rauhalammi S, Ahmed N, Mordi I, McEntegart M,
35. Engblom H, Heiberg E, Erlinge D, Jensen SE, Nordrehaug JE, Dubois-Randé
Petrie MC, Eteiba H, Hood S, Watkins S, Lindsay M, Mahrous A, Ford I, JL, Halvorsen S, Hoffmann P, Koul S, Carlsson M, Atar D, Arheden H.
Tzemos N, Sattar N, Welsh P, Radjenovic A, Oldroyd KG, Berry C. Prog- Sample size in clinical cardioprotection trials using myocardial salvage in-
nostic significance of infarct core pathology revealed by quantitative non- dex, infarct size, or biochemical markers as endpoint. J Am Heart Assoc.
contrast in comparison with contrast cardiac magnetic resonance imag- 2016;5:e002708. doi: 10.1161/JAHA.115.002708.
ing in reperfused ST-elevation myocardial infarction survivors. Eur Heart J. 36. Kim RJ, Fieno DS, Parrish TB, Harris K, Chen EL, Simonetti O, Bundy J, Finn
2016;37:1044–1059. doi: 10.1093/eurheartj/ehv372. JP, Klocke FJ, Judd RM. Relationship of MRI delayed contrast enhance-
20. Kumar A, Green JD, Sykes JM, Ephrat P, Carson JJ, Mitchell AJ, Wisen- ment to irreversible injury, infarct age, and contractile function. Circula-
berg G, Friedrich MG. Detection and quantification of myocardial reperfu- tion. 1999;100:1992–2002.
sion hemorrhage using T2*-weighted CMR. JACC Cardiovasc Imaging. 37. Krug A, Du Mesnil de Rochemont R, Korb G. Blood supply of the myocar-
2011;4:1274–1283. doi: 10.1016/j.jcmg.2011.08.016. dium after temporary coronary occlusion. Circ Res. 1966;19:57–62.
21. Kali A, Tang RL, Kumar A, Min JK, Dharmakumar R. Detection of acute 38. Bulluck H, Rosmini S, Abdel-Gadir A, Bhuva AN, Treibel TA, Fontana M,
reperfusion myocardial hemorrhage with cardiac MR imaging: T2 versus Gonzalez-Lopez E, Ramlall M, Hamarneh A, Sirker A, Herrey AS, Man-
T2. Radiology. 2013;269:387–395. doi: 10.1148/radiol.13122397. isty C, Yellon DM, Moon JC, Hausenloy DJ. Diagnostic performance of
22. Ortiz-Pérez JT, Meyers SN, Lee DC, Kansal P, Klocke FJ, Holly TA, David- T1and T2mapping to detect intramyocardial hemorrhage in reperfused ST-
son CJ, Bonow RO, Wu E. Angiographic estimates of myocardium at segment elevation myocardial infarction (STEMI) patients. J Magn Reson
risk during acute myocardial infarction: validation study using cardiac Imaging. 2017;46:877–886. doi: 10.1002/jmri.25638.
magnetic resonance imaging. Eur Heart J. 2007;28:1750–1758. doi: 39. Hamirani YS, Wong A, Kramer CM, Salerno M. Effect of microvascular
10.1093/eurheartj/ehm212. obstruction and intramyocardial hemorrhage by CMR on LV remod-
23. Aletras AH, Tilak GS, Natanzon A, Hsu LY, Gonzalez FM, Hoyt RF Jr, Arai eling and outcomes after myocardial infarction: a systematic review
AE. Retrospective determination of the area at risk for reperfused acute and meta-analysis. JACC Cardiovasc Imaging. 2014;7:940–952. doi:
myocardial infarction with T2-weighted cardiac magnetic resonance 10.1016/j.jcmg.2014.06.012.
imaging: histopathological and displacement encoding with stimulated 40. Arai AE. Magnetic resonance imaging for area at risk, myocardial infarc-
echoes (DENSE) functional validations. Circulation. 2006;113:1865–1870. tion, and myocardial salvage. J Cardiovasc Pharmacol Ther. 2011;16:313–
doi: 10.1161/CIRCULATIONAHA.105.576025. 320. doi: 10.1177/1074248411412378.
24. Berry C, Kellman P, Mancini C, Chen MY, Bandettini WP, Lowrey T, 41. Bulluck H, Hammond-Haley M, Fontana M, Knight DS, Sirker A, Herrey
Hsu LY, Aletras AH, Arai AE. Magnetic resonance imaging delineates AS, Manisty C, Kellman P, Moon JC, Hausenloy DJ. Quantification of both
the ischemic area at risk and myocardial salvage in patients with acute the area at risk and acute myocardial infarct size in ST-segment eleva-
myocardial infarction. Circ Cardiovasc Imaging. 2010;3:527–535. doi: tion myocardial infarction using T1-mapping. J Cardiovasc Magn Reson.
10.1161/CIRCIMAGING.109.900761. 2017;19:57. doi: 10.1186/s12968-017-0370-6.

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1961


Bulluck et al The Role of CMR in STEMI

42. Kali A, Choi EY, Sharif B, Kim YJ, Bi X, Spottiswoode B, Cokic I, Yang HJ, aging. doi: 10.1016/j.jcmg.2017.08.027. https://www.sciencedirect.com/
Tighiouart M, Conte AH, Li D, Berman DS, Choi BW, Chang HJ, Dharma- science/article/pii/S1936878X17309166?via%3Dihub.
STATE OF THE ART

kumar R. Native T1 mapping by 3-T CMR imaging for characterization of 56. Reinstadler SJ, Stiermaier T, Liebetrau J, Fuernau G, Eitel C, de Waha S,
chronic myocardial infarctions. JACC Cardiovasc Imaging. 2015;8:1019– Desch S, Reil JC, Poss J, Metzler B, Lucke C, Gutberlet M, Schuler G, Thiele
1030. doi: 10.1016/j.jcmg.2015.04.018. H, Eitel I. Prognostic significance of remote myocardium alterations as-
43. Liu D, Borlotti A, Viliani D, Jerosch-Herold M, Alkhalil M, De Maria GL, sessed by quantitative noncontrast T1 mapping in ST-segment elevation
Fahrni G, Dawkins S, Wijesurendra R, Francis J, Ferreira V, Piechnik S, Rob- myocardial infarction. JACC Cardiovasc Imaging. 2018;11:411–419. doi:
son MD, Banning A, Choudhury R, Neubauer S, Channon K, Kharbanda R, 10.1016/j.jcmg.2017.03.015.
Dall’Armellina E. CMR native T1 mapping allows differentiation of revers- 57. Carrick D, Haig C, Rauhalammi S, Ahmed N, Mordi I, McEntegart M, Petrie
ible versus irreversible myocardial damage in ST-segment-elevation myo- MC, Eteiba H, Lindsay M, Watkins S, Hood S, Davie A, Mahrous A, Sattar
cardial infarction: an OxAMI Study (Oxford Acute Myocardial Infarction). N, Welsh P, Tzemos N, Radjenovic A, Ford I, Oldroyd KG, Berry C. Patho-
Circ Cardiovasc Imaging. 2017;10:e005986. physiology of LV remodeling in survivors of STEMI: inflammation, remote
44. Garg P, Broadbent D, Swoboda P, Foley JR, Fent GJ, Musa TA, Ripley DP, myocardium, and prognosis. JACC Cardiovasc Imaging. 2015;8:779–789.
Erhayiem B, Dobson LE, McDiarmid A, Haaf P, Kidambi A, van der Geest doi: 10.1016/j.jcmg.2015.03.007.
RJ, Greenwood JP, Plein S. Acute infarct extracellular volume mapping to 58. Garg P, Broadbent DA, Swoboda PP, Foley JRJ, Fent GJ, Musa TA, Ripley
quantify myocardial area at risk and chronic infarct size on cardiovascular DP, Erhayiem B, Dobson LE, McDiarmid AK, Haaf P, Kidambi A, Crandon
magnetic resonance imaging. Circ Cardiovasc Imaging. 2017;10:e006182. S, Chew PG, van der Geest RJ, Greenwood JP, Plein S. Extra-cellular expan-
45. Bulluck H, Hausenloy DJ. Mapping myocardial salvage index by ex- sion in the normal, non-infarcted myocardium is associated with worsen-
tracellular volume fraction: are we there yet? Circ Cardiovasc Imaging. ing of regional myocardial function after acute myocardial infarction. J
2017;10:e006680. Cardiovasc Magn Reson. 2017;19:73. doi: 10.1186/s12968-017-0384-0.
46. Kidambi A, Motwani M, Uddin A, Ripley DP, McDiarmid AK, Swoboda 59. Stone GW, Selker HP, Thiele H, Patel MR, Udelson JE, Ohman EM, Maehara
PP, Broadbent DA, Musa TA, Erhayiem B, Leader J, Croisille P, Clarysse A, Eitel I, Granger CB, Jenkins PL, Nichols M, Ben-Yehuda O. Relationship
P, Greenwood JP, Plein S. Myocardial extracellular volume estimation by between infarct size and outcomes following primary PCI: patient-level
CMR predicts functional recovery following acute MI. JACC Cardiovasc analysis from 10 randomized trials. J Am Coll Cardiol. 2016;67:1674–
Imaging. 2017;10:989–999. doi: 10.1016/j.jcmg.2016.06.015. 1683. doi: 10.1016/j.jacc.2016.01.069.
47. Ørn S, Manhenke C, Greve OJ, Larsen AI, Bonarjee VV, Edvardsen T, Dick- 60. Symons R, Pontone G, Schwitter J, Francone M, Iglesias JF, Barison A,
stein K. Microvascular obstruction is a major determinant of infarct heal- Zalewski J, de Luca L, Degrauwe S, Claus P, Guglielmo M, Nessler J, Car-
ing and subsequent left ventricular remodelling following primary per- bone I, Ferro G, Durak M, Magistrelli P, Lo Presti A, Aquaro GD, Eeckhout
cutaneous coronary intervention. Eur Heart J. 2009;30:1978–1985. doi: E, Roguelov C, Andreini D, Vogt P, Guaricci AI, Mushtaq S, Lorenzoni V,
10.1093/eurheartj/ehp219. Muller O, Desmet W, Agati L, Janssens S, Bogaert J, Masci PG. Long-
48. van Kranenburg M, Magro M, Thiele H, de Waha S, Eitel I, Cochet A, term incremental prognostic value of cardiovascular magnetic resonance
Cottin Y, Atar D, Buser P, Wu E, Lee D, Bodi V, Klug G, Metzler B, Delewi after ST-segment elevation myocardial infarction: a study of the collabora-
R, Bernhardt P, Rottbauer W, Boersma E, Zijlstra F, van Geuns RJ. Prog- tive registry on CMR in STEMI [published online ahead of print August
nostic value of microvascular obstruction and infarct size, as measured by 11, 2017]. JACC Cardiovasc Imaging. doi: 10.1016/j.jcmg.2017.05.023.
CMR in STEMI patients. JACC Cardiovasc Imaging. 2014;7:930–939. doi: https://www.sciencedirect.com/science/article/pii/S1936878X17306204?
10.1016/j.jcmg.2014.05.010. via%3Dihub.
49. de Waha S, Patel MR, Granger CB, Ohman EM, Maehara A, Eitel I, Ben- 61. Eitel I, de Waha S, Wöhrle J, Fuernau G, Lurz P, Pauschinger M, Desch S,
Downloaded from http://ahajournals.org by on March 3, 2020

Yehuda O, Jenkins P, Thiele H, Stone GW. Relationship between micro- Schuler G, Thiele H. Comprehensive prognosis assessment by CMR imag-
vascular obstruction and adverse events following primary percutaneous ing after ST-segment elevation myocardial infarction. J Am Coll Cardiol.
coronary intervention for ST-segment elevation myocardial infarction: an 2014;64:1217–1226. doi: 10.1016/j.jacc.2014.06.1194.
individual patient data pooled analysis from seven randomized trials. Eur 62. Amier RP, Tijssen RYG, Teunissen PFA, Fernandez-Jimenez R, Pizarro G,
Heart J. 2017;38:3502–3510. doi: 10.1093/eurheartj/ehx414. Garcia-Lunar I, Bastante T, van de Ven PM, Beek AM, Smulders MW, Bek-
50. Westman PC, Lipinski MJ, Luger D, Waksman R, Bonow RO, Wu E, Epstein kers S, van Royen N, Ibanez B, Nijveldt R. Predictors of intramyocardial
SE. Inflammation as a driver of adverse left ventricular remodeling after hemorrhage after reperfused ST-segment elevation myocardial infarction.
acute myocardial infarction. J Am Coll Cardiol. 2016;67:2050–2060. doi: J Am Heart Assoc. 2017;6:e005651.
10.1016/j.jacc.2016.01.073. 63. Kanazawa H, Tseliou E, Malliaras K, Yee K, Dawkins JF, De Couto G, Smith
51. Bulluck H, Go YY, Crimi G, Ludman AJ, Rosmini S, Abdel-Gadir A, Bhuva RR, Kreke M, Seinfeld J, Middleton RC, Gallet R, Cheng K, Luthringer
AN, Treibel TA, Fontana M, Pica S, Raineri C, Sirker A, Herrey AS, Manisty D, Valle I, Chowdhury S, Fukuda K, Makkar RR, Marbán L, Marbán E.
C, Groves A, Moon JC, Hausenloy DJ. Defining left ventricular remodeling Cellular postconditioning: allogeneic cardiosphere-derived cells reduce
following acute ST-segment elevation myocardial infarction using cardio- infarct size and attenuate microvascular obstruction when administered
vascular magnetic resonance. J Cardiovasc Magn Reson. 2017;19:26. doi: after reperfusion in pigs with acute myocardial infarction. Circ Heart Fail.
10.1186/s12968-017-0343-9. 2015;8:322–332. doi: 10.1161/CIRCHEARTFAILURE.114.001484.
52. Kali A, Kumar A, Cokic I, Tang RL, Tsaftaris SA, Friedrich MG, Dharmaku- 64. Higgins CB, Herfkens R, Lipton MJ, Sievers R, Sheldon P, Kaufman L,
mar R. Chronic manifestation of postreperfusion intramyocardial hemor- Crooks LE. Nuclear magnetic resonance imaging of acute myocardial in-
rhage as regional iron deposition: a cardiovascular magnetic resonance farction in dogs: alterations in magnetic relaxation times. Am J Cardiol.
study with ex vivo validation. Circ Cardiovasc Imaging. 2013;6:218–228. 1983;52:184–188.
doi: 10.1161/CIRCIMAGING.112.000133. 65. Friedrich MG. Myocardial edema: a new clinical entity? Nat Rev Cardiol.
53. Kali A, Cokic I, Tang R, Dohnalkova A, Kovarik L, Yang HJ, Kumar A, Prato 2010;7:292–296. doi: 10.1038/nrcardio.2010.28.
FS, Wood JC, Underhill D, Marban E, Dharmakumar R. Persistent micro- 66. Tilak GS, Hsu LY, Hoyt RF Jr, Arai AE, Aletras AH. In vivo T2-weighted
vascular obstruction after myocardial infarction culminates in the conflu- magnetic resonance imaging can accurately determine the ischemic area
ence of ferric iron oxide crystals, proinflammatory burden, and adverse at risk for 2-day-old nonreperfused myocardial infarction. Invest Radiol.
remodeling. Circ Cardiovasc Imaging. 2016;9:e004996. 2008;43:7–15. doi: 10.1097/RLI.0b013e3181558822.
54. Roghi A, Poggiali E, Duca L, Mafrici A, Pedrotti P, Paccagnini S, Brenna S, 67. Ubachs JF, Engblom H, Koul S, Kanski M, Andersson P, van der Pals J,
Galli A, Consonni D, Cappellini MD. Role of non-transferrin-bound iron in Carlsson M, Erlinge D, Arheden H. Myocardium at risk can be determined
the pathogenesis of cardiotoxicity in patients with ST-elevation myocardial by ex vivo T2-weighted magnetic resonance imaging even in the pres-
infarction assessed by cardiac magnetic resonance imaging. Int J Cardiol. ence of gadolinium: comparison to myocardial perfusion single pho-
2015;199:326–332. doi: 10.1016/j.ijcard.2015.07.056. ton emission computed tomography. Eur Heart J Cardiovasc Imaging.
55. Carberry J, Carrick D, Haig C, Ahmed N, Mordi I, McEntegart M, Petrie 2013;14:261–268. doi: 10.1093/ehjci/jes142.
MC, Eteiba H, Hood S, Watkins S, Lindsay M, Davie A, Mahrous A, Ford 68. Kim HW, Van Assche L, Jennings RB, Wince WB, Jensen CJ, Rehwald WG,
I, Sattar N, Welsh P, Radjenovic A, Oldroyd KG, Berry C. Persistent iron Wendell DC, Bhatti L, Spatz DM, Parker MA, Jenista ER, Klem I, Crowley
within the infarct core after ST-segment elevation myocardial infarction: AL, Chen EL, Judd RM, Kim RJ. Relationship of T2-weighted MRI myocar-
implications for left ventricular remodeling and health outcomes [pub- dial hyperintensity and the ischemic area at risk. Circ Res. 2015;117:254–
lished online ahead of print November 10, 2017]. JACC Cardiovasc Im- 265. doi: 10.1161/CIRCRESAHA.117.305771.

1962 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693


Bulluck et al The Role of CMR in STEMI

69. Bulluck H, White SK, Fröhlich GM, Casson SG, O’Meara C, New- in delayed enhancement of the myocardium during the 1st week after re-
ton A, Nicholas J, Weale P, Wan SM, Sirker A, Moon JC, Yellon DM, perfusion. Radiology. 2010;254:88–97. doi: 10.1148/radiol.09090660.

STATE OF THE ART


Groves A, Menezes L, Hausenloy DJ. Quantifying the area at risk in 81. Jablonowski R, Engblom H, Kanski M, Nordlund D, Koul S, van der Pals
reperfused ST-segment-elevation myocardial infarction patients us- J, Englund E, Heiberg E, Erlinge D, Carlsson M, Arheden H. Contrast-
ing hybrid cardiac positron emission tomography-magnetic reso- enhanced CMR overestimates early myocardial infarct size: mechanistic
nance imaging. Circ Cardiovasc Imaging. 2016;9:e003900. doi: insights using ECV measurements on day 1 and day 7. JACC Cardiovasc
10.1161/CIRCIMAGING.115.003900. Imaging. 2015;8:1379–1389. doi: 10.1016/j.jcmg.2015.08.015.
70. Hammer-Hansen S, Ugander M, Hsu LY, Taylor J, Thune JJ, Køber L, Kell- 82. Hammer-Hansen S, Bandettini WP, Hsu LY, Leung SW, Shanbhag S, Mancini C,
man P, Arai AE. Distinction of salvaged and infarcted myocardium within Greve AM, Køber L, Thune JJ, Kellman P, Arai AE. Mechanisms for overesti-
the ischaemic area at risk with T2 mapping. Eur Heart J Cardiovasc Imag- mating acute myocardial infarct size with gadolinium-enhanced cardiovascu-
ing. 2014;15:1048–1053. doi: 10.1093/ehjci/jeu073. lar magnetic resonance imaging in humans: a quantitative and kinetic study.
71. Dall’Armellina E, Karia N, Lindsay AC, Karamitsos TD, Ferreira V, Rob- Eur Heart J Cardiovasc Imaging. 2016;17:76–84. doi: 10.1093/ehjci/jev123.
son MD, Kellman P, Francis JM, Forfar C, Prendergast BD, Banning AP, 83. Rodríguez-Palomares JF, Ortiz-Pérez JT, Lee DC, Bucciarelli-Ducci C, Teje-
Channon KM, Kharbanda RK, Neubauer S, Choudhury RP. Dynamic dor P, Bonow RO, Wu E. Time elapsed after contrast injection is crucial to
changes of edema and late gadolinium enhancement after acute determine infarct transmurality and myocardial functional recovery after
myocardial infarction and their relationship to functional recovery an acute myocardial infarction. J Cardiovasc Magn Reson. 2015;17:43.
and salvage index. Circ Cardiovasc Imaging. 2011;4:228–236. doi: doi: 10.1186/s12968-015-0139-8.
10.1161/CIRCIMAGING.111.963421. 84. Mather AN, Fairbairn TA, Artis NJ, Greenwood JP, Plein S. Timing of car-
72. Carlsson M, Ubachs JF, Hedström E, Heiberg E, Jovinge S, Arheden H. diovascular MR imaging after acute myocardial infarction: effect on esti-
Myocardium at risk after acute infarction in humans on cardiac magnetic mates of infarct characteristics and prediction of late ventricular remodel-
resonance: quantitative assessment during follow-up and validation with ing. Radiology. 2011;261:116–126. doi: 10.1148/radiol.11110228.
single-photon emission computed tomography. JACC Cardiovasc Imaging. 85. Engblom H, Hedström E, Heiberg E, Wagner GS, Pahlm O, Arheden H.
2009;2:569–576. doi: 10.1016/j.jcmg.2008.11.018. Rapid initial reduction of hyperenhanced myocardium after reperfused
73. Fernández-Jiménez R, Sánchez-González J, Agüero J, García-Prieto J, first myocardial infarction suggests recovery of the peri-infarction zone:
López-Martín GJ, García-Ruiz JM, Molina-Iracheta A, Rosselló X, Fernán- one-year follow-up by MRI. Circ Cardiovasc Imaging. 2009;2:47–55. doi:
dez-Friera L, Pizarro G, García-Álvarez A, Dall’Armellina E, Macaya C, 10.1161/CIRCIMAGING.108.802199.
Choudhury RP, Fuster V, Ibáñez B. Myocardial edema after ischemia/re- 86. Lønborg J, Vejlstrup N, Kelbæk H, Holmvang L, Jørgensen E, Helqvist S,
perfusion is not stable and follows a bimodal pattern: imaging and histo- Saunamäki K, Ahtarovski KA, Bøtker HE, Kim WY, Clemmensen P, Eng-
logical tissue characterization. J Am Coll Cardiol. 2015;65:315–323. doi:
strøm T. Final infarct size measured by cardiovascular magnetic resonance
10.1016/j.jacc.2014.11.004.
in patients with ST elevation myocardial infarction predicts long-term
74. Fernández-Jiménez R, García-Prieto J, Sánchez-González J, Agüero J,
clinical outcome: an observational study. Eur Heart J Cardiovasc Imaging.
López-Martín GJ, Galán-Arriola C, Molina-Iracheta A, Doohan R, Fuster V,
2013;14:387–395. doi: 10.1093/ehjci/jes271.
Ibáñez B. Pathophysiology underlying the bimodal edema phenomenon
87. Wu E, Ortiz JT, Tejedor P, Lee DC, Bucciarelli-Ducci C, Kansal P, Carr JC, Holly
after myocardial ischemia/reperfusion. J Am Coll Cardiol. 2015;66:816–
TA, Lloyd-Jones D, Klocke FJ, Bonow RO. Infarct size by contrast enhanced
828. doi: 10.1016/j.jacc.2015.06.023.
cardiac magnetic resonance is a stronger predictor of outcomes than left
75. Thuny F, Lairez O, Roubille F, Mewton N, Rioufol G, Sportouch C, Sanchez
ventricular ejection fraction or end-systolic volume index: prospective co-
I, Bergerot C, Thibault H, Cung TT, Finet G, Argaud L, Revel D, Derumeaux
hort study. Heart. 2008;94:730–736. doi: 10.1136/hrt.2007.122622.
G, Bonnefoy-Cudraz E, Elbaz M, Piot C, Ovize M, Croisille P. Post-condi-
Downloaded from http://ahajournals.org by on March 3, 2020

88. Thiele H, Kappl MJ, Conradi S, Niebauer J, Hambrecht R, Schuler G.


tioning reduces infarct size and edema in patients with ST-segment eleva-
Reproducibility of chronic and acute infarct size measurement by de-
tion myocardial infarction. J Am Coll Cardiol. 2012;59:2175–2181. doi:
layed enhancement-magnetic resonance imaging. J Am Coll Cardiol.
10.1016/j.jacc.2012.03.026.
2006;47:1641–1645. doi: 10.1016/j.jacc.2005.11.065.
76. White SK, Frohlich GM, Sado DM, Maestrini V, Fontana M, Treibel TA,
89. Bulluck H, Rosmini S, Abdel-Gadir A, Bhuva AN, Treibel TA, Fontana M,
Tehrani S, Flett AS, Meier P, Ariti C, Davies JR, Moon JC, Yellon DM,
Weinmann S, Sirker A, Herrey AS, Manisty C, Moon JC, Hausenloy DJ. Impact
Hausenloy DJ. Remote ischemic conditioning reduces myocardial in-
of microvascular obstruction on semiautomated techniques for quantifying
farct size and edema in patients with ST-segment elevation myocar-
dial infarction. JACC Cardiovasc Interv. 2015;8(pt B):178–188. doi: acute and chronic myocardial infarction by cardiovascular magnetic reso-
10.1016/j.jcin.2014.05.015. nance. Open Heart. 2016;3:e000535. doi: 10.1136/openhrt-2016-000535.
77. Eitel I, Stiermaier T, Rommel KP, Fuernau G, Sandri M, Mangner N, Linke 90. Yang HJ, Sharif B, Pang J, Kali A, Bi X, Cokic I, Li D, Dharmakumar R.
A, Erbs S, Lurz P, Boudriot E, Mende M, Desch S, Schuler G, Thiele H. Car- Free-breathing, motion-corrected, highly efficient whole heart T2 map-
dioprotection by combined intrahospital remote ischaemic percondition- ping at 3T with hybrid radial-cartesian trajectory. Magn Reson Med.
ing and postconditioning in ST-elevation myocardial infarction: the ran- 2016;75:126–136. doi: 10.1002/mrm.25576.
domized LIPSIA CONDITIONING trial. Eur Heart J. 2015;36:3049–3057. 91. Kellman P, Xue H, Spottiswoode BS, Sandino CM, Hansen MS, Abdel-
doi: 10.1093/eurheartj/ehv463. Gadir A, Treibel TA, Rosmini S, Mancini C, Bandettini WP, McGill LA,
78. Ibanez B, Macaya C, Sánchez-Brunete V, Pizarro G, Fernández-Friera L, Gatehouse P, Moon JC, Pennell DJ, Arai AE. Free-breathing T2* mapping
Mateos A, Fernández-Ortiz A, García-Ruiz JM, García-Álvarez A, Iñiguez using respiratory motion corrected averaging. J Cardiovasc Magn Reson.
A, Jiménez-Borreguero J, López-Romero P, Fernández-Jiménez R, Goico- 2015;17:3. doi: 10.1186/s12968-014-0106-9.
lea J, Ruiz-Mateos B, Bastante T, Arias M, Iglesias-Vázquez JA, Rodriguez 92. Kellman P, Arai AE. Cardiac imaging techniques for physicians: late
MD, Escalera N, Acebal C, Cabrera JA, Valenciano J, Pérez de Prado A, enhancement. J Magn Reson Imaging. 2012;36:529–542. doi:
Fernández-Campos MJ, Casado I, García-Rubira JC, García-Prieto J, Sanz- 10.1002/jmri.23605.
Rosa D, Cuellas C, Hernández-Antolín R, Albarrán A, Fernández-Vázquez 93. Seropian IM, Toldo S, Van Tassell BW, Abbate A. Anti-inflammatory strat-
F, de la Torre-Hernández JM, Pocock S, Sanz G, Fuster V. Effect of early egies for ventricular remodeling following ST-segment elevation acute
metoprolol on infarct size in ST-segment-elevation myocardial infarction myocardial infarction. J Am Coll Cardiol. 2014;63:1593–1603. doi:
patients undergoing primary percutaneous coronary intervention: the Ef- 10.1016/j.jacc.2014.01.014.
fect of Metoprolol in Cardioprotection During an Acute Myocardial In- 94. Stiermaier T, Jobs A, de Waha S, Fuernau G, Poss J, Desch S, Thiele H,
farction (METOCARD-CNIC) trial. Circulation. 2013;128:1495–1503. doi: Eitel I. Optimized prognosis assessment in ST-segment-elevation myocar-
10.1161/CIRCULATIONAHA.113.003653. dial infarction using a cardiac magnetic resonance imaging risk score. Circ
79. Lønborg J, Vejlstrup N, Kelbæk H, Bøtker HE, Kim WY, Mathiasen AB, Cardiovasc Imaging. 2017;10:e006774.
Jørgensen E, Helqvist S, Saunamäki K, Clemmensen P, Holmvang L, 95. Pontone G, Guaricci AI, Andreini D, Ferro G, Guglielmo M, Baggiano A,
Thuesen L, Krusell LR, Jensen JS, Køber L, Treiman M, Holst JJ, Engstrøm Fusini L, Muscogiuri G, Lorenzoni V, Mushtaq S, Conte E, Annoni A, For-
T. Exenatide reduces reperfusion injury in patients with ST-segment el- menti A, Mancini ME, Carita P, Verdecchia M, Pica S, Fazzari F, Cosentino
evation myocardial infarction. Eur Heart J. 2012;33:1491–1499. doi: N, Marenzi G, Rabbat MG, Agostoni P, Bartorelli AL, Pepi M, Masci PG.
10.1093/eurheartj/ehr309. Prognostic Stratification of Patients With ST-Segment-Elevation Myocar-
80. Ibrahim T, Hackl T, Nekolla SG, Breuer M, Feldmair M, Schömig A, Schwaiger dial Infarction (PROSPECT): a cardiac magnetic resonance study. Circ Car-
M. Acute myocardial infarction: serial cardiac MR imaging shows a decrease diovasc Imaging. 2017;10:e006428.

Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693 May 1, 2018 1963


Bulluck et al The Role of CMR in STEMI

96. Dharmakumar R. “Rusty hearts”: is it time to rethink iron chelation cardiovascular magnetic resonance for the prediction of adverse arrhyth-
therapies in post-myocardial-infarction setting? Circ Cardiovasc Imaging. mic cardiac events after a first noncomplicated ST-segment-elevation
STATE OF THE ART

2016;9:e005541. myocardial infarction. Circ Cardiovasc Imaging. 2013;6:755–761. doi:


97. O’Gara PT, Kushner FG, Ascheim DD, Casey DE Jr, Chung MK, de Lemos 10.1161/CIRCIMAGING.113.000702.
JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, Granger CB, Krumholz 101. Watanabe E, Abbasi SA, Heydari B, Coelho-Filho OR, Shah R, Neilan TG,
HM, Linderbaum JA, Morrow DA, Newby LK, Ornato JP, Ou N, Radford MJ, Murthy VL, Mongeon FP, Barbhaiya C, Jerosch-Herold M, Blankstein R,
Tamis-Holland JE, Tommaso CL, Tracy CM, Woo YJ, Zhao DX; American Hatabu H, van der Geest RJ, Stevenson WG, Kwong RY. Infarct tissue het-
College of Cardiology Foundation; American Heart Association Task erogeneity by contrast-enhanced magnetic resonance imaging is a novel
Force on Practice Guidelines; American College of Emergency Physicians; predictor of mortality in patients with chronic coronary artery disease and
Society for Cardiovascular Angiography and Interventions. 2013 ACCF/ left ventricular dysfunction. Circ Cardiovasc Imaging. 2014;7:887–894.
AHA guideline for the management of ST-elevation myocardial infarc- doi: 10.1161/CIRCIMAGING.113.001293.
tion: executive summary: a report of the American College of Cardiology 102. Klem I, Weinsaft JW, Bahnson TD, Hegland D, Kim HW, Hayes B,
Foundation/American Heart Association Task Force on Practice Guidelines: Parker MA, Judd RM, Kim RJ. Assessment of myocardial scarring
developed in collaboration with the American College of Emergency improves risk stratification in patients evaluated for cardiac defi-
Physicians and Society for Cardiovascular Angiography and Interventions. brillator implantation. J Am Coll Cardiol. 2012;60:408–420. doi:
Catheter Cardiovasc Interv. 2013;82:E1–E27. doi: 10.1002/ccd.24776. 10.1016/j.jacc.2012.02.070.
98. Zaman S, Kovoor P. Sudden cardiac death early after myocardial infarc- 103. Cokic I, Kali A, Yang HJ, Yee R, Tang R, Tighiouart M, Wang X, Jackman
tion: pathogenesis, risk stratification, and primary prevention. Circulation. WS, Chugh SS, White JA, Dharmakumar R. Iron-sensitive cardiac mag-
2014;129:2426–2435. doi: 10.1161/CIRCULATIONAHA.113.007497. netic resonance imaging for prediction of ventricular arrhythmia risk
99. Pouleur AC, Barkoudah E, Uno H, Skali H, Finn PV, Zelenkofske SL, in patients with chronic myocardial infarction: early evidence. Circ
Belenkov YN, Mareev V, Velazquez EJ, Rouleau JL, Maggioni AP, Cardiovasc Imaging. 2015;8:e003642.
Køber L, Califf RM, McMurray JJ, Pfeffer MA, Solomon SD; VALIANT 104. Treibel TA, Fontana M, Maestrini V, Castelletti S, Rosmini S, Simpson
Investigators. Pathogenesis of sudden unexpected death in a clini- J, Nasis A, Bhuva AN, Bulluck H, Abdel-Gadir A, White SK, Manisty
cal trial of patients with myocardial infarction and left ventricular dys- C, Spottiswoode BS, Wong TC, Piechnik SK, Kellman P, Robson MD,
function, heart failure, or both. Circulation. 2010;122:597–602. doi: Schelbert EB, Moon JC. Automatic measurement of the myocardial
10.1161/CIRCULATIONAHA.110.940619. interstitium: synthetic extracellular volume quantification without he-
100. Izquierdo M, Ruiz-Granell R, Bonanad C, Chaustre F, Gomez C, Ferrero A, matocrit sampling. JACC Cardiovasc Imaging. 2016;9:54–63. doi:
Lopez-Lereu P, Monmeneu JV, Nuñez J, Chorro FJ, Bodi V. Value of early 10.1016/j.jcmg.2015.11.008.
Downloaded from http://ahajournals.org by on March 3, 2020

1964 May 1, 2018 Circulation. 2018;137:1949–1964. DOI: 10.1161/CIRCULATIONAHA.117.030693

You might also like