Guidelines Clinical Practice Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Cardiol. Jul 26, 2011; 3(7): 219-229
Published online Jul 26, 2011. doi: 10.4330/wjc.v3.i7.219
Non-invasive detection of vulnerable coronary plaque
Faisal Sharif, Department of Cardiology, Regional Hospital Galway, and Regenerative Medicine Institute, National University of Ireland Galway, County Galway, Ireland
Derek G Lohan, Department of Radiology, Regional Hospital Galway, County Galway, Ireland
William Wijns, Department of Cardiology, Cardiovascular Centre, OLV Ziekenhuis, Aalst, 9300, Belgium
Author contributions: All authors contributed equally to the paper.
Correspondence to: Dr. Faisal Sharif, Senior Lecturer Regenerative Medicine/Consultant Cardiologist, Department of Cardiology, Regional Hospital Galway, and Regenerative Medicine Institute, National University of Ireland Galway, County Galway, Ireland. faisal.sharif@nuigalway.ie
Telephone: +353-91-542186 Fax: +353-91-527197
Received: March 28, 2011
Revised: June 16, 2011
Accepted: June 23, 2011
Published online: July 26, 2011

Abstract

Critical coronary stenoses have been shown to contribute to only a minority of acute coronary syndromes and sudden cardiac death. Autopsy studies have identified a subgroup of high-risk patients with disrupted vulnerable plaque and modest stenosis. Consequently, a clinical need exists to develop methods to identify these plaques prospectively before disruption and clinical expression of disease. Recent advances in invasive and non-invasive imaging techniques have shown the potential to identify these high-risk plaques. Non-invasive imaging with magnetic resonance imaging, computed tomography and positron emission tomography holds the potential to differentiate between low- and high-risk plaques. There have been significant technological advances in non-invasive imaging modalities, and the aim is to achieve a diagnostic sensitivity for these technologies similar to that of the invasive modalities. Molecular imaging with the use of novel targeted nanoparticles may help in detecting high-risk plaques that will ultimately cause acute myocardial infarction. Moreover, nanoparticle-based imaging may even provide non-invasive treatments for these plaques. However, at present none of these imaging modalities are able to detect vulnerable plaque nor have they been shown to definitively predict outcome. Further trials are needed to provide more information regarding the natural history of high-risk but non-flow-limiting plaque to establish patient specific targeted therapy and to refine plaque stabilizing strategies in the future.

Key Words: Atherosclerotic plaque, Magnetic resonance imaging, Multidetector row computed tomography, Single photon emission computed tomography



INTRODUCTION

Technological advances in cardiovascular imaging in parallel with significant development in biomedical science has changed the way we assess coronary atherosclerosis. Interestingly, two very separate but intermingled concepts have emerged. In the first concept, regardless of the extent of coronary atheroma and luminal stenosis (as observed by coronary angiography), coronary pressure measurement is used to evaluate functional ischemia of the myocardium supplied by the stenotic epicardial vessel. The decision to revascularize a stenotic epicardial vessel is based on the presence or absence of a flow-limiting trans-stenotic coronary pressure gradient. The usefulness of this concept has been clinically validated[1-3]. The second concept evolved after reports that claimed that most acute coronary events and coronary thromboses form on angiographically non-obstructive atheroma[4-6]. Standard coronary angiography often fails to identify the culprit lesion of non trans-mural acute myocardial infarction (AMI)[7]. In addition, the plaque burden, its delineation and constituents cannot be assessed by coronary angiography. These potentially lethal but mechanically non-obstructive plaques were later labeled as high-risk plaques or vulnerable plaques[8].

The extent of underlying plaque burden causing plaque rupture is a contentious issue. Early, and in fact some recent studies, have shown that coronary thrombosis and AMI are directly proportional to the severity of the coronary stenosis[9]. However, in contrast to this notion, there is also substantial evidence in the literature to believe that coronary thrombosis can develop in as many as two-thirds of cases with non-obstructive, high-risk vulnerable coronary plaques[4-6]. Regardless of the extent of underlying coronary atheroma leading to acute coronary syndromes (ACS), it is well documented that high-risk vulnerable plaques exist and are prone to rupture[10]. These plaques are generally treated conservatively and the possibility of adaptive remodeling is routinely overlooked. Most currently available diagnostic tests are unable to predict the risk of thrombosis associated with any particular lesion in the coronary arteries. Consequently, a clinical need exists to develop new techniques that are capable of identifying vulnerable plaques before disruption occurs.

VULNERABLE PLAQUE

The composition of atherosclerotic plaque is heterogeneous by nature and contains: (1) fibrocellular components [extracellular matrix and smooth muscle cells (SMCs)]; (2) lipid-cellular components (crystalline cholesterol and cholesterol esters mixed with macrophages); (3) thrombotic components (platelets and fibrin); and (4) calcium[11-14]. Vulnerable plaques that result in rupture have been now well described as thin cap fibroatheroma (TCFA). The term vulnerable plaque was first instituted by Muller et al[10]. These plaques contain extensive necrotic lipid core and a thin fibrous cap (< 65 μm)[14,15]. The TCFA that is prone to rupture is deficient in SMCs but contains type 1 collagen and infiltrating active macrophages. These macrophages release matrix metalloproteinases (MMPs) 1, 8 and 13 that weaken the fibrous cap and consequently result in rupture[16]. About 65%-70% of all coronary thrombi result from plaque rupture. Although the term thin (< 65 μm) fibrous cap is generally accepted to define vulnerable plaques, some conflicting reports have used higher thresholds (> 200 μm) to describe vulnerable plaques[17,18]. It is important to realize that that the pathological description of vulnerable plaque lacks physiological data. The histological observations are made on static and inert tissue while plaque rupture is a more dynamic process as recently reported by Abela et al[19]. Cholesterol expands in volume when crystallizing from a liquid to a solid, potentially leading to rupture. In addition to plaque rupture, plaque erosion and calcified nodules can also result in thrombotic plaque disruption[20]. Erosive plaques usually have intimal thickening and are composed of fibrotic tissue with a thick fibrous cap. The thick fibrous cap in contrast to a thin cap contains an abundance of SMCs, proteoglycans and type III collagen, but very few inflammatory cells[20]. Determination of plaque characteristics is important to appreciate the pathophysiological process of atherothrombosis, and may also provide us with a means to establish risk assessment for individual plaques in individual patients. Accordingly, imaging modalities are required to reliably evaluate plaque composition and thereby allow implementation of treatment strategies to prevent adverse coronary events.

INVASIVE IMAGING MODALITIES

Invasive techniques to determine the vulnerable plaques have received more attention. In particular three different aspects of vulnerable plaques have been investigated using different invasive technologies. The first technique focuses on imaging the microanatomy of the plaque to identify the plaque components, and includes high frequency intra-vascular ultrasound (IVUS), virtual histology-IVUS®, intravascular optical coherence tomography and intravascular magnetic resonance imaging (MRI). The second set of techniques is directed at measuring metabolic activity of the plaque to predict the risk of plaque disruption, and includes intravascular thermography and elastography. The third technique relies on measuring plaque chemical composition and detailed characterization by employing near infrared reflectance or Raman spectroscopy. Although it is likely that initial prospective identification of vulnerable plaque will first be achieved by one of these competing intra-coronary technologies, several drawbacks exist in the widespread use of these technologies. Most invasive imaging modalities are novel and therefore require specific training and highly skilled staff, they are expensive to run and consequently are not feasible for routine clinical application. Readers are directed to specific reviews on invasive imaging to detect plaque[21]. This article will focus on current non-invasive modalities available to detect vulnerable plaque with especial focus on computed tomography (CT), MRI and positron emission tomography (PET) imaging.

NON-INVASIVE IMAGING TECHNIQUES

By their very nature, invasive imaging techniques are undesirable, with a lower level of patient acceptability than non-invasive alternatives, and thus face significant hurdles if they are to be accepted into routine clinical practice[22-26]. Non-invasive imaging modalities, namely CT, MRI and scintigrahic nuclear imaging techniques may provide an alternative to invasive imaging, and have shown considerable promise in recent studies[27-32]. Continued technological advances have bridged the gap between the accuracies of these non-invasive “modern” techniques and their “traditional” invasive counterparts, with the result that the former have been accepted into routine practice for an ever-increasing spectrum of clinical conditions and indications. One of the ways to achieve increased sensitivity is through the use of nanoparticle-based molecular imaging[27,33,34]. Nanoparticle-enhanced MRI or single photon emission CT (SPECT) can identify a thrombus by detecting fibrin, confirm the presence of an inflammatory process by detecting leukocyte and macrophage infiltration, and recognize plaque angiogenesis by detecting specific integrins involved in the formation of new blood vessels[35-38]. In addition, molecules can also be used to label specific cells both in vivo and ex vivo for imaging, for example, stem cells for cardiac regeneration and lymphocytes for specific tumors. Furthermore, and in contrast to invasive imaging, molecular imaging may also be used to provide local treatments by targeting with specific therapeutic molecules. Finally, nanoparticle-enhanced MRI and PET scanners may play an important role in the development of new drugs by providing an in vivo assessment of the desired molecular effect of the investigational drug. This information could be very useful when deciding whether expensive phase III trials for new pharmaceutical agents are justified.

The "ideal" non-invasive imaging modality

In order to understand the relative strengths and weaknesses of modalities in current practice, it is important to first determine the characteristics of the “ideal” non-invasive imaging modality (Table 1). Such a non-invasive modality would combine patient acceptability, acceptable clinical indications, safety, speed and high technical specification to produce unequivocal objective data upon which future management could be confidently based. It is apparent that modern techniques, while far superior to their predecessors, fall considerably short of these model standards.

Table 1 Characteristics of the “ideal” non-invasive imaging modality.
Patient-related factorsTechnical factors
Absence of ionizing radiationRapid image acquisition
Spacious (minimize claustrophobia)High spatial resolution
Suitable for all (not precluded by aneurysm clips/pacemaker leads)High contrast resolution
Absence of breath-holdingHigh temporal resolution
Administration of extrinsic contrast agents unnecessaryElectrocardiogram and respiratory gating
Wide range of clinical indicationsNot limited by cardiac arrhythmia
Provides both anatomic and metabolic information
Reproducible
Accurate
MRI

Rapid innovation and development in MR technology has allowed for widespread acceptance of cardiovascular MRI as a valuable non-invasive in vivo imaging modality for assessment of myocardial contractility, viability and valvular function. This is due in no small part to the commercial availability of increasingly more robust coil and gradient technology in combination with novel pulse sequence design. This technique exploits differences in proton density, proton mobility, water content, chemical composition, molecular motion and diffusion to allow for exquisite soft tissue depiction, a reflection of the high contrast resolution of MRI. Most versatile at field strengths of 1.5 Tesla, this modality fulfils several of the desirable characteristics outlined in Table 1, including absence of ionizing radiation and requirement for breath-holding or extrinsic contrast agents as well as facilitating electrocardiogram (ECG) gating with high spatial and contrast resolution.

Despite the technological advances referred to above, coronary imaging has until recent times been beyond the capabilities of MRI. This reflects the diminutive size of the coronary vessels and thus poor contrast/signal to noise ratios, a short-lived “rest period” during diastole during which coronary motion is minimized, temporal restrictions imposed by the patient’s ability to suspend respiration and the resultant limited spatial resolution attainable during that time period. Several of these issues have been addressed with considerable success by the introduction of T2 preparation navigator-gated and -corrected 3-dimensional segmented techniques which, although time-consuming, allow for volumetric imaging of the coronary arteries in their entirety with subsequent multiplanar reconstruction. However, coronary MR angiographic techniques remain technically demanding and suboptimal to merit widespread acceptance into the clinical arena.

HIGH RESOLUTION MRI FOR CORONARY PLAQUE IMAGING

MR angiographic evaluation of coronary arterial luminal patency has traditionally proven challenging for the reasons outlined above. One can appreciate therefore why attempts at coronary plaque imaging add an additional level of complexity, further stressing the boundaries of MRI capability. High resolution imaging using T1, T2 and proton density weighting allows for identification of calcium which is hypointense on all imaging sequences, lipid which is T1- and PD-hyperintense and T2-hypointense, and fibrous tissue which produces increased signal intensity on PD-weighted imaging, while it is isointense on T1 and isointense-to-hyperintense on T2-weighted imaging. These multi-contrast signal intensity “signatures” allow for characterization of various plaque components and plaque morphology on high-resolution imaging and therefore assessment of plaque vulnerability. In order to have a null signal from blood within the adjacent coronary lumen and thereby maximize contrast resolution, real-time respiratory navigated black blood fast spin echo sequences are generally utilized. Real-time slice position correction has also been employed. The accuracy of this technique has been histologically confirmed ex vivo[39]. This technique has also been employed to assess coronary wall thickness, with validation in both animal models and humans[40-43]. Fayad et al[41] reported positive remodeling and significant coronary wall thickening in patients with coronary artery disease in comparison with control patients. Such approaches may prove useful for non-invasive coronary plaque burden measurement in the absence of ionizing radiation exposure.

MOLECULAR MR FOR PLAQUE IMAGING

Evaluation of coronary plaque at the molecular level requires the addition of targeted contrast agents (Table 2). Paramagnetic gadolinium chelates are the most commonly used extracellular contrast agent for MRI. Although in nature this metal has a short half-life, this has been compensated by novel gadolinium constructs with albumin, high-density lipoprotein and liposomes[27,33,44-47]. In addition to a longer half-life, these new generation gadolinium chelates have improved affinity for adjacent protons, allowing superior imaging of the adjoining tissue. This affinity for protons is referred to as relaxivity time, R1 for longitudinal relaxivity (adjacent tissue appears bright) and R2 for transverse relaxivity (adjacent tissue appears dark).

Table 2 Selected molecular imaging agents in cardiovascular disease.
Biological processAgentTargetImaging platform
Plaque rupture/thrombosisGadolinium carrying perfluorocarbon and peptides (EP-2104R)FibrinMRI
Phage display nanoparticles generated by CEST 99mTc-apcitideGlycoprotein IIb/IIIa receptorSPECT
Inflammation/apoptosisMNP + specific antibodiesE-selectinMRI
Monocrystalline iron oxideICAM/VCAMPET
Cross-linked iron oxide 18FDGMacrophages glucose transporter-1, hexokinaseSPECT
99mTc-annexinPhosphatidylserine/caspasesSPECT
99mTc-interleukin-2Lymphocytes
AngiogenesisMNP + specific antibody 99mNC100692VCAM-1/integrin αvβ3MRI
Myocardial infarctionMNP 111Indium oxideStem cell labelingMRI/SPECT

Magnetic iron oxide is another class of paramagnetic nanoparticle (MNP) that has been used as a molecular agent for detection of plaque characteristics. Newer generations of MNP [termed monodisperse iron oxide (MION)] are pretreated with polymer coatings that offer several advantages including better in vivo stability and the ability to target multiple molecules by allowing stable conjugation of a variety of ligands to the nanoparticle. In addition, a cross-linked derivative of MION (CLIO) can be conjugated to near infrared fluorochromes to allow dual modality imaging with fluorescence microscopy and MRI[48,49]. These magnetofluorescent nanoparticle have been used to target specific molecules in vivo[49-52]. Furthermore, a longer half-life, a high relaxivity time and a small diameter allow this nanoparticle to be a useful molecular agent for plaque and myocardial imaging[53-56]. In addition to imaging static cellular markers, MNPs can be used to identify dynamic cellular targets (specific enzymes, e.g. proteases, oxidases) by using novel magnetic relaxation switches[57,58]. These magnetic relaxation switches produce a change in relaxivity time (R2) of the nanoparticle by undergoing reversible modification in structure in the presence of a specific enzyme, which is then detected by T2-weighted MRI[57,58]. These magnetic relaxation switches hold significant potential for identifying a large set of proteins relevant to clinical cardiology including troponin, brain natriuretic peptide and C-reactive protein.

After administration, the molecule-targeting nanoparticles can approach the plaque either through the lumen of the coronary artery or through the vasa vasorum in the outer vessel wall. These molecular agents can detect cell surface markers and therefore identify cells of interest. The cell surface markers are detected by attaching a targeted ligand to the nanoparticle, while cells are identified after cellular uptake and internalization of treated nanoparticles. To target and identify specific peptides or proteins, phage display screening libraries are normally used. Molecular MR approaches to image vulnerable plaque has focused on plaque thrombosis, plaque lipid content, plaque inflammation and plaque angiogenesis. To date, these molecular agents have successfully targeted several plaque components, including fibrin, cellular markers, e.g. vascular cellular adhesion molecule (VCAM), and angiogenesis markers, e.g. integrin αvβ3.

Imaging targets of plaque rupture

Fibrin deposition on the plaque surface is the first step after endothelial disruption following plaque rupture. Therefore targeting fibrin on the plaque surface can potentially identify high-risk plaques that are prone to disruption. The first fibrin-targeting agent was perfluorocarbon nanoparticles, which contained a liquid perfluorocarbon core and an encapsulating phospholipid monolayer. These nanoparticles have the advantage of carrying > 90 000 gadolinium atoms, hence allowing superior T1-weighted contrast enhancement[59]. Fibrin can also be targeted using phage display methods, whereby fibrin-targeted gadolinium-labeled peptides are used. These peptides (e.g. EP-1873) allow MR detection of fibrin deposits on the surface of ruptured plaques with good histological correlation in an animal model of coronary thrombosis[60,61]. The new generations of fibrin specific peptides (e.g. EP-2104R) are more selective for fibrin and have demonstrated superior targeting of a thrombus in vivo[62]. The main limitation of fibrin-specific peptides is the small number of gadolinium atoms (only 4 atoms) that can be attached at one time, hence requiring accumulation in sufficient quantities at the site of imaging. Nonetheless, more recent chemical exchange saturation transfer (CEST) technology is now applied to nanoparticles allowing generation of stronger MR signals. CEST contrast can originate from endogenous amide or hydroxyl protons or from exchangeable sites on exogenous CEST agents. In this technology, exchangeable protons transfer magnetization to the strong signal of bulk water after irradiation. CEST contrast agents include a liposome-based nanoparticle LIPOCEST, and other paramagnetic nanoparticles PARACEST[63-66]. In vitro fibrin clots are targeted with the use of anti-fibrin antibody formulated with perfluorocarbon nanoparticle PARACEST contrast agents[66].

Imaging cell surface markers

One of the foremost goals of molecular imaging is to detect the early stages of vulnerable plaque formation. Inflammation plays a critical role in the initiation and pathogenesis of atherosclerosis[67]. Exposure to inflammatory cytokines leads to over-expression of cell surface adhesion molecules especially E-selectin, intercellular adhesion molecule-1 and VCAM-1. These adhesion molecules mediate adhesion and migration of leukocytes along the endothelial surface to the inflammatory site. Therefore significant attention has been paid to develop probes that can detect these activated molecules on the surface of endothelial cells.

The expression of E-selectin has been targeted in vivo by generation of pegylated paramagnetic liposomes formulated with anti E-selectin antibody. E-selectin could be successfully imaged in the collared carotid arteries of apolipoprotein E-deficient apoE -/- mice as compared with controls[37]. Several generations of MNPs have been used to target VCAM-1. More recently, a MNP phage display with linear peptide based probe was used to successfully image in vivo VCAM-1 expression in the aortic roots of apoE -/- mice[52]. Furthermore, statin treatment of these mice blunted the imaging signal by reducing accumulation of the probe in the aortic root, thereby demonstrating sufficient dynamic range to detect a treatment effect. The specificity of the MNP VCAM-1 probe has also been evaluated in ex vivo human carotid endarterectomy samples. Incubation of the samples with this probe resulted in co-localization of VCAM-1-expressing cells and the MNP probe on immunohistochemistry, and resulted in a reduction T2 signal[68].

Imaging cellular targets in atherosclerotic plaque

Gadofluorine is a more lipophilic chelate of gadolinium and forms micelles due to their hydrophobic fluorinated side chain[69]. This probe has been shown to accumulate in lipid rich atherosclerotic plaques in hypercholesterolemic animal models[35,70]. High density lipoprotein (HDL) plays a key role in removing excess cholesterol from the plaques, and therefore may be a suitable candidate for transfer of nanoparticles into the plaque. HDL-like nanoparticles containing gadolinium have been developed to image the plaque in vivo. It has been shown that these HDL-like nanoparticles accumulate in atherosclerotic plaques after their intravenous injection[44]. The rate of this contrast uptake appears to be related to the lipid and macrophage content of the plaques in apoE -/- mice[27]. In addition to gadolinium chelates, CLIO-MNPs have been used to image macrophages in both animal and human atherosclerotic plaques with good histological correlation[54,55,71]. Molecular targeting of macrophages could identify the presence of inflammation in the vulnerable plaques[33,44,72]. MNP-enhanced MRI has been used in a clinical trial to assess the effect of statin dose on the level of macrophage accumulation in patients with carotid atheromatosis[73].

Imaging for angiogenesis

Growing atherosclerotic plaques initiate angiogenesis to meet their increased metabolic needs. The microvessels can cause intra-plaque hemorrhage, thereby converting these plaques to high risk and prone to rupture. New vessel formation starts from the vasa vasorum in the outer wall of the arteries and is related to the key mediator of new blood vessel formation, integrin αvβ3[36]. This integrins may represent an important molecular target for diagnosing and treating angiogenesis-related diseases. In this context, an antagonist of integrin αvβ3 has been used to induce tumor regression by targeting inhibition of neovascularization both in animals and humans[74,75]. A paramagnetic liposome containing anti-αvβ3 antibodies has been used to image this integrin in an animal model[38]. Moreover, the use of an αvβ3 integrin antagonist (fumagillin) in an animal model of atherosclerosis resulted in an anti-angiogenic effect without affecting pre-existing normal blood vessels[34,74]. Other potential investigative targets for angiogenesis include vascular endothelial growth factors and integrin α1β5.

MRI has several distinct advantages over other techniques including the absence of ionizing radiation, use of significantly less nephrotoxic contrast agent, and facilitation of high spatial resolution imaging with superb soft tissue characterization. These traits can be further enhanced by the use of specific and targeted contrast agents. Molecular imaging can help detect vulnerable plaques and may provide a risk assessment of these plaques. Furthermore, novel therapeutic nanoparticles are being developed to target these high risk plaques to provide local treatment.

CARDIAC CT

In contrast to MRI, cardiac CT incurs significant ionizing radiation exposure (10-20 mSv for retrospective gated techniques and 1-10 mSv for prospective “single phase” techniques). This modality allows for rapid data acquisition at high spatial resolution, invokes less patient anxiety given its spacious, short gantry, and has fewer contraindications than MRI. However, patient cooperation with breath-holding instructions and administration of potentially nephrotoxic contrast agents are pre-requisites for diagnostic imaging.

Electron beam CT (EBCT), which featured non-mechanical movement of the electron source and fixed detectors has been replaced in recent times by multidetector row CT (MDCT) during which both the radiation source and detectors rotate during patient motion through the CT gantry. More recently, the introduction of dual-source CT methodologies have allowed for coronary imaging without the need for β blockade. Retrospective ECG gating allows coupling of MDCT data with the corresponding phase of cardiac contraction, providing multiphasic data with superior temporal resolution when compared with EBCT. This initially incurred penalties by means of increased radiation exposure although many “low dose” protocols have since been developed and accepted into routine clinical practice.

MDCT systems allow for non-invasive characterization of different plaque components[76]. Calcium can be detected with high sensitivity and has become the established means for detection and quantification of coronary artery calcification[77]. MDCT can also be used to detect non-calcified plaque in both ex-vivo[78] and in vivo[79,80] studies. The assessment of calcification within the arterial wall may provide an independent risk factor for coronary artery disease, but it fails to identify high risk vulnerable plaques. Early studies using contrast enhanced 4-slice MDCT of coronary plaques demonstrated good correlation in differentiating between soft, intermediate and calcified plaques, as compared with IVUS[81]. In another study, 4-slice MDCT demonstrated that non-calcified plaque contributed more to total plaque burden in patients with AMI in comparison to patients with stable angina[28]. More recent studies have attempted to quantify the total volume of non-calcified atherosclerotic plaque using 64-slice MDCT. One such study evaluated 50 patients some 17 mo apart and documented a mean annualized increase of 22% in plaque volume[31]. Inter-observer variability for the quantification of non-calcified plaque volumes was found to be substantial. In a separate study the same investigators reported strong correlation between CT plaque attenuation, positive remodeling and lipid content of the plaque at contrast enhanced 64-slice CT[82]. The progression of atherosclerotic lesions resulting in focal change in luminal patency is referred to as vessel wall remodeling[83]. Positive arterial remodeling is an adaptive compensatory mechanism aiming to maintain luminal patency. In contrast, negative remodeling results in luminal narrowing irrespective of the plaque volume[84-86]. It has been shown in studies that both positive remodeling and plaque lipid content determine plaque vulnerability[84,87,88]. This hypothesis was further confirmed in another clinical study where 16/64-slice CT was performed in 38 patients with ACS and 33 patients with stable angina. The investigators reported high positive predictive value for plaque vulnerability in the presence of positive remodeling, non-calcified plaque < 30 HU, and spotty calcification[89]. Low CT attenuation, positive remodeling and spotty calcification were further shown to be associated with high risk plaque in 147 patients by 64-slice CT[90]. Other investigators have reported a 97% sensitivity for 64-slice CT when compared with IVUS in detecting plaque in 26 patients who underwent both investigations, although MDCT performed less optimally in differentiating between soft and fibrous plaque composition[32]. It has been reported that CT tends to overestimate the volume of atherosclerosis compared with IVUS[91]. In a more recent study, Pflederer et al[92] compared morphological features of plaque in patients with ACS and stable patients using contrast-enhanced coronary dual-source CT. The culprit lesions in patients with ACS were reported to have spotty calcification, low CT attenuation, large plaque volume and higher remodeling indices, as compared to control stable lesions.

There is little doubt that future generations of MDCT scanners will allow for improved coronary arterial plaque detection and characterization. Continuing improvements in spatial and temporal resolution, combined with innovative techniques such as dual-energy CT and the non-invasive nature of CT may pave the way to making this modality highly attractive for the identification of vulnerable plaque in the wider population. High definition detectors, multi-source tube-detector configurations and flat panel detectors are likely to feature prominently in the near future.

SCINTIGRAPHIC IMAGING

Scintigraphic techniques including SPECT and PET hold the potential for superior functional and molecular atherosclerotic imaging for prediction of the risk of plaque rupture. These techniques allow study of changes at a cellular and molecular level, and have been used for clinical and research purposes to study myocardial perfusion, innervation, angiogenesis, gene expression and stem cell labeling. While such metabolic information is surplus to that provided by MRI and MDCT, scintigraphic imaging techniques are limited by relatively poor spatial and temporal resolution. Both SPECT and PET involve significant ionizing radiation exposures, and as a result, these techniques also fall considerably short of the “ideal” described earlier.

Imaging cellular targets in atherosclerotic plaque

Fluorine-18 fluorodeoxyglucose (18FDG) is a glucose analog that becomes concentrated in metabolically active cells. It has been suggested that its uptake within the atherosclerotic plaque is proportional to the degree of inflammation and macrophage density[93]. Preclinical studies have suggested that PET scanning can detect 18FDG accumulation within the atherosclerotic plaque[29]. Rudd et al[30] demonstrated that human carotid plaque inflammation can be imaged with 18FDG-PET and that symptomatic plaques accumulate more 18FDG than asymptomatic lesions. In addition, histological examination of the excised symptomatic plaques in this study revealed heavy macrophage infiltration. This finding confirms that inflammation is present to a greater degree in symptomatic plaques. In a separate study, Tawakol et al[94] also showed noticeable correlation between 18FDG-PET in vivo signals and macrophage content on histological examination after carotid endarterectomy.

Imaging proteases

Matrix degrading MMPs present in the fibrous cap of the vulnerable plaque can also provide a surrogate marker of plaque instability. Activated MMPs cause proteolysis of the extracellular matrix of the fibrous cap causing plaque remodeling and rendering it susceptible to rupture. Specific radiotracers based on inhibitors of MMPs (123I-HO-I-CGS 27023A) have been generated and tested successfully in animal models[95,96]. Increased levels of macrophage or leukocyte apoptosis in the atherosclerotic plaque may contribute to plaque instability.

Imaging apoptosis

Annexin V is an endogenous protein that binds to phosphatidylserine, a negatively charged membrane phospholipid externalized to the cell surface during early cell apoptosis. 99mTc radiolabeled annexin V uptake in animal plaque has shown a good correlation with apoptosis[97]. In another similar animal model, treatment with statins resulted in a reduction in 99mTc annexin V accumulation within the plaque signifying plaque stabilization[98]. Intracellular activation of enzyme caspases is responsible for initiation of apoptosis and progress is ongoing to develop intracellular radiotracers to target these enzymes.

Imaging vasoconstricting peptides

Endothelins are 21 amino acid vasoconstricting peptides produced primarily by the endothelial cells and play a critical role in vascular homeostasis. Endothelin has three isoforms (ET-1, ET-2, ET-3), that bind to two endothelin receptors ETA and ETB, the latter being present on vascular endothelial cells. Elevated levels of ET-1 have been implicated in several vascular pathological processes including atherosclerosis, stent restenosis, endothelial dysfunction and angiogenesis. Positron emitting 18F-labeled ET-1 has shown good receptor affinity in vitro[99]. 99mTc-labeled ETs have been used in vivo with high uptake in atherosclerotic plaques in animal models[100]. In addition, specific radiolabeled antagonists of ET receptors have been demonstrated in animal studies[101].

SPECT or PET imaging could be used to identify unstable plaque and therefore may allow target treatment of high-risk plaques regardless of their angiographic appearances. The shortcomings of PET scanning are the need for ionizing radiation, substantial background uptake by the active myocardium and poor spatial resolution; however, this has been overcome with new technological advances combining CT with a PET scanner in new hybrid CT/PET devices. The combined molecular and anatomical imaging with SPECT or PET combined with CT, MRI or echocardiography may increase anatomical localization of the radiotracer signal. At present, imaging of moving coronary arteries with SPECT and PET represents a challenge.

WHY IS PLAQUE DETECTION IMPORTANT?

Detection of vulnerable plaque may help avert subsequent acute coronary syndrome by facilitating timely preventive regional and local therapies to the coronary arteries. Identification and aggressive medical treatment of these high-risk plaques can stabilize these plaques and potentially reduce the incidence of AMI and sudden cardiac death. To date there has been no prospective clinical data available upon which to develop treatment criteria for these plaques. There are several shortcomings in current vulnerable plaque detection techniques. Most invasive and non-invasive methods of vulnerable plaque detection lack sensitivity and specificity. In addition, the natural history of vulnerable plaque is unclear and until the dynamic progression of such lesions is defined, it will be difficult to implement any coherent proven treatment strategy. Furthermore, at present there are no data to prove that interventional treatment strategies with percutaneous coronary intervention for these high risk but asymptomatic plaques are superior to conventional medical treatment. In our opinion it is therefore essential to refine the available non-invasive techniques to assess the natural progression of vulnerable plaque. An established gold-standard may then be used in a longitudinal study in patients with established coronary artery disease to truly characterize this complex and dynamic process.

CONCLUSION

Significant progress has been made in the last decade to advance our understanding of the biology of atherosclerosis. This has resulted in identification of vulnerable plaques that are prone to acute thrombotic complications, which can account for the suddenness of clinical presentation in the majority of patients with coronary artery disease. A number of different novel imaging modalities have been investigated to define the specific characteristics of vulnerable plaque. However, most of these techniques are still undergoing constant refinement and cannot reliably identify vulnerable plaque in the clinical setting. It is important to realize that plaque composition is not equal to plaque vulnerability. Most of the methodologies described in this review are able to detect particular components of plaque, for example lipids and calcium. However, at present there is no definitive evidence that in vivo plaque composition is directly related to plaque vulnerability nor that the observed characteristics of a plaque are related to outcome. Further research is required to increase the sensitivity and specificity of these modalities to more accurately predict adverse events in the context of high-risk plaque. In our investigation of vulnerable plaque, it is essential that we do not forget to treat cardiovascular risk factors that initiate endothelial dysfunction, which remains the earliest pathological signal of atherosclerosis. In addition, peripheral blood can contain unique cells and cytokines that may also help in identifying the general population at risk for new sudden cardiovascular events. Many unanswered questions remain, but there is a real clinical imperative to understand the natural history of these non-obstructive plaques in order to better implement preventive strategies in the future.

Footnotes

Peer reviewers: Yuri V Bobryshev, PhD, Associate Professor, School of Medical Sciences, Faculty of Medicine University of New South Wales, Kensington NSW 2052, Australia; Masamichi Takano, MD, PhD, Cardiovascular Center, Chiba-Hokusoh Hospital, NipponMedical School, 1715 Kamakari, Imba, Chiba, 270-1694, Japan; Cristina Vassalle, PhD, G. Monasterio Foundation and Institute of Clinical Physiology, Via Moruzzi 1, I-56124, Pisa, Italy

S- Editor Cheng JX L- Editor Cant MR E- Editor Zheng XM

References
1.  Berger A, Botman KJ, MacCarthy PA, Wijns W, Bartunek J, Heyndrickx GR, Pijls NH, De Bruyne B. Long-term clinical outcome after fractional flow reserve-guided percutaneous coronary intervention in patients with multivessel disease. J Am Coll Cardiol. 2005;46:438-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 111]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
2.  Pijls NH, van Schaardenburgh P, Manoharan G, Boersma E, Bech JW, van't Veer M, Bär F, Hoorntje J, Koolen J, Wijns W. Percutaneous coronary intervention of functionally nonsignificant stenosis: 5-year follow-up of the DEFER Study. J Am Coll Cardiol. 2007;49:2105-2111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1099]  [Cited by in F6Publishing: 1114]  [Article Influence: 65.5]  [Reference Citation Analysis (0)]
3.  Tonino PA, De Bruyne B, Pijls NH, Siebert U, Ikeno F, van' t Veer M, Klauss V, Manoharan G, Engstrøm T, Oldroyd KG. Fractional flow reserve versus angiography for guiding percutaneous coronary intervention. N Engl J Med. 2009;360:213-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2974]  [Cited by in F6Publishing: 2851]  [Article Influence: 190.1]  [Reference Citation Analysis (0)]
4.  Ambrose JA, Tannenbaum MA, Alexopoulos D, Hjemdahl-Monsen CE, Leavy J, Weiss M, Borrico S, Gorlin R, Fuster V. Angiographic progression of coronary artery disease and the development of myocardial infarction. J Am Coll Cardiol. 1988;12:56-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1063]  [Cited by in F6Publishing: 1103]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
5.  Hackett D, Davies G, Maseri A. Pre-existing coronary stenoses in patients with first myocardial infarction are not necessarily severe. Eur Heart J. 1988;9:1317-1323.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Lichtlen PR, Nikutta P, Jost S, Deckers J, Wiese B, Rafflenbeul W. Anatomical progression of coronary artery disease in humans as seen by prospective, repeated, quantitated coronary angiography. Relation to clinical events and risk factors. The INTACT Study Group. Circulation. 1992;86:828-838.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Kerensky RA, Wade M, Deedwania P, Boden WE, Pepine CJ. Revisiting the culprit lesion in non-Q-wave myocardial infarction. Results from the VANQWISH trial angiographic core laboratory. J Am Coll Cardiol. 2002;39:1456-1463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 106]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
8.  Muller JE, Tofler GH, Stone PH. Circadian variation and triggers of onset of acute cardiovascular disease. Circulation. 1989;79:733-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 938]  [Cited by in F6Publishing: 889]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
9.  Frøbert O, van't Veer M, Aarnoudse W, Simonsen U, Koolen JJ, Pijls NH. Acute myocardial infarction and underlying stenosis severity. Catheter Cardiovasc Interv. 2007;70:958-965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
10.  Muller JE, Abela GS, Nesto RW, Tofler GH. Triggers, acute risk factors and vulnerable plaques: the lexicon of a new frontier. J Am Coll Cardiol. 1994;23:809-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 335]  [Cited by in F6Publishing: 343]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
11.  Fuster V, Badimon L, Badimon JJ, Chesebro JH. The pathogenesis of coronary artery disease and the acute coronary syndromes (2). N Engl J Med. 1992;326:310-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1132]  [Cited by in F6Publishing: 1175]  [Article Influence: 36.7]  [Reference Citation Analysis (0)]
12.  Fuster V, Badimon L, Badimon JJ, Chesebro JH. The pathogenesis of coronary artery disease and the acute coronary syndromes (1). N Engl J Med. 1992;326:242-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2165]  [Cited by in F6Publishing: 1988]  [Article Influence: 62.1]  [Reference Citation Analysis (0)]
13.  Stary HC. Natural history and histological classification of atherosclerotic lesions: an update. Arterioscler Thromb Vasc Biol. 2000;20:1177-1178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 492]  [Cited by in F6Publishing: 468]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
14.  Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 2000;20:1262-1275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2834]  [Cited by in F6Publishing: 2696]  [Article Influence: 112.3]  [Reference Citation Analysis (0)]
15.  Davies MJ, Richardson PD, Woolf N, Katz DR, Mann J. Risk of thrombosis in human atherosclerotic plaques: role of extracellular lipid, macrophage, and smooth muscle cell content. Br Heart J. 1993;69:377-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 837]  [Cited by in F6Publishing: 757]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
16.  Libby P. Current concepts of the pathogenesis of the acute coronary syndromes. Circulation. 2001;104:365-372.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Schaar JA, De Korte CL, Mastik F, Strijder C, Pasterkamp G, Boersma E, Serruys PW, Van Der Steen AF. Characterizing vulnerable plaque features with intravascular elastography. Circulation. 2003;108:2636-2641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 219]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
18.  Mann JM, Davies MJ. Vulnerable plaque. Relation of characteristics to degree of stenosis in human coronary arteries. Circulation. 1996;94:928-931.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Abela GS, Aziz K, Vedre A, Pathak DR, Talbott JD, Dejong J. Effect of cholesterol crystals on plaques and intima in arteries of patients with acute coronary and cerebrovascular syndromes. Am J Cardiol. 2009;103:959-968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 145]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
20.  Farb A, Burke AP, Tang AL, Liang TY, Mannan P, Smialek J, Virmani R. Coronary plaque erosion without rupture into a lipid core. A frequent cause of coronary thrombosis in sudden coronary death. Circulation. 1996;93:1354-1363.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Sharif F, Murphy RT. Current status of vulnerable plaque detection. Catheter Cardiovasc Interv. 2010;75:135-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
22.  Fujimoto JG, Boppart SA, Tearney GJ, Bouma BE, Pitris C, Brezinski ME. High resolution in vivo intra-arterial imaging with optical coherence tomography. Heart. 1999;82:128-133.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Moore MP, Spencer T, Salter DM, Kearney PP, Shaw TR, Starkey IR, Fitzgerald PJ, Erbel R, Lange A, McDicken NW. Characterisation of coronary atherosclerotic morphology by spectral analysis of radiofrequency signal: in vitro intravascular ultrasound study with histological and radiological validation. Heart. 1998;79:459-467.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Rodriguez-Granillo GA, García-García HM, Mc Fadden EP, Valgimigli M, Aoki J, de Feyter P, Serruys PW. In vivo intravascular ultrasound-derived thin-cap fibroatheroma detection using ultrasound radiofrequency data analysis. J Am Coll Cardiol. 2005;46:2038-2042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 246]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
25.  von Birgelen C, Klinkhart W, Mintz GS, Papatheodorou A, Herrmann J, Baumgart D, Haude M, Wieneke H, Ge J, Erbel R. Plaque distribution and vascular remodeling of ruptured and nonruptured coronary plaques in the same vessel: an intravascular ultrasound study in vivo. J Am Coll Cardiol. 2001;37:1864-1870.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Yabushita H, Bouma BE, Houser SL, Aretz HT, Jang IK, Schlendorf KH, Kauffman CR, Shishkov M, Kang DH, Halpern EF. Characterization of human atherosclerosis by optical coherence tomography. Circulation. 2002;106:1640-1645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 898]  [Cited by in F6Publishing: 812]  [Article Influence: 36.9]  [Reference Citation Analysis (0)]
27.  Frias JC, Ma Y, Williams KJ, Fayad ZA, Fisher EA. Properties of a versatile nanoparticle platform contrast agent to image and characterize atherosclerotic plaques by magnetic resonance imaging. Nano Lett. 2006;6:2220-2224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 94]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
28.  Leber AW, Knez A, White CW, Becker A, von Ziegler F, Muehling O, Becker C, Reiser M, Steinbeck G, Boekstegers P. Composition of coronary atherosclerotic plaques in patients with acute myocardial infarction and stable angina pectoris determined by contrast-enhanced multislice computed tomography. Am J Cardiol. 2003;91:714-718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 193]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
29.  Lederman RJ, Raylman RR, Fisher SJ, Kison PV, San H, Nabel EG, Wahl RL. Detection of atherosclerosis using a novel positron-sensitive probe and 18-fluorodeoxyglucose (FDG). Nucl Med Commun. 2001;22:747-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 114]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
30.  Rudd JH, Warburton EA, Fryer TD, Jones HA, Clark JC, Antoun N, Johnström P, Davenport AP, Kirkpatrick PJ, Arch BN. Imaging atherosclerotic plaque inflammation with [18F]-fluorodeoxyglucose positron emission tomography. Circulation. 2002;105:2708-2711.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 976]  [Cited by in F6Publishing: 893]  [Article Influence: 40.6]  [Reference Citation Analysis (0)]
31.  Schmid M, Achenbach S, Ropers D, Komatsu S, Ropers U, Daniel WG, Pflederer T. Assessment of changes in non-calcified atherosclerotic plaque volume in the left main and left anterior descending coronary arteries over time by 64-slice computed tomography. Am J Cardiol. 2008;101:579-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
32.  Sun J, Zhang Z, Lu B, Yu W, Yang Y, Zhou Y, Wang Y, Fan Z. Identification and quantification of coronary atherosclerotic plaques: a comparison of 64-MDCT and intravascular ultrasound. AJR Am J Roentgenol. 2008;190:748-754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 159]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
33.  Winter PM, Morawski AM, Caruthers SD, Fuhrhop RW, Zhang H, Williams TA, Allen JS, Lacy EK, Robertson JD, Lanza GM. Molecular imaging of angiogenesis in early-stage atherosclerosis with alpha(v)beta3-integrin-targeted nanoparticles. Circulation. 2003;108:2270-2274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 569]  [Cited by in F6Publishing: 482]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
34.  Winter PM, Neubauer AM, Caruthers SD, Harris TD, Robertson JD, Williams TA, Schmieder AH, Hu G, Allen JS, Lacy EK. Endothelial alpha(v)beta3 integrin-targeted fumagillin nanoparticles inhibit angiogenesis in atherosclerosis. Arterioscler Thromb Vasc Biol. 2006;26:2103-2109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 331]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
35.  Barkhausen J, Ebert W, Heyer C, Debatin JF, Weinmann HJ. Detection of atherosclerotic plaque with Gadofluorine-enhanced magnetic resonance imaging. Circulation. 2003;108:605-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 143]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
36.  Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science. 1994;264:569-571.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Mulder WJ, Douma K, Koning GA, van Zandvoort MA, Lutgens E, Daemen MJ, Nicolay K, Strijkers GJ. Liposome-enhanced MRI of neointimal lesions in the ApoE-KO mouse. Magn Reson Med. 2006;55:1170-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
38.  Sipkins DA, Cheresh DA, Kazemi MR, Nevin LM, Bednarski MD, Li KC. Detection of tumor angiogenesis in vivo by alphaVbeta3-targeted magnetic resonance imaging. Nat Med. 1998;4:623-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 695]  [Cited by in F6Publishing: 725]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
39.  Yuan C, Hatsukami TS, Obrien KD. High-Resolution magnetic resonance imaging of normal and atherosclerotic human coronary arteries ex vivo: discrimination of plaque tissue components. J Investig Med. 2001;49:491-499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
40.  Botnar RM, Stuber M, Kissinger KV, Manning WJ. Free-breathing 3D coronary MRA: the impact of "isotropic" image resolution. J Magn Reson Imaging. 2000;11:389-393.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Fayad ZA, Fuster V, Fallon JT, Jayasundera T, Worthley SG, Helft G, Aguinaldo JG, Badimon JJ, Sharma SK. Noninvasive in vivo human coronary artery lumen and wall imaging using black-blood magnetic resonance imaging. Circulation. 2000;102:506-510.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Kim WY, Stuber M, Börnert P, Kissinger KV, Manning WJ, Botnar RM. Three-dimensional black-blood cardiac magnetic resonance coronary vessel wall imaging detects positive arterial remodeling in patients with nonsignificant coronary artery disease. Circulation. 2002;106:296-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 287]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
43.  Worthley SG, Helft G, Fuster V, Fayad ZA, Rodriguez OJ, Zaman AG, Fallon JT, Badimon JJ. Noninvasive in vivo magnetic resonance imaging of experimental coronary artery lesions in a porcine model. Circulation. 2000;101:2956-2961.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Frias JC, Williams KJ, Fisher EA, Fayad ZA. Recombinant HDL-like nanoparticles: a specific contrast agent for MRI of atherosclerotic plaques. J Am Chem Soc. 2004;126:16316-16317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 225]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
45.  Lipinski MJ, Amirbekian V, Frias JC, Aguinaldo JG, Mani V, Briley-Saebo KC, Fuster V, Fallon JT, Fisher EA, Fayad ZA. MRI to detect atherosclerosis with gadolinium-containing immunomicelles targeting the macrophage scavenger receptor. Magn Reson Med. 2006;56:601-610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 136]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
46.  Morawski AM, Winter PM, Crowder KC, Caruthers SD, Fuhrhop RW, Scott MJ, Robertson JD, Abendschein DR, Lanza GM, Wickline SA. Targeted nanoparticles for quantitative imaging of sparse molecular epitopes with MRI. Magn Reson Med. 2004;51:480-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 186]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
47.  Yu X, Song SK, Chen J, Scott MJ, Fuhrhop RJ, Hall CS, Gaffney PJ, Wickline SA, Lanza GM. High-resolution MRI characterization of human thrombus using a novel fibrin-targeted paramagnetic nanoparticle contrast agent. Magn Reson Med. 2000;44:867-872.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Kircher MF, Mahmood U, King RS, Weissleder R, Josephson L. A multimodal nanoparticle for preoperative magnetic resonance imaging and intraoperative optical brain tumor delineation. Cancer Res. 2003;63:8122-8125.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Schellenberger EA, Sosnovik D, Weissleder R, Josephson L. Magneto/optical annexin V, a multimodal protein. Bioconjug Chem. 2004;15:1062-1067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 172]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
50.  Sosnovik DE, Schellenberger EA, Nahrendorf M, Novikov MS, Matsui T, Dai G, Reynolds F, Grazette L, Rosenzweig A, Weissleder R. Magnetic resonance imaging of cardiomyocyte apoptosis with a novel magneto-optical nanoparticle. Magn Reson Med. 2005;54:718-724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 200]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
51.  Kelly KA, Nahrendorf M, Yu AM, Reynolds F, Weissleder R. In vivo phage display selection yields atherosclerotic plaque targeted peptides for imaging. Mol Imaging Biol. 2006;8:201-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 95]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
52.  Nahrendorf M, Jaffer FA, Kelly KA, Sosnovik DE, Aikawa E, Libby P, Weissleder R. Noninvasive vascular cell adhesion molecule-1 imaging identifies inflammatory activation of cells in atherosclerosis. Circulation. 2006;114:1504-1511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 472]  [Cited by in F6Publishing: 430]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
53.  Denis MC, Mahmood U, Benoist C, Mathis D, Weissleder R. Imaging inflammation of the pancreatic islets in type 1 diabetes. Proc Natl Acad Sci U S A. 2004;101:12634-12639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 151]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
54.  Jaffer FA, Nahrendorf M, Sosnovik D, Kelly KA, Aikawa E, Weissleder R. Cellular imaging of inflammation in atherosclerosis using magnetofluorescent nanomaterials. Mol Imaging. 2004;5:85-92.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Kooi ME, Cappendijk VC, Cleutjens KB, Kessels AG, Kitslaar PJ, Borgers M, Frederik PM, Daemen MJ, van Engelshoven JM. Accumulation of ultrasmall superparamagnetic particles of iron oxide in human atherosclerotic plaques can be detected by in vivo magnetic resonance imaging. Circulation. 2003;107:2453-2458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 634]  [Cited by in F6Publishing: 659]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
56.  Krombach GA, Wendland MF, Higgins CB, Saeed M. MR imaging of spatial extent of microvascular injury in reperfused ischemically injured rat myocardium: value of blood pool ultrasmall superparamagnetic particles of iron oxide. Radiology. 2002;225:479-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
57.  Harris TJ, von Maltzahn G, Derfus AM, Ruoslahti E, Bhatia SN. Proteolytic actuation of nanoparticle self-assembly. Angew Chem Int Ed Engl. 2006;45:3161-3165.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Perez JM, Simeone FJ, Saeki Y, Josephson L, Weissleder R. Viral-induced self-assembly of magnetic nanoparticles allows the detection of viral particles in biological media. J Am Chem Soc. 2003;125:10192-10193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 448]  [Cited by in F6Publishing: 450]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
59.  Flacke S, Fischer S, Scott MJ, Fuhrhop RJ, Allen JS, McLean M, Winter P, Sicard GA, Gaffney PJ, Wickline SA. Novel MRI contrast agent for molecular imaging of fibrin: implications for detecting vulnerable plaques. Circulation. 2001;104:1280-1285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 405]  [Cited by in F6Publishing: 428]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
60.  Botnar RM, Buecker A, Wiethoff AJ, Parsons EC, Katoh M, Katsimaglis G, Weisskoff RM, Lauffer RB, Graham PB, Gunther RW. In vivo magnetic resonance imaging of coronary thrombosis using a fibrin-binding molecular magnetic resonance contrast agent. Circulation. 2004;110:1463-1466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 168]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
61.  Botnar RM, Perez AS, Witte S, Wiethoff AJ, Laredo J, Hamilton J, Quist W, Parsons EC, Vaidya A, Kolodziej A. In vivo molecular imaging of acute and subacute thrombosis using a fibrin-binding magnetic resonance imaging contrast agent. Circulation. 2004;109:2023-2029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 227]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
62.  Spuentrup E, Fausten B, Kinzel S, Wiethoff AJ, Botnar RM, Graham PB, Haller S, Katoh M, Parsons EC, Manning WJ. Molecular magnetic resonance imaging of atrial clots in a swine model. Circulation. 2005;112:396-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 90]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
63.  Aime S, Delli Castelli D, Terreno E. Highly sensitive MRI chemical exchange saturation transfer agents using liposomes. Angew Chem Int Ed Engl. 2005;44:5513-5515.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Aime S, Barge A, Delli Castelli D, Fedeli F, Mortillaro A, Nielsen FU, Terreno E. Paramagnetic lanthanide(III) complexes as pH-sensitive chemical exchange saturation transfer (CEST) contrast agents for MRI applications. Magn Reson Med. 2002;47:639-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 285]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
65.  Terreno E, Cabella C, Carrera C, Delli Castelli D, Mazzon R, Rollet S, Stancanello J, Visigalli M, Aime S. From spherical to osmotically shrunken paramagnetic liposomes: an improved generation of LIPOCEST MRI agents with highly shifted water protons. Angew Chem Int Ed Engl. 2007;46:966-968.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Winter PM, Cai K, Chen J, Adair CR, Kiefer GE, Athey PS, Gaffney PJ, Buff CE, Robertson JD, Caruthers SD. Targeted PARACEST nanoparticle contrast agent for the detection of fibrin. Magn Reson Med. 2006;56:1384-1388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 85]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
67.  Ross R. Atherosclerosis--an inflammatory disease. N Engl J Med. 1999;340:115-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15621]  [Cited by in F6Publishing: 15151]  [Article Influence: 606.0]  [Reference Citation Analysis (0)]
68.  Kelly KA, Allport JR, Tsourkas A, Shinde-Patil VR, Josephson L, Weissleder R. Detection of vascular adhesion molecule-1 expression using a novel multimodal nanoparticle. Circ Res. 2005;96:327-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in F6Publishing: 300]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
69.  Staatz G, Nolte-Ernsting CC, Adam GB, Grosskortenhaus S, Misselwitz B, Bücker A, Günther RW. Interstitial T1-weighted MR lymphography: lipophilic perfluorinated gadolinium chelates in pigs. Radiology. 2001;220:129-134.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Sirol M, Itskovich VV, Mani V, Aguinaldo JG, Fallon JT, Misselwitz B, Weinmann HJ, Fuster V, Toussaint JF, Fayad ZA. Lipid-rich atherosclerotic plaques detected by gadofluorine-enhanced in vivo magnetic resonance imaging. Circulation. 2004;109:2890-2896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 173]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
71.  Trivedi RA, U-King-Im JM, Graves MJ, Cross JJ, Horsley J, Goddard MJ, Skepper JN, Quartey G, Warburton E, Joubert I. In vivo detection of macrophages in human carotid atheroma: temporal dependence of ultrasmall superparamagnetic particles of iron oxide-enhanced MRI. Stroke. 2004;35:1631-1635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 261]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
72.  Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ, Pittenger MF, Hare JM, Bulte JW. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation. 2003;107:2290-2293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 569]  [Cited by in F6Publishing: 600]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
73.  Heyn C, Bowen CV, Rutt BK, Foster PJ. Detection threshold of single SPIO-labeled cells with FIESTA. Magn Reson Med. 2005;53:312-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 194]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
74.  Brooks PC, Montgomery AM, Rosenfeld M, Reisfeld RA, Hu T, Klier G, Cheresh DA. Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell. 1994;79:1157-1164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1738]  [Cited by in F6Publishing: 1667]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
75.  Brooks PC, Strömblad S, Klemke R, Visscher D, Sarkar FH, Cheresh DA. Antiintegrin alpha v beta 3 blocks human breast cancer growth and angiogenesis in human skin. J Clin Invest. 1995;96:1815-1822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 608]  [Cited by in F6Publishing: 648]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
76.  Becker CR. Assessment of coronary arteries with CT. Radiol Clin North Am. 2002;40:773-782, vi.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
77.  Becker CR, Jakobs TF, Aydemir S, Becker A, Knez A, Schoepf UJ, Bruening R, Haberl R, Reiser MF. Helical and single-slice conventional CT versus electron beam CT for the quantification of coronary artery calcification. AJR Am J Roentgenol. 2000;174:543-547.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Becker CR, Nikolaou K, Muders M, Babaryka G, Crispin A, Schoepf UJ, Loehrs U, Reiser MF. Ex vivo coronary atherosclerotic plaque characterization with multi-detector-row CT. Eur Radiol. 2003;13:2094-2098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 205]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
79.  Becker CR, Knez A, Leber A, Treede H, Haberl R, Reiser MF. [Angiography with multi-slice spiral CT. Detecting plaque, before it causes symptoms]. MMW Fortschr Med. 2001;143:30-32.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Becker CR, Knez A, Ohnesorge B, Schoepf UJ, Reiser MF. Imaging of noncalcified coronary plaques using helical CT with retrospective ECG gating. AJR Am J Roentgenol. 2000;175:423-424.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Schroeder S, Kopp AF, Baumbach A, Meisner C, Kuettner A, Georg C, Ohnesorge B, Herdeg C, Claussen CD, Karsch KR. Noninvasive detection and evaluation of atherosclerotic coronary plaques with multislice computed tomography. J Am Coll Cardiol. 2001;37:1430-1435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 554]  [Cited by in F6Publishing: 583]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
82.  Schmid M, Pflederer T, Jang IK, Ropers D, Sei K, Daniel WG, Achenbach S. Relationship between degree of remodeling and CT attenuation of plaque in coronary atherosclerotic lesions: an in-vivo analysis by multi-detector computed tomography. Atherosclerosis. 2008;197:457-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
83.  Losordo DW, Rosenfield K, Kaufman J, Pieczek A, Isner JM. Focal compensatory enlargement of human arteries in response to progressive atherosclerosis. In vivo documentation using intravascular ultrasound. Circulation. 1994;89:2570-2577.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Pasterkamp G, Schoneveld AH, van der Wal AC, Haudenschild CC, Clarijs RJ, Becker AE, Hillen B, Borst C. Relation of arterial geometry to luminal narrowing and histologic markers for plaque vulnerability: the remodeling paradox. J Am Coll Cardiol. 1998;32:655-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 261]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
85.  Schoenhagen P, Ziada KM, Kapadia SR, Crowe TD, Nissen SE, Tuzcu EM. Extent and direction of arterial remodeling in stable versus unstable coronary syndromes : an intravascular ultrasound study. Circulation. 2000;101:598-603.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Varnava AM, Mills PG, Davies MJ. Relationship between coronary artery remodeling and plaque vulnerability. Circulation. 2002;105:939-943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 383]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
87.  Smits PC, Pasterkamp G, Quarles van Ufford MA, Eefting FD, Stella PR, de Jaegere PP, Borst C. Coronary artery disease: arterial remodelling and clinical presentation. Heart. 1999;82:461-464.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Stiel GM, Stiel LS, Schofer J, Donath K, Mathey DG. Impact of compensatory enlargement of atherosclerotic coronary arteries on angiographic assessment of coronary artery disease. Circulation. 1989;80:1603-1609.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Motoyama S, Kondo T, Sarai M, Sugiura A, Harigaya H, Sato T, Inoue K, Okumura M, Ishii J, Anno H. Multislice computed tomographic characteristics of coronary lesions in acute coronary syndromes. J Am Coll Cardiol. 2007;50:319-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 772]  [Cited by in F6Publishing: 747]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
90.  Kitagawa T, Yamamoto H, Horiguchi J, Ohhashi N, Tadehara F, Shokawa T, Dohi Y, Kunita E, Utsunomiya H, Kohno N. Characterization of noncalcified coronary plaques and identification of culprit lesions in patients with acute coronary syndrome by 64-slice computed tomography. JACC Cardiovasc Imaging. 2009;2:153-160.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Otsuka M, Bruining N, Van Pelt NC, Mollet NR, Ligthart JM, Vourvouri E, Hamers R, De Jaegere P, Wijns W, Van Domburg RT. Quantification of coronary plaque by 64-slice computed tomography: a comparison with quantitative intracoronary ultrasound. Invest Radiol. 2008;43:314-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 77]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
92.  Pflederer T, Marwan M, Schepis T, Ropers D, Seltmann M, Muschiol G, Daniel WG, Achenbach S. Characterization of culprit lesions in acute coronary syndromes using coronary dual-source CT angiography. Atherosclerosis. 2010;211:437-444.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Tawakol A, Migrino RQ, Hoffmann U, Abbara S, Houser S, Gewirtz H, Muller JE, Brady TJ, Fischman AJ. Noninvasive in vivo measurement of vascular inflammation with F-18 fluorodeoxyglucose positron emission tomography. J Nucl Cardiol. 2005;12:294-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 231]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
94.  Tawakol A, Migrino RQ, Bashian GG, Bedri S, Vermylen D, Cury RC, Yates D, LaMuraglia GM, Furie K, Houser S. In vivo 18F-fluorodeoxyglucose positron emission tomography imaging provides a noninvasive measure of carotid plaque inflammation in patients. J Am Coll Cardiol. 2006;48:1818-1824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 710]  [Cited by in F6Publishing: 682]  [Article Influence: 37.9]  [Reference Citation Analysis (0)]
95.  Kopka K, Breyholz HJ, Wagner S, Law MP, Riemann B, Schröer S, Trub M, Guilbert B, Levkau B, Schober O. Synthesis and preliminary biological evaluation of new radioiodinated MMP inhibitors for imaging MMP activity in vivo. Nucl Med Biol. 2004;31:257-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 58]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
96.  Schäfers M, Riemann B, Kopka K, Breyholz HJ, Wagner S, Schäfers KP, Law MP, Schober O, Levkau B. Scintigraphic imaging of matrix metalloproteinase activity in the arterial wall in vivo. Circulation. 2004;109:2554-2559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 178]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
97.  Kolodgie FD, Petrov A, Virmani R, Narula N, Verjans JW, Weber DK, Hartung D, Steinmetz N, Vanderheyden JL, Vannan MA. Targeting of apoptotic macrophages and experimental atheroma with radiolabeled annexin V: a technique with potential for noninvasive imaging of vulnerable plaque. Circulation. 2003;108:3134-3139.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Hartung D, Sarai M, Petrov A, Kolodgie F, Narula N, Verjans J, Virmani R, Reutelingsperger C, Hofstra L, Narula J. Resolution of apoptosis in atherosclerotic plaque by dietary modification and statin therapy. J Nucl Med. 2005;46:2051-2056.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Johnström P, Harris NG, Fryer TD, Barret O, Clark JC, Pickard JD, Davenport AP. (18)F-Endothelin-1, a positron emission tomography (PET) radioligand for the endothelin receptor system: radiosynthesis and in vivo imaging using microPET. Clin Sci (Lond). 2002;103 Suppl 48:4S-8S.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Dinkelborg LM, Duda SH, Hanke H, Tepe G, Hilger CS, Semmler W. Molecular imaging of atherosclerosis using a technetium-99m-labeled endothelin derivative. J Nucl Med. 1998;39:1819-1822.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Johnström P, Rudd JH, Richards HK, Fryer TD, Clark JC, Weissberg PL, Pickard JD, Davenport AP. Imaging endothelin ET(B) receptors using [18F]-BQ3020: in vitro characterization and positron emission tomography (microPET). Exp Biol Med (Maywood). 2006;231:736-740.  [PubMed]  [DOI]  [Cited in This Article: ]