115 - vrije universiteit amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus...

17
VU Research Portal From Population Screening to Advanced Detection of Coronary Disease Laufer, E.M. 2019 document version Publisher's PDF, also known as Version of record Link to publication in VU Research Portal citation for published version (APA) Laufer, E. M. (2019). From Population Screening to Advanced Detection of Coronary Disease. General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal ? Take down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. E-mail address: [email protected] Download date: 09. Mar. 2021

Upload: others

Post on 13-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

VU Research Portal

From Population Screening to Advanced Detection of Coronary Disease

Laufer, E.M.

2019

document versionPublisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)Laufer, E. M. (2019). From Population Screening to Advanced Detection of Coronary Disease.

General rightsCopyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright ownersand it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights.

• Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal ?

Take down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

E-mail address:[email protected]

Download date: 09. Mar. 2021

Page 2: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

114

Page 3: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

115

Chapter 7.

Molecular Imaging of Macrophage Cell Death for the Assessment of Plaque Vulnerability

Eduard M. Laufer 1,2 , Mark H.M. Winkens 1,2 , Jagat Narula 3 and Leonard

Hofstra 1,2

1. Department of Cardiology, Maastricht University Medical center, the Netherlands

2. Cardiovascular research Institute Maastricht, The Netherlands

3. Department of Cardiology, University of California Irvine

Arterioscler Thromb Vasc Biol. 2009 Jul;29(7):1031-8.

Page 4: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

116

Introduction

Atherosclerotic disease remains killer number one in both developed and

developing nations. It afflicts women and men alike. Because of the epidemic

proportions of obesity and aging of the population, it is expected that the

impact of atherosclerotic disease will remain high in the new century as well.

The first signs of the impact of the escalating proportions of obesity may have

influenced recent event rates in the United States, wherein the decline in the

incidence of acute myocardial infarction in men has come to a halt, and in

women the numbers have started to escalate.

The main challenge for clinicians is to identify individuals who have a

high likelihood of developing myocardial infarction. Clinical risk profiling

algorithms, such as the Framingham and the Procam risk scores, have proven

to be useful tools for the identification of individuals with a high risk to

develop acute myocardial infarction in the next 10 years. Typically, the

clinical risk scoring profiles categorize individuals in low risk (less than 5%

10-year risk), intermediate risk (5% to 20%), and high risk (20% or more) to

develop acute myocardial infarction.1 However, clinical risk profiling does

not provide a clear answer about the time frame when events may occur in

the next 10 years. We may also need to find ways to improve the area under

the ROC curve to enhance predictive value. Recent studies have shown that

the addition of novel serum biomarkers to clinical risk profiling had limited

beneficial effect, and that novel strategies need to be developed to improve

the predictive value of diagnostic algorithms.2

One way to obtain better predictive values and more precise information is

the use of imaging technology for identification of plaques that are vulnerable

to rupture.3 Traditional imaging methods have provided excellent information

about the location and the extent of luminal narrowing of atherosclerotic

lesions.4 Clinical research studies have demonstrated that the occurrence of

acute myocardial infarction is related to plaque characteristics. In at least

70% to 80% of cases, acute myocardial infarction is attributable to rupture

of a vulnerable plaque, resulting in local thrombus formation and acute

disruption in the coronary blood supply in the affected area.5 These studies

have shown that vulnerable plaques are characterized by a large necrotic

core, a thin fibrous cap, and by the presence of macrophages.6 Cell death of

inflammatory cells by the process of apoptosis has recently been proposed

to contribute to plaque vulnerability. Apoptosis has been linked to thinning

of the fibrous cap, the development of the necrotic core, and positive vessel

remodeling.7 Identification of apoptosis in atherosclerotic lesions may help

Page 5: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

117

Ch

apter 7 | M

olecu

lar Imagin

g of M

acrop

hage C

ell Death

Figure 1. Cartoon diagram illustrating where measurements of cell type and apoptosis were performed in a ruptured (above) and a stable (below) plaque. The color key at the bottom left describes the various plaque constituents. The black-hatched regions represent areas where measurements were taken. Quantitatively, the size of the rupture site varied between cases such that cell counts were performed in areas ranging from 0.12 to 0.20 mm2. Histological photomicrographs in the upper row demonstrate apoptosis in culprit plaques. The micrograph, next to the cartoon, is a cross-section of an epicardial coronary artery that shows the plaque rupture with an acute luminal thrombus (Th); note the thin fibrous cap, boxed area (H&E stain; original magnification, ×30). The boxed area from its serial section after DNA fragmentation staining by shows numerous apoptotic cells (blue nuclear staining, arrowheads) at the plaque rupture site (eosin counterstain; original magnification, ×150). The inset shows a high-power view illustrating the nuclear detail; note the fragmented nucleus (arrowhead; original magnification, ×1000). ISEL-positive nuclei were visualized with diaminobenzidine (dark-brown reaction product) and antibody staining was detected using an antimouse IgG conjugated with biotin and an avidin alkaline phosphatase-substrate system (red reaction product). The methyl green counterstain yields blue-green nuclei. The next 3 photomicrographs are stained for CD68 and ISEL, ASMA and ISEL, and anticaspase-1 antibody. Note the majority of apoptotic cells are macrophages (original magnification, ×150), and that most of the apoptotic nuclei are negative for SMCs (original magnification, ×300). Apoptotic macrophages demonstrate abundant caspase-1 at the site of plaque rupture (reddish-brown reaction product; original magnification, ×150). Overall, 45% of macrophages were recognized as apoptotic. On the other hand, a cross-section of an epicardial coronary artery shows an eccentric stable plaque in the bottom row. The lesion is characterized by dense fibrous cap, boxed area, overlying a calcified region (H&E; original magnification, ×30). Serial section after DNA fragmentation staining shows a paucity of apoptotic cells (arrowheads) relative to plaque rupture site (eosin counterstain; original magnification, ×150). Stable plaque shows rare apoptotic macrophages (original magnification, ×300), or SMCs (arrowheads) in the fibrous cap; no definite caspase-1 staining is seen (reddish-brown reaction product; original magnification, ×150). MF indicates macrophages; SMC, smooth muscle cells. L, lumen; NC, necrotic core; Th, thrombus; FC, fibrous cap. Modified with permission from Kolodgie et al.10

Page 6: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

118

identify vulnerable atherosclerotic plaques in patients. Because apoptosis is

characterized most prominently by the phosphatidylserine (PS) shift from the

intracellular leaflet to the extracellular leaflet of the plasma cell membrane,8

externalized PS provides an attractive target for apoptosis detection. Annexin

A5, a normally circulating protein, demonstrates a strong affinity for PS in the

nanomolar range and has proven to be an effective means to detect apoptosis

both in vitro as well as in vivo.9

Apoptosis in Plaque VulnerabilityA number of key characteristics in plaque vulnerability are related to

apoptotic cell death. A large part of the necrotic core is likely derived from

cell death of macrophage foam cells, and the cell death of smooth muscle

cells leads to thinning of the fibrous cap and positive remodeling of the

involved coronary segment. The thin-capped fibro-atheroma has been defined

as an atherosclerotic lesion with a fibrous cap of 65 mm or less. The fibrous

cap is made up of collagen, which is mainly derived from smooth muscle cells

(SMCs) of the plaque. The extracellular collagen helps to develop a stable and

thick fibrous cap, which prevents exposure of the thrombogenic necrotic core

to the blood. Regular loss of SMCs through apoptosis perpetuates attenuation

and weakening of the fibrous cap. Further, it has been demonstrated that

extensive and collective apoptosis of macrophages at the

site of plaque rupture is associated with the acute coronary event. Based on

these observations it appears that apoptosis and inflammation are substrates

for plaque disruption.6,11

In the study of culprit lesions obtained from victims of sudden coronary

death, the plaques were divided into 2 mechanisms of death: ruptured plaques

with acute thrombosis and stable plaques with and without healed myocardial

infarction.10 Apoptotic cells were identified by staining of fragmented DNA, and

expression and activation of 2 inducers of apoptosis, caspases-1 and -3. Unlike

stable lesions, ruptured plaques showed extensive macrophage infiltration of

the fibrous cap, in particular at rupture sites (Figure 1). Plaque rupture sites

demonstrated a strong immunoreactivity to caspase-1 within the apoptotic

macrophages; weak staining for caspase-3 was also observed. Although this

study demonstrated extensive apoptosis of macrophages limited to the site

of plaque rupture, it was not clear whether macrophage apoptosis is essential

to acute plaque rupture or is a response to the rupture itself.

Macrophage apoptosis may also facilitate the acute thrombotic event

arising from the rupture itself.13 They captured the shed membrane

microparticles originating from culprit carotid plaques as the products of

Page 7: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

119

Ch

apter 7 | M

olecu

lar Imagin

g of M

acrop

hage C

ell Death

apoptotic cells by using annexin A5. These microparticles were primarily

monocytic in origin and very rich in tissue factor and hence procoagulant

activity. Thus, it is conceivable that microparticles from macrophage apoptosis

at the rupture site would induce thrombogenecity.

Figure 2. Histological characterization of Annexin A5 uptake in experimental atherosclerotic lesions in rabbits. The lesions were classified according to the AHA classification (top) into type II lesions or fatty streaks, type III lesions with extracellular lipid pools, and type IV lesions with formation of necrotic core. Lesions beyond type IV lesions were not seen. Type IV lesions comprised 50% of total lesions, type III 30%, and type II 20%, respectively. Significant Annexin A5 uptake was visible only in type IV lesions. In situ end labelling studies demonstrated higher macrophage apoptosis in type IV lesions (below) compared to type II and III lesions and explain higher Annexin uptake in type IV lesions. Modified with permission from Kolodgie et al. 12

Page 8: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

120

Feasibility of Imaging Apoptosis in Atherosclerotic LesionsThe proof of concept of exogenous radiolabeled Annexin A5 to image apoptosis

and inflammation was provided in an experimental model of atherosclerosis in

rabbits.12 Atherosclerotic lesions were formed by balloon de-endothelialization

of the infradiaphragmatic aorta, followed by 16 weeks of high-cholesterol high-

fat diet. Control rabbits were studied without manipulation. In vivo imaging

was performed after intravenous administration of 0.5 to 1 mg of Annexin

A5 labeled with technetium-99m, followed by harvesting of the abdominal

aortas, ex vivo imaging, and macro-autoradiography. In addition, histological

examination was performed to localize the radiotracer in the atherosclerotic

lesions. Two hours after injection of the radiolabeled tracer, there was

clear demarcation of the radiolabel in the aorta by in vivo gamma imaging

(Figure 2). Ex vivo imaging demonstrated a clear uptake of radiotracer in the

aorta corresponding to the in vivo images and confirming the macroscopic

localization of atherosclerotic lesions.

The accumulation of 99mTc-Annexin A5 in atherosclerotic lesions in the

region of the aorta affected by vascular injury was about 9.3-fold greater than

in aortas of the control animals. The mean (±SEM) percent-injected dose

per gram uptake in the aortas with atherosclerotic lesions (0.054± 0.0095%)

was significantly higher than the activity control specimens (0.0058±0.001,

P<0.000). The Annexin A5 uptake was significantly higher in advanced

lesions (AHA type IV lesions), where the incidence of apoptosis was maximal.

Furthermore, the uptake correlated with the extent of macrophage presence

in the plaques. Although the rabbit model of atherosclerosis does not develop

vulnerable lesions as seen in humans, the data indicate that Annexin A5

targets to macrophages in the plaque and is related to plaque apoptosis.

Because human vulnerable atherosclerotic lesions exhibit a great quantity

of activated or apoptotic macrophages, these data suggest that Annexin

A5 may be an imaging marker for plaque vulnerability in patients with

atherosclerotic disease.

Page 9: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

121

Ch

apter 7 | M

olecu

lar Imagin

g of M

acrop

hage C

ell Death

Figure 3. Top left, Ex vivo images of control mice, apoE-/- mice without cholesterol diet and with cholesterol diet with Annexin A5 imaging. No obvious Annexin A5 uptake is seen on images of control animals. Top right, Distinct uptake is observed in the arch and abdominal aorta in the transgenic mice. Quantitative Annexin A5 uptake is provided for normal mice, ApoE-/- and LDLR-/- mice; uptake was highest in cholesterol-fed apoE-/- mice, followed by chow-fed apoE-/- and control mice in lesions at arch, thoracic, or abdominal level. Histopathologic characterization of atherosclerotic lesions including Movat pentachrome staining (×100), Mac-3 antibody staining (×400), a-actin staining (×400), and TUNEL staining (×400) in control, chow-fed LDLR-/-, cholesterol-fed LDLR-/-, chow-fed apoE-/-, and cholesterol-fed apoE-/- mice. Prevalence of Mac-3–positive cells is significantly higher in cholesterol- and chow-fed apoE-

/- and cholesterol-fed LDLR-/- mice than that in chow-fed LDLR-/- mice. Prevalence of SMC is significantly lower compared with macrophages. SMCs are relatively more commonly observed in cholesterol- and chow-fed apoE-/- mice than in cholesterol- or chow-fed LDLR-

/- mice. Apoptotic nuclei in core region are more frequently observed in apoE-/- mice than in LDLR-/- mice and more so in cholesterol-fed animals (arrows). Modified with permission from Isobe et al.14

Page 10: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

122

Figure 4. Noninvasive imaging of vulnerable carotid artery atherosclerotic lesion with radiolabeled Annexin-V. A, Transverse and coronal SPECT images from 1 of the patients who suffered from left sided TIA 3 days before imaging. B, Histopathologic analysis of endarterectomy tissue from the same patient demonstrated significant macrophage infiltration in the neointima with extensive presence of Annexin A5. C, SPECT images from a patient, who had suffered from a right sided TIA 3 months before imaging. No Annexin A5 uptake in the carotid region on either side was shown; however, Doppler ultrasound revealed a severe stenotic lesion on the culprit side. D, Histopathologic analysis of endarterectomy tissue from this patient showed a smooth muscle cell (SMC) rich lesion, and no Annexin A5 presence. Modified with permission from Kietselaer et al.15

Confirmation of Annexin A5 Uptake in Other Animal ModelsThe imaging of apoptotic cell death has been demonstrated in various

other animal models of atherosclerosis, including transgenic mice such as

apolipoprotein E– deficient (apoE-/-) and low-density-lipoprotein receptor–

deficient (LDLR-/-) mice, which develop spontaneous atherosclerotic lesions,

and the swine model of coronary atherosclerosis produced by endothelial

injury and high-cholesterol diet. Radiolabeled Annexin A5 was successfully

used for noninvasively image imaging in transgenic apoE-/- and LDLR-/- mice

with or without high-cholesterol diet.14 The Annexin A5 uptake was compared

with normal wild-type (C57BL/6) mice of the same genetic background.

Noninvasive imaging was performed using micro–SPECT-CT, followed by

ex vivo imaging of the explanted aorta and quantitative assessment of the

radiotracer localization by gamma counting of the percentage injected dose

per gram (%ID/g) annexin uptake (Figure 3). Subsequently, histological and

immunohistochemical characterization of the aortic samples was performed.

Aortic lesions were clearly visualized noninvasively by micro-SPECT, and

produced by endothelial injury and high-cholesterol diet.Radiolabeled Annexin A5 was successfully used for nonin-vasively image imaging in transgenic apoE�/� and LDLR�/�

mice with or without high-cholesterol diet.14 The Annexin A5uptake was compared with normal wild-type (C57BL/6) miceof the same genetic background. Noninvasive imaging wasperformed using micro–SPECT-CT, followed by ex vivoimaging of the explanted aorta and quantitative assessment ofthe radiotracer localization by gamma counting of the per-centage injected dose per gram (%ID/g) annexin uptake(Figure 3). Subsequently, histological and immunohisto-chemical characterization of the aortic samples was per-formed. Aortic lesions were clearly visualized noninvasivelyby micro-SPECT, and aorta calcification was detected bymicro-CT. The quantitative uptake of Annexin A5 washighest in the cholesterol-fed apoE�/� mice, followed by thenormal chow-fed apoE�/�, the cholesterol-fed LDLR�/�, thechow-fed LDLR�/�, and the disease control normal mice.The histological extent of atherosclerosis paralleled radio-tracer uptake, and immunohistochemical studies revealed asignificant correlation between radiotracer uptake and bothmacrophage infiltration and the extent of apoptosis. Intrave-nously injected biotinylated Annexin A5 localized in apoptot-ic and nonapoptotic macrophages.

One of the major challenges is to develop a noninvasiveimaging technology that is able to detect vulnerable plaquesin the coronary tree. In a study in a swine model ofatherosclerosis it was shown that the visualization of AnnexinA5 uptake in the coronary tree is feasible using nuclearimaging.15 In male swine, atherosclerosis was produced byhigh-fat diet combined with vascular injury to the coronaryvessels. After an average of 2 months, the swine underwentcoronary angiography, followed by radiolabeled Annexin A5and SPECT imaging. After in vivo imaging the animals were

euthanized, the coronary vessels imaged by autoradiographyand counted in the well counter. Immunostaining was per-formed for �-actin and caspase 3. Histopathology showedthat the coronary lesions were early, mild, and mainlycharacterized by smooth muscle cells. Out of the abnormalvessels, 60% showed focal uptake of Anexin A5 in vivo,which corresponded to uptake on autoradiography. The countratio of the injured vessel compared to control vessel wasalmost 3:1 for positive scans and 1.3:1 for negative scans.Scans positive for Annexin A5 uptake correlated withcaspase-positive staining. These data provide proof of con-cept that noninvasive detection of the Annexin A5 uptake inatherosclerotic lesions of the coronary tree is feasible.

Clinical Feasibility of Apoptosis ImagingIn a first attempt to evaluate apoptosis imaging in a clinicalsetting, Annexin A5 imaging was performed in patients withrecent (n�2) or remote history (n�2) of transient ischemicattack (TIA), respectively, 1 to 3 days before the scheduledcarotid endarterectomy.16 Six hours after injection of 600 to800 MBq 99mTc-Annexin A5, SPECT imaging was per-formed. The 2 patients who had suffered from a TIA 3 to 4days before imaging showed clear Annexin A5 uptake (Fig-ure 4) in the area of the culprit carotid artery lesion, as wasidentified by ultrasound. Histopathologic assessment of theendarterectomy tissue showed vulnerable plaque morphol-ogy, including substantial macrophage infiltration and in-traplaque hemorrhage. Immunohistochemical evaluationshowed binding of Annexin A5 predominantly to macro-phages. The 2 patients, who had a remote history of TIA 3 to4 months before imaging, showed no Annexin A5 uptake inthe carotid artery on SPECT imaging. These patients had asimilar carotid lesion as compared to the patients with arecent history of TIA. These patients had been treated with

Figure 4. Noninvasive imaging of vulnerable carotid artery atherosclerotic lesion with radiolabeled Annexin-V. A, Transverse andcoronal SPECT images from 1 of the patients who suffered from left sided TIA 3 days before imaging. B, Histopathologic analysisof endarterectomy tissue from the same patient demonstrated significant macrophage infiltration in the neointima with extensivepresence of Annexin A5. C, SPECT images from a patient, who had suffered from a right sided TIA 3 months before imaging. NoAnnexin A5 uptake in the carotid region on either side was shown; however, Doppler ultrasound revealed a severe stenotic lesionon the culprit side. D, Histopathologic analysis of endarterectomy tissue from this patient showed a smooth muscle cell (SMC)rich lesion, and no Annexin A5 presence. Modified with permission from Kietselaer et al.17

Laufer et al Macrophage Cell Death and Plaque Vulnerability 1035

at Universiteitsbibliotheek Utrecht on February 3, 2015http://atvb.ahajournals.org/Downloaded from

Page 11: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

123

Ch

apter 7 | M

olecu

lar Imagin

g of M

acrop

hage C

ell Death

aorta calcification was detected by micro-CT. The quantitative uptake of

Annexin A5 was highest in the cholesterol-fed apoE-/- mice, followed by the

normal chow-fed apoE-/-, the cholesterol-fed LDLR-/-, the chow-fed LDLR-/-, and

the disease control normal mice. The histological extent of atherosclerosis

paralleled radiotracer uptake, and immunohistochemical studies revealed

a significant correlation between radiotracer uptake and both macrophage

infiltration and the extent of apoptosis. Intravenously injected biotinylated

Annexin A5 localized in apoptotic and nonapoptotic macrophages.

One of the major challenges is to develop a noninvasive imaging technology

that is able to detect vulnerable plaques in the coronary tree. In a study in

a swine model of atherosclerosis it was shown that the visualization of

Annexin A5 uptake in the coronary tree is feasible using nuclear imaging.16

In male swine, atherosclerosis was produced by high-fat diet combined with

vascular injury to the coronary vessels. After an average of 2 months, the

swine underwent coronary angiography, followed by radiolabeled Annexin A5

and SPECT imaging. After in vivo imaging the animals were euthanized, the

coronary vessels imaged by autoradiography and counted in the well counter.

Immunostaining was performed for a-actin and caspase 3. Histopathology

showed that the coronary lesions were early, mild, and mainly characterized

by smooth muscle cells. Out of the abnormal vessels, 60% showed focal uptake

of Annexin A5 in vivo, which corresponded to uptake on autoradiography.

The count ratio of the injured vessel compared to control vessel was almost

3:1 for positive scans and 1.3:1 for negative scans. Scans positive for Annexin

A5 uptake correlated with caspase-positive staining. These data provide

proof of concept that noninvasive detection of the Annexin A5 uptake in

atherosclerotic lesions of the coronary tree is feasible.

Page 12: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

124

Figure 5. Annexin A5 uptake within abdominal aortas in various study groups. Ex vivo images of explanted aortas showed no Annexin A5 uptake in normal aorta (N-Annexin A5), but intense Annexin A5 uptake within abdominal aortas in rabbits fed high-cholesterol (HC-Annexin A5) diet for 4 months. There was only a negligible Annexin A5 uptake seen in diet withdrawal (DW-Annexin A5) and statin (Rx-Annexin A5) therapy group. Similar to the control group, no Annexin A5 uptake was observed in rabbits receiving high-cholesterol diet for 4 months and imaged with mutant Annexin A5 (HC+mAnnexin A5). Bar graphs (below) show quantitative Annexin A5 uptake within abdominal aortas in various study groups, represented as mean ±SD %ID/g; Annexin A5 uptake corresponds to ex vivo images as above. Maximum Annexin A5 uptake (%ID/g) was seen in HC animals (P=0.0001) compared with N animals (controls). Uptake was significantly lower in DW and RX groups than in HC animals. No significant difference was observed between animals in DW and Rx groups or between N and HC-mAnnexin A5 groups (P was not significant [NS]). Quantitative results confirmed ex vivo imaging observations. Photomicrographs in right panel of abdominal atherosclerotic lesions are stained for histology (with Movat pentachrome stain), macrophages (with Ram-11), alpha smooth muscle actin (with HHF35), and apoptosis (ISEL) from untreated atherosclerotic animal of HC-Annexin A5 (top), DW (middle), and Rx (bottom) groups. HC group demonstrates AHA type IV lesion with well-formed necrotic core in neointimal region. Staining with antibody specific for macrophages (CD68; RAM-11) shows widespread presence of macrophage infiltration in neointima. Staining with antibody against actin isotypes a and ß (HHF-35) shows absence of SMC in neointima. SMC staining is visible only in normal medial layer of atherosclerotic vessel. DNA fragmentation staining (blue–black nuclei; arrow) by in situ end labeling (ISEL) and methyl green counterstaining demonstrate extensive apoptosis of macrophages. Stabilization of atherosclerotic lesions in diet withdrawal and statin therapy groups is evident by increased SMC content, decreased macrophage populations, and decreased apoptosis after diet withdrawal (middle row) and statin therapy (bottom row). The statin group is significantly superior to the DW group. (Magnification ×100 for top row and ×35 for the middle and bottom). Modified with permission from Hartung et al.17

Page 13: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

125

Ch

apter 7 | M

olecu

lar Imagin

g of M

acrop

hage C

ell Death

Clinical Feasibility of Apoptosis ImagingIn a first attempt to evaluate apoptosis imaging in a clinical setting, Annexin

A5 imaging was performed in patients with recent (n=2) or remote history

(n=2) of transient ischemic attack (TIA), respectively, 1 to 3 days before the

scheduled carotid endarterectomy.18 Six hours after injection of 600 to 800

MBq 99mTc-Annexin A5, SPECT imaging was performed. The 2 patients who had

suffered from a TIA 3 to 4 days before imaging showed clear Annexin A5 uptake

(Figure 4) in the area of the culprit carotid artery lesion, as was identified by

ultrasound. Histopathologic assessment of the endarterectomy tissue showed

vulnerable plaque morphology, including substantial macrophage infiltration

and intraplaque hemorrhage. Immunohistochemical evaluation showed

binding of Annexin A5 predominantly to macrophages. The 2 patients, who

had a remote history of TIA 3 to 4 months before imaging, showed no Annexin

A5 uptake in the carotid artery on SPECT imaging. These patients had a similar

carotid lesion as compared to the patients with a recent history of TIA. These

patients had been treated with statins and antiplatelet agents after the acute

event. Endarterectomy specimens from the patients with a remote history

demonstrated stable plaque characteristics with nonsubstantial macrophage

infiltration and no intraplaque hemorrhage; only slight Annexin binding was

observed on immunohistochemical analysis. These pilot data demonstrate

that 99mTc-Annexin A5 targets to macrophage-rich lesions of plaques with

vulnerable plaque morphology. In addition, the data show that noninvasive

Annexin A5 imaging of vulnerable atherosclerotic lesions is clinically feasible

and may recognize patients at risk for acute vascular events.

Page 14: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

126

Figure 6. The extent of apoptosis in various animal groups represented by quantitative Annexin A5 uptake and histological characterization of untreated and broad caspase-inhibited animals. Modified with permission from Sarai et al.19 The left panel demonstrates the uptake of Annexin A5 in abdominal aortas. Disease control normal animals (N) with no atherosclerotic lesions are represented by an open bar, and the remaining atherosclerotic animals by solid bars. Atherosclerotic animals imaged with mutant Annexin were used as tracer controls (gray bar), and the remaining atherosclerotic animals were either untreated (black bar) or treated with various caspase inhibitors (colored bars). Broad and caspase-3 inhibition bars are shown in pink, caspase-8 and -9 inhibitors differentiating between mitochondrial and extramitochondrial pathways are in blue, and capase-1–specific inhibition is represented by a green solid bar. The statistical significance of caspase inhibition, in comparison with the disease control and untreated atherosclerotic animals, is shown above and below the bars, respectively. The uptake is significantly higher in the abdominal aortas of untreated atherosclerotic animals compared with that in the disease and tracer control groups. Treatment with a broad caspase inhibitor or selective caspase-1, -9, or -3 inhibitor significantly reduced the apoptotic activity as represented by lower Annexin uptake. On the other hand, selective caspase-8 inhibition did not affect apoptosis. The right panel compares histological characterization of atherosclerotic lesions in atherosclerotic untreated animals and atherosclerotic animals receiving the broad caspase inhibitor. Movat pentachrome (M5Ch), smooth muscle cell (a-actin), and macrophage (Ram-11) staining (×200) in untreated atherosclerotic animals (left) and broad caspase inhibitor-treated animals (right) demonstrate similar cholesterol crystal-rich necrotic core, foam cell-rich, and smooth muscle-deficient lesions. The morphological characteristics of the lesions in untreated and caspase-treated atherosclerosis are unchanged. However, terminal deoxyribonucleotide transferase-mediated nick-end labeling (TUNEL) reveals evidence of marked apoptotic nuclei in untreated atherosclerotic animals but marked resolution in the atherosclerotic lesions in caspase-inhibited animals.

Page 15: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

127

Ch

apter 7 | M

olecu

lar Imagin

g of M

acrop

hage C

ell Death

Acute and Chronic Resolution of Apoptosis in Atherosclerotic PlaquesAlthough apoptosis within atherosclerotic plaques is associated with

plaque vulnerability and rupture, it has been proposed17,19 that the role of

inhibition of the apoptotic process can be clarified by evaluating the impact

of interventions which result in favorable outcomes. As such the influence

of dietary modification and statin therapy on the incidence of apoptosis

in experimental atherosclerotic lesions was seen. A group of rabbits with

experimental atherosclerosis17 was randomized as follows: high-cholesterol

diet for 4 months (untreated atherosclerotic group), high-cholesterol diet for

3 months, and normal chow diet for 1 month (diet withdrawal group) and

high-cholesterol diet for 4 months and simvastatin orally every day of the

last month (statin therapy group). Radiolabeled Annexin A5 was used for the

noninvasive detection of the extent of apoptosis. For control experiments,

either radiolabeled mutant Annexin A5 was used in the hypercholesterolemic

rabbits or Annexin A5 in normal rabbits receiving normal chow. Quantitative

Annexin 5 uptake in the abdominal aorta was determined and compared

with the histological and immunohistochemical characteristics of the

atherosclerotic lesions. Maximum Annexin A5 uptake, as expected, was

observed in the untreated atherosclerotic animals. The uptake was substantially

reduced in the diet withdrawal and statin therapy groups. The plaques in

the untreated high-cholesterol group demonstrated advanced atherosclerotic

lesions, which were substantially resolved, including the resolution of

macrophage infiltration and an increase in smooth muscle cell content in

the diet withdrawal and statin groups. There was a marked reduction in the

apoptosis of macrophages. No significant uptake of Annexin A5 or mutant

Annexin A5 was seen in rabbits on the normal chow diet or atherosclerotic

rabbits, respectively (Figure 5). Because apoptosis of macrophages contributes

significantly to plaque rupture, it is logical to presume that acute repression

of apoptosis may be of therapeutic benefit in acute coronary events. Such

a proposal may be of added value because resolution of apoptosis has also

been shown to be beneficial in limiting myocardial injury in experimental

myocardial infarction. Resolution of apoptosis by statins takes at least 4

weeks, a period wherein most recurrences of ischemic events are seen.

As such, the role of caspase inhibitors on acute resolution of apoptosis in

atherosclerotic lesions has been evaluated by imaging with Annexin A5. For

this purpose, a large number of atherosclerotic rabbits received broad caspase,

caspase-1, caspase-3, caspase-8, or caspase-9 inhibitors. These caspase-treated

animals were compared with the magnitude of apoptosis in the untreated

animal group and normal disease control animals. Radiolabeled Annexin A5

Page 16: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

128

traced the atherosclerotic lesions best in untreated atherosclerotic rabbits.

Quantitative Annexin A5 uptake, defined as the percent of injected dose

per gram of abdominal aorta tissue, was significantly higher in untreated

atherosclerotic animals compared with the normal rabbits or atherosclerotic

rabbits receiving mutant Annexin. Among all caspase inhibitor-treated

rabbits, uptake was 40% lower than the untreated atherosclerotic animals. In

fact, the uptake was significantly lower in the rabbits receiving broad caspase

or caspase-1, -3, or -9 inhibitors. Caspase-8 inhibitor did not affect apoptosis.

On histological characterization, a substantial decrease in macrophage

apoptosis was observed in caspase-inhibited animals. Because caspase-8

inhibition had no significant effect on apoptosis, and caspase-9 inhibition

significantly blocked it, the apoptosis of foam cells is likely to involve the

mitochondrial pathway and not the death receptor pathway (Figure 6). In

addition, the caspase-1 inhibitor also blocked apoptosis to a level similar to

the broad caspase and specific caspase-3 inhibitors, suggesting that caspase-1

may also be involved in apoptosis in atherosclerotic lesions; this confirms

the initial observations of caspase-1 activations in the victims of sudden

death attributable to plaque rupture.10 Activation of caspase-1 in apoptosis

in atherosclerosis in the absence of caspase-8 involvement suggests its

interaction with mitochondrial pathway of apoptosis. Such interaction has

recently been demonstrated in traumatic brain injuries, wherein increased

levels of cytochrome c, caspase-3, and caspase-1 have been observed in the

cerebrospinal fluid, and caspase-3 and -1 activation in brain tissue.20 It is likely

that reactive oxygen species generation, which is downstream of caspase-1,

plays an important role in caspase-1 based apoptotic cascade.

ConclusionsMolecular imaging strategies have been developed to detect apoptosis,

an important pathological substrate of plaque instability. Some of these

techniques have been used in clinical settings and demonstrate the possibility

of their application in risk-stratification of asymptomatic subjects falling in the

high-risk categories (more than 20% 10-year risk). In addition to the potential

diagnostic application, molecular imaging also helps to better understand the

pathogenesis of vulnerable plaques because it is the only strategy that can be

used for the study of subcellular events in living organisms.

Page 17: 115 - Vrije Universiteit Amsterdam 7.pdfof a vulnerable plaque, resulting in local thrombus formation and acute disruption in the coronary blood supply in the affected area.5 These

129

Ch

apter 7 | M

olecu

lar Imagin

g of M

acrop

hage C

ell Death

References

1. Braunwald, E. Epilogue: what do clinicians expect from imagers? J. Am. Coll. Cardiol. 47, C101–3 (2006).2. Zethelius, B. et al. Use of multiple biomarkers to improve the prediction of death from cardiovascular

causes. N. Engl. J. Med. 358, 2107–2116 (2008).3. Sanz, J. & Fayad, Z. A. Imaging of atherosclerotic cardiovascular disease. Nature 451, 953–957 (2008).4. Osborn, E. A. & Jaffer, F. A. Advances in molecular imaging of atherosclerotic vascular disease. Curr.

Opin. Cardiol. 23, 620–628 (2008).5. Burke, A. P. et al. Coronary risk factors and plaque morphology in men with coronary disease who died

suddenly. N. Engl. J. Med. 336, 1276–1282 (1997).6. Virmani, R., Burke, A. P., Kolodgie, F. D. & Farb, A. Vulnerable plaque: the pathology of unstable coronary

lesions. J Interv Cardiol 15, 439–446 (2002).7. Virmani, R., Burke, A. P., Farb, A. & Kolodgie, F. D. Pathology of the vulnerable plaque. J. Am. Coll. Cardiol.

47, C13–8 (2006).8. van Engeland, M., Nieland, L. J., Ramaekers, F. C., Schutte, B. & Reutelingsperger, C. P. Annexin V-affinity

assay: a review on an apoptosis detection system based on phosphatidylserine exposure. Cytometry 31, 1–9 (1998).

9. Koopman, G. et al. Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis. Blood 84, 1415–1420 (1994).

10. Kolodgie, F. D. et al. Localization of apoptotic macrophages at the site of plaque rupture in sudden coronary death. Am. J. Pathol. 157, 1259–1268 (2000).

11. Libby, P. What have we learned about the biology of atherosclerosis? The role of inflammation. AJC 88, 3J–6J (2001).

12. Kolodgie, F. D. et al. Targeting of apoptotic macrophages and experimental atheroma with radiolabeled annexin V: a technique with potential for noninvasive imaging of vulnerable plaque. Circulation 108, 3134–3139 (2003).

13. Mallat, Z. et al. Shed Membrane Microparticles With Procoagulant Potential in Human Atherosclerotic Plaques. Circulation 99, 348–353 (1999).

14. Isobe, S. et al. Noninvasive imaging of atherosclerotic lesions in apolipoprotein E-deficient and low-density-lipoprotein receptor-deficient mice with annexin A5. J. Nucl. Med. 47, 1497–1505 (2006).

15. Kietselaer, B. L. J. H. et al. Noninvasive detection of plaque instability with use of radiolabeled annexin A5 in patients with carotid-artery atherosclerosis. N. Engl. J. Med. 350, 1472–1473 (2004).

16. Johnson, L. L., Schofield, L., Donahay, T., Narula, N. & Narula, J. 99mTc-annexin V imaging for in vivo detection of atherosclerotic lesions in porcine coronary arteries. J. Nucl. Med. 46, 1186–1193 (2005).

17. Hartung, D. et al. Resolution of apoptosis in atherosclerotic plaque by dietary modification and statin therapy. J. Nucl. Med. 46, 2051–2056 (2005).

18. Kietselaer, B. L. J. H. et al. Noninvasive detection of programmed cell loss with 99mTc-labeled annexin A5 in heart failure. J. Nucl. Med. 48, 562–567 (2007).

19. Sarai, M. et al. Broad and specific caspase inhibitor-induced acute repression of apoptosis in atherosclerotic lesions evaluated by radiolabeled annexin A5 imaging. J. Am. Coll. Cardiol. 50, 2305–2312 (2007).

20. Satchell, M. A. et al. Cytochrome c, a biomarker of apoptosis, is increased in cerebrospinal fluid from infants with inflicted brain injury from child abuse. J. Cereb. Blood Flow Metab. 25, 919–927 (2005).