480
Views
14
CrossRef citations to date
0
Altmetric
REVIEW ARTICLE

Characterization and molecular detection of atherothrombosis by magnetic resonance—potential tools for individual risk assessment and diagnostics

, &
Pages 322-336 | Published online: 08 Jul 2009

Abstract

This review focuses on recent non‐invasive or minimally invasive magnetic resonance (MR) approaches to study atherothrombosis. The potential benefits of combining diverse metabolic information obtained by the variety of MR techniques from tissues in vivo and ex vivo and from body fluids in vitro are also briefly discussed. A well established methodology is available for lipoprotein subclass quantification from plasma by 1H MR spectroscopy providing information for assessing the long‐term risk of atherosclerosis. Multi‐contrast MR imaging in vivo relying on endogenous contrast allows partial characterization of components in atherothrombotic plaques. The use of exogenous contrast agents in MR angiography enhances blood‐tissue contrast and provides functional information on plaque metabolism, improving plaque characterization and assessment of plaque vulnerability by MR imaging. Recent applications of molecular targeted MR imaging have revealed novel opportunities for specific early detection of atherothrombotic processes, such as angiogenesis and accumulation of macrophages. Currently, MR imaging and spectroscopy can produce such metabolic in vivo and in vitro information that in combination could facilitate the screening, identification and follow‐up of cardiovascularly vulnerable patients in research settings. The recent developments imply that in the near future MR techniques will be part of clinical protocols for individual diagnostics in atherothrombosis.

View correction statement:
ERRATUM

Abbreviations
ACS=

acute coronary syndrome

CAD=

coronary artery disease

CHD=

coronary heart disease

HDL=

high‐density lipoprotein

LDL=

low‐density lipoprotein

MR=

magnetic resonance

MRA=

magnetic resonance angiography

MRI=

magnetic resonance imaging

MRS=

magnetic resonance spectroscopy

rHDL=

reconstituted high‐density lipoprotein

VLDL=

very low‐density lipoprotein

Atherothrombosis—life is full of risks

Atherosclerosis is a diffuse systemic disease that is characterized by the local build‐up of lipid‐rich plaques within the walls of large arteries. The processes are multigenetic, being influenced also by dietary and environmental factors, and are apparent as early as the second decade in life with an increased incidence in the elderly Citation1. Inflammatory processes are in a key role in the development of atherothrombosis Citation2. In advanced disease plaque rupture as well as erosion can lead to arterial thrombosis and thereby to an acute coronary syndrome (ACS) Citation3.

A common practice for assessing the risk of an individual for atherosclerosis is to measure the lipoprotein lipids in blood plasma or serum: high levels of low‐density lipoprotein (LDL) and low levels of high‐density lipoprotein (HDL) cholesterol are pro‐atherogenic Citation4. The lipoprotein subclasses are also characteristically related to the risk of atherosclerosis Citation5. Thus, in principle, with the lipoprotein profile of a person one can approximate the risk value for a clinical manifestation of atherosclerosis, i.e., the probability of a cardiovascular event, in a specified period of time. However, the predictive accuracy of the lipoprotein and other blood compartment‐associated risk factors is too poor to be clinically useful in predicting the individual incidence of ACS in the near future Citation6,7. Therefore, the biochemistry of plasma seems not to be able to consistently reflect the atherothrombotic processes in the vessel wall. This is not surprising since atherothrombotic plaques take many forms depending on the relative proportion of, e.g., a lipid‐rich core, a fibrous cap, vascular smooth‐muscle cells, extracellular matrix components, and inflammatory cells Citation3.

Currently the standard method for the evaluation of coronary artery disease (CAD) is invasive angiographic visualization and quantification of significant coronary stenosis Citation8. Angiography (with either X‐ray or magnetic resonance (MR)) images only the vessel lumen and the silhouette of lesions that impinge on the lumen Citation9. However, it is possible that lumen calibre preserves unaltered or minimally altered despite the presence of a large plaque Citation10. In view of that, for example, the coronary artery which subsequently occluded had less than a 50% stenosis on the first angiogram in two‐thirds of patients Citation11. In other words, ACSs often result from disruption of mildly stenotic plaques with large extracellular lipid cores, thin fibrous caps and inflammatory cell infiltrates Citation3. In the coronary arteries, the pathobiological behaviour of plaque is consequently determined by its molecular composition and not by its size. In addition, there can be striking heterogeneity in the composition of human atherothrombotic plaques even in the same individual Citation3. Accordingly, angiography alone is not adequate for diagnosing a CAD or the risk for an ACS.

Hence, there is a fundamental requirement for non‐invasive in vivo imaging tools, firstly, to allow for direct detection of atherothrombotic tissues, i.e., to identify and probe atherothrombosis Citation12, and, secondly, to characterize the composition of various plaques in order to understand and assess their risk for rupture and thrombotic complications, i.e., to predict an ACS on an individual basis Citation13. As a summary, presents a potential scheme for the utilization of MR methodologies in the assessment of long‐term and short‐term risk for atherothrombotic events. This scheme can be seen as one option to elucidate the potential of MR in detecting individual intermediate atherothrombotic end‐points and utilizing their prognostic value before the occurrence of a definite end‐point as discussed in Raggi et al. Citation14.

Figure 1. A potential scheme utilizing magnetic resonance(MR) methodologies in the risk assessment of long‐term risk for atherothrombotic events (non‐symptomatic individuals) and of short‐term risk for recurrent cardiovascular events after an experienced acute coronary syndrome (ACS) (symptomatic patients). At risk assessment point I the molecular constituents of serum, including lipoprotein subclasses, could be assessed by in vitro1H magnetic resonance spectroscopy (MRS) metabonomics for non‐symptomatic individuals. If high long‐term risk for atherothrombotic events is indicated, non‐invasive in vivo magnetic resonance imaging (MRI) could follow for the potential detection of plaque (risk assessment point IIa) and subsequent compositional evaluation of the vulnerability of the detected plaque(s) for rupture or erosion (IIb). Depending on the outcome from the plaque detection and assessment by MRI the individual could accordingly be directed for further actions. If vulnerable plaque at point IIb would be detected, considerations for aggressive drug therapy or invasive therapies such as angiographic stenting or bypass surgery would be needed. In the case of an individual with an experienced ACS (III) in vitro1H MRS metabonomics could be used to complement the clinical protocols when evaluating the risk for recurrent cardiovascular events and the proper individual treatment options. For some symptomatic patients in vivo MRI might also be feasible at point III for direct assessment of plaque composition and vulnerability. This scheme can be seen as one option to elucidate the potential of MR in detecting individual intermediate atherothrombotic end‐points and utilizing their prognostic value before the occurrence of a definite end‐point. The recent MR findings and developments summarized in this review awaken confidence that this kind of scheme might be operational in the near future saving both human suffering and societal health costs.

Figure 1. A potential scheme utilizing magnetic resonance(MR) methodologies in the risk assessment of long‐term risk for atherothrombotic events (non‐symptomatic individuals) and of short‐term risk for recurrent cardiovascular events after an experienced acute coronary syndrome (ACS) (symptomatic patients). At risk assessment point I the molecular constituents of serum, including lipoprotein subclasses, could be assessed by in vitro1H magnetic resonance spectroscopy (MRS) metabonomics for non‐symptomatic individuals. If high long‐term risk for atherothrombotic events is indicated, non‐invasive in vivo magnetic resonance imaging (MRI) could follow for the potential detection of plaque (risk assessment point IIa) and subsequent compositional evaluation of the vulnerability of the detected plaque(s) for rupture or erosion (IIb). Depending on the outcome from the plaque detection and assessment by MRI the individual could accordingly be directed for further actions. If vulnerable plaque at point IIb would be detected, considerations for aggressive drug therapy or invasive therapies such as angiographic stenting or bypass surgery would be needed. In the case of an individual with an experienced ACS (III) in vitro1H MRS metabonomics could be used to complement the clinical protocols when evaluating the risk for recurrent cardiovascular events and the proper individual treatment options. For some symptomatic patients in vivo MRI might also be feasible at point III for direct assessment of plaque composition and vulnerability. This scheme can be seen as one option to elucidate the potential of MR in detecting individual intermediate atherothrombotic end‐points and utilizing their prognostic value before the occurrence of a definite end‐point. The recent MR findings and developments summarized in this review awaken confidence that this kind of scheme might be operational in the near future saving both human suffering and societal health costs.

Key messages

  • Magnetic resonance imaging (MRI) is a non‐invasive method allowing detection, characterization and follow‐up of atherothrombosis in vivo.

  • The use of in vitro1H magnetic resonance spectroscopy (MRS), to detect lipoprotein subclasses and plasma metabolites, together with in vivo multi‐contrast MRI, to characterize plaque components, could provide a genuinely new approach to uncover individual intermediate atherothrombosis before any clinical manifestations.

  • In general, contrast agents enhance characterization of atherothrombotic processes, and, in particular, molecular targeted MRI offers a kind of in vivo equivalent to immunohistochemistry for detecting early atherosclerotic processes, such as accumulation of macrophages and angiogenesis.

A vulnerable patient—magnetic resonance imaging and spectroscopy

Vulnerable plaques are not the only culprit factors for the ACSs but vulnerable blood (prone to thrombosis) and vulnerable myocardium (prone to fatal arrhythmia) also play an important role in the outcome. Hence, a concept of a ‘cardiovascular vulnerable patient’ was recently proposed to define subjects susceptible to an ACS or sudden cardiac death based on plaque, blood, or myocardial vulnerability (for example, 1‐year risk ⩾5%) Citation6,7. For the detection of a vulnerable plaque, five major criteria were presented, namely active inflammation, a thin cap with a large lipid core, endothelial denudation with superficial platelet aggregation, fissured or injured plaque and severe stenosis. Interestingly, magnetic resonance imaging (MRI) seems to be the only non‐invasive methodology capable of detecting all these major criteria for plaque vulnerability Citation6. Several serum markers of vulnerable blood (reflecting metabolic and immune disorders), e.g., abnormal lipoprotein profiles, diabetes, hypertriglyceridaemia, and lipid peroxidation are also such that they influence characteristically the MR spectroscopy (MRS) information of plasma and serum Citation15.

In this review we will discuss the potential use of MRI and MRS in the characterization and risk assessment of atherothrombosis. The applications of endogenous contrast in MRI will be dealt with before discussing the use of exogenous contrast agents. Molecular targeted MRI is then discussed in relation to the molecular biology of atherothrombosis followed by a brief note on the role of MRS in lipoprotein quantification and plaque characterization.

Magnetic resonance imaging and angiography

It should be noted that because the MR method operates at the radio frequency region of the electromagnetic radiation it is intrinsically non‐ionizing, non‐destructive and non‐invasive. In routine clinical settings MRI offers three‐dimensional data with high spatial resolution (approximately 1 mm) and in advanced research settings for two decades less (∼10 µm). The MRI techniques for the evaluation of vascular anatomy are collectively called magnetic resonance angiography (MRA). Currently, MRI and MRA are often seen as a favourable technology for the detection, visualization and characterization of atherothrombosis: also two extensive text‐books of cardiovascular magnetic resonance (CMR) have recently appeared Citation16,17.

Anatomical aspects

X‐ray tomography, with the aid of ionizing radiation and nephrotoxic contrast media, is at present the most accurate and robust way to image coronary arteries Citation18. However, a new non‐invasive MRA technique, that does not even use a contrast agent, now allows imaging of the whole coronary artery tree within 30 minutes with 0.55×0.55×0.75 mm3 voxel size Citation19.

Characterization and quantification of plaque using endogenous contrast

By using multi‐contrast MRI, i.e., a combination of T1‐, T2‐, and proton density‐weighted images, with sub‐mm spatial resolution, plaque anatomy and components have been determined in various test animal and human artery specimens ex vivo and in vivoCitation20. Unfortunately, in general there is an overlap of signal intensities between the different MRI pulse sequences (e.g., T1‐, T2‐, and proton density weighting) and the various plaque components (e.g., fibrous cap, lipid core, calcification, thrombus, media and adventitia), which makes unique identification of the plaque components difficult and requires particular attention on the image analysis. Recently, however, a spatially enhanced iterative cluster analysis has been developed for the automated analysis of multi‐contrast ex vivo MR images of human coronary artery specimens. It turned out that the resulting MR segmentation had an excellent match with histopathology images for all American Heart Association plaque classification types I–VI Citation21. A result using the new cluster analysis method is illustrated in together with an applied colour composite MR image as well as a histopathology for comparison. In addition, a stepwise logistic regression model has been developed to identify human atherosclerotic carotid plaque components based on multi‐sequence in vivo MRI Citation22. As a complement to the above plaque classifications, a detailed description of the MRI appearance of a plaque rupture‐associated thrombosis in histologically validated platelet‐rich thrombi has recently been presented Citation23. These studies jointly suggest that MRI seems to be intrinsically capable of imaging the whole cascade of atherothrombotic phenomena non‐invasively without contrast agents.

Figure 2. Illustration of the spatially enhanced iterative cluster analysis for the multi‐contrast ex vivo magnetic resonance (MR) images of human coronary artery specimens. On the left are shown the T1‐, proton density, and T2‐weighted (T1W, PDW and T2W, respectively) axial plane images that were assigned red (R), green (G), and blue (B) colours, respectively, and combined to yield a colour composite and pseudo‐colour cluster image of the coronary artery with colour distributions corresponding to different tissue types. On the right a comparable histopathology image via a combined Masson's elastic (CME) stain is shown. This type Vb‐Vc lesion exhibits eccentric fibrous deposits and a dark area of calcification. The identified areas include dense fibrous tissue (df), calcium deposits (cal), media (med), and fibrous cap (fc). The CME histopathology shows an excellent agreement with the MR images. (With permission from Itskovich et al. 2004 Citation21.)

Figure 2. Illustration of the spatially enhanced iterative cluster analysis for the multi‐contrast ex vivo magnetic resonance (MR) images of human coronary artery specimens. On the left are shown the T1‐, proton density, and T2‐weighted (T1W, PDW and T2W, respectively) axial plane images that were assigned red (R), green (G), and blue (B) colours, respectively, and combined to yield a colour composite and pseudo‐colour cluster image of the coronary artery with colour distributions corresponding to different tissue types. On the right a comparable histopathology image via a combined Masson's elastic (CME) stain is shown. This type Vb‐Vc lesion exhibits eccentric fibrous deposits and a dark area of calcification. The identified areas include dense fibrous tissue (df), calcium deposits (cal), media (med), and fibrous cap (fc). The CME histopathology shows an excellent agreement with the MR images. (With permission from Itskovich et al. 2004 Citation21.)

In apoE‐/‐ mice, MRI has been shown to accurately quantify aortic atherosclerosis in vivoCitation24. Importantly for genetic and pharmacological studies of plaque composition in the mouse models of atherosclerosis, a new method, based on ex vivo three‐dimensional MRI at 11.7 T and at a resolution of 47×47×62.5 µm3 in apoE‐/‐ mice, was recently developed that enables accurate quantification of lipid‐rich/necrotic core and cell‐rich cap areas in atherosclerotic lesions Citation25.

While the non‐invasive MRI of sub‐mm aortic plaques tackles such problems as obtaining sufficient sensitivity and exclusion of artefacts due to respiratory motion and blood flow, the ultimate goal to image coronary artery plaques tends to amplify all these problems Citation20. However, human coronary artery lumen and wall imaging has been achieved in vivo by the so‐called black‐blood MRI method, the application of which maximizes the signal from static tissue and results in a signal void in blood Citation26. In addition, three‐dimensional black‐blood MRI of human coronary vessel wall has detected positive arterial remodelling in patients with non‐significant CAD Citation27. These results suggest that non‐invasive in vivo MRI investigations of human coronaries has potential to improve risk stratification in individual patients, thereby being able to guide and follow‐up treatment options and ultimately to help in preventing ACSs.

Due to its non‐invasiveness MRI is particularly well suited for in vivo follow‐up and pharmacological studies. An MRI study of the participants of the Framingham Heart Study offspring cohort, who were free of clinically apparent CAD, identified aortic atherosclerosis in 38% of women and in 41% of men. The prevalence of atherosclerosis increased with age and was more apparent in the abdominal than in the thoracic aorta Citation28. MRI can also detect lowered aortic distensibility and impaired flow‐mediated vasodilation indicative of endothelial dysfunction in young healthy smokers Citation29. These data demonstrate the ability of MRI in detecting subclinical atherosclerotic disease and to assist in the risk stratification and treatment of patients with an asymptomatic disease.

The clinical significance of carotid plaque characterization has been demonstrated in a case‐control study of patients undergoing carotid endarterectomy: a recent (within 90 days) history of transient ischaemic attack or stroke was strongly associated with the presence of thin or ruptured plaque identified preoperatively by MRI. The risk of recent ischaemic neurological symptoms was increased by a remarkable 23‐fold in cases in which ruptured plaque was identified compared with a thick fibrous cap Citation30. These observations are enhancing the use of MRI in prospective evaluation of plaque behaviour. However, in clinical practice detection of fibrotic caps or plaque components is not currently used, but the treatment decision for endarterectomy is based on the luminal stenosis of carotid arteries according to clinical multi‐centre trials such as the European Carotid Surgery Trial Citation31.

Statins are widely used to treat hypercholesterolaemia and atherosclerotic disease. A regression of aortic as well as carotid plaques after lipid‐lowering therapy with simvastatin has been demonstrated using MRI. After 12 months of treatment, MRI showed that statistically significant reductions occurred in vessel wall thickness and vessel wall area both in carotid arteries and aorta, but no increase in vessel lumen area was detected. The increase in lumen size measurement achieved statistical significance after 18 and 24 months of treatment for both carotid and aortic lesions Citation32. More recently, 1‐year lipid‐lowering therapy with 20‐mg atorvastatin was demonstrated to induce a significant plaque regression in the thoracic aorta (along a marked LDL cholesterol reduction). In the abdominal aorta, even the 20‐mg dose resulted in only a retardation of plaque progression, and a significant progression was observed with the 5‐mg dose treatment Citation33. These results demonstrate that MRI can be used to monitor the effects of pharmacological interventions to atherosclerosis and suggest plaque‐specific follow‐up to optimize the individual treatment options. However, monitoring the effects within an individual patient calls for high inter‐study reproducibility that may not be currently available in clinical routine at 1.5 T.

It has also been revealed, using MRI and multivariate analysis, that LDL cholesterol levels correlate with the plaque extent in the thoracic aorta but not in the abdominal aorta Citation34. Therefore, plaque formation in the thoracic aorta appears to be more closely associated with hypercholesterolaemia than in the abdominal aorta. Hence, lipid‐lowering therapy is more likely to be effective for plaque regression in the thoracic aorta. In general, these data point towards a specific role of plasma lipoproteins in the progression and regression of atherothrombotic plaques and thereby call for follow‐up studies that combine quantification of plasma lipoprotein levels and plaque components. A genuinely new MR approach would be the use of in vitro1H MRS of plasma for the lipoprotein subclass quantification Citation5,Citation15 and in vivo multi‐contrast MRI to characterize the plaque components Citation20; see .

It has recently been shown in rabbits at 2 T that MRI can be used to monitor progression and regression of atherosclerosis also in a short‐term model (cholesterol feeding and normal diet both for 12 weeks). The results indicated that positive remodelling, a process in which the arteries compensate for the progressive growth of atherosclerotic plaques by increasing their external circumference and thereby maintaining the lumen diameter, occurs early during progression and persists through regression of atherosclerotic lesions Citation35. An interesting finding, also from an MRI study of rabbits treated with thromboxane A2 receptor inhibitor (to potentially improve endothelial function and to reduce the inflammatory component of atherosclerosis in addition to antiplatelet activity), is that regression of already established advanced atherosclerotic lesions occurs after 6 months of treatment while maintaining the atherogenic diet. Also, a change in the phenotype of the atherosclerotic plaques was observed in the treatment group using histological stainings Citation36. The reduction in the content of macrophages, apoptotic cells, metalloproteinases and endothelin‐1, and the increase in vascular smooth muscle cells, suggested that selective inhibition of thromboxane A2 pathway may transform lesions towards a more stable plaque phenotype. These findings also demonstrate the additional value of non‐invasive MR techniques, enabling molecular and cellular specification, in the clarification and follow‐up of pathophysiological processes of atherothrombosis in vivo.

Plaque imaging with the aid of exogenous contrast agents

Contrast agents serve as a fascinating option to enhance the capability of MRI and MRA in cardiovascular imaging and they have been extensively used in the detection of ischaemic myocardial injuries and vascular stenosis Citation37. A typical MR contrast agent, such as widely clinically used gadolinium chelate Gd‐DTPA (gadopenetate, Magnevist), is paramagnetic material containing unpaired electrons, which influence the relaxation properties and thereby the intensity (contrast) of the detectable nuclei. The MR contrast agents (unlike in X‐ray studies) are not directly observable. The MR contrast agents are a heterogeneous group of molecules, the effects of which in the actual MR experiment depend on, e.g., the structural properties, dosage, distribution, tissue, pulse sequence, and magnetic field strength Citation38.

The advantage of using contrast agents in the MRI studies of atherothrombosis is two‐fold. First, they may improve tissue contrast between the plaque components as illustrated in showing an example of Gd‐DTPA‐enhanced MRI differentiation of a fibrous cap Citation39. Second, the distribution of contrast agents in the body as well as the time course of observed contrast may provide valuable structural and functional information in tissue characterizations, such as the extent of vascularization and the size of an intercellular matrix. In MRA gadolinium is used to enhance the blood‐tissue contrast, i.e., to improve the detection of anatomical details such as a vessel lumen.

Figure 3. A T1‐weighted image from a patient with abdominal aortic aneurysm demonstrating bright signal within thrombus and darker signal in lipid core towards vessel wall (on the left). The cap region is faintly seen on the T1‐weighted image. A comparable T1‐weighted image, but several minutes after infusion of Gd‐DTPA, is shown on the right. Note the improved delineation of fibrous cap by bright signal within (arrow). (With permission from Kramer et al. 2004 Citation39.)

Figure 3. A T1‐weighted image from a patient with abdominal aortic aneurysm demonstrating bright signal within thrombus and darker signal in lipid core towards vessel wall (on the left). The cap region is faintly seen on the T1‐weighted image. A comparable T1‐weighted image, but several minutes after infusion of Gd‐DTPA, is shown on the right. Note the improved delineation of fibrous cap by bright signal within (arrow). (With permission from Kramer et al. 2004 Citation39.)

In atherothrombosis new microvessels form that may be associated with local inflammation Citation3. The presence of new vessels has also been associated with carotid plaque instability, and in addition they have been connected to abnormal permeation of plasma proteins, such as albumin and fibrinogen Citation37. Using a gadolinium‐based contrast agent in MRI of carotid arteries, plaque microvascularization was found to affect the contrast enhancement thereby suggesting potential improvement in the detection of vulnerable carotid plaque Citation40. Using MS‐325, a gadolinium‐based contrast agent that binds albumin, areas of high signal intensity have been observed in the aortic or iliac arterial wall. This finding may not only reflect increased plaque vascularity but also leaking of these microvessels due to active inflammatory processes Citation37.

The most extensively used MR contrast agent Gd‐DTPA is highly hydrophilic and thus does not penetrate cellular membranes. In contrast, gadofluorine, also a gadolinium‐based contrast agent, is lipophilic. As a matter of fact, gadofluorine has recently been shown to enhance MRI of atherosclerotic plaques and to improve plaque detection in non‐stenotic lesions, not visible when using endogenous contrast Citation41. It has also been suggested that gadofluorine has a high affinity towards lipid‐rich plaque areas Citation42. On the other hand, a recent study using a Gd‐DTPA‐type of a contrast agent reports good results in quantifying intact fibrous cap and lipid‐rich necrotic core size in human atherosclerotic carotid plaques Citation43.

Molecular targeted MRI

The detection of contrast in conventional MRI relies on anatomical, physiological, or metabolic heterogeneity in the tissues. On the other hand, the rising interest in applying ‘molecular imaging’ is based on an assumption that disease processes, i.e., atherothrombosis in this particular case, can be detected and characterized via specific detections along the molecular and cellular development and pathways. It has been proposed that molecular imaging offers an in vivo equivalent to immunohistochemistry, in situ hybridization or in situ zymography Citation1.

The general idea in molecular imaging is to target the specific molecule of interest using a specific ligand, which is conjugated to a carrier particle loaded with signal affecting elements. For example, an αvβ3‐integrin may be targeted using a monoclonal antibody conjugated to a suitable nanoparticle loaded with paramagnetic chelates. In an effective situation the carrier particles, and thereby also the MR signal‐affecting elements, accumulate in the vicinity of the target molecules in the tissue thus potentially modifying the detectable signal. Though the molecular imaging appears very appealing there are several pitfalls in both the biological and MRI side of potential applications Citation1,Citation12,Citation37. The contrast agent must identify the target tissue with high specificity and induce distinctive signal changes within the corresponding voxels in comparison to voxels containing untargeted tissue. It is notable that if only a small number of paramagnetic chelates are conjugated to the targeting molecule, sufficient contrast is difficult to achieve. Therefore, specific nanoparticle approaches have been developed in which extensive shipment of paramagnetic or superparamagnetic agents is feasible Citation1. However, a limitation of the nanoparticle approach is the steric hindrance imposed by such relatively large particles. Also the issues of toxicity and systemic clearance of the agent are important. The following chapters briefly introduce a few recent molecular targeted MRI studies of fundamental importance in atherothrombosis research.

Angiogenesis and αvβ3‐integrins—perfluorocarbon nanoparticles

Angiogenesis is involved in the early phases of atherosclerotic plaque development and it may also be one of the issues relating to vulnerable plaque. The αvβ3‐integrin is expressed on endothelial cells during angiogenesis, but not in the resting state, and atherosclerotic lesions are known to be highly vascular in comparison to normal vessel tissue Citation44. An MRI application at 1.5 T using αvβ3‐integrin‐targeted, paramagnetic perfluorocarbon nanoparticles in hyperlipidaemic rabbits was successful in detecting angiogenesis within the aortic wall (but it could not be spatially resolved to a specific vascular layer, e.g., adventitia) Citation45. The nanoparticles used were ∼273 nm in diameter and contained ∼94 200 Gd3+‐atoms per particle. These data indicated that αvβ3‐integrin‐targeted, paramagnetic nanoparticles might be used for molecular imaging of neovascularization throughout the vascular wall and that αvβ3‐integrin expression shows considerable heterogeneity within individual aortic segments substantiating that early atherosclerosis evolves as a diffuse process. These findings are demonstrated in . Variation in the gross severity of atherosclerosis from the diaphragm to the renal arteries generally corresponded to the calculated MRI signal enhancement and the magnitude of neovascular proliferation observed histologically. These findings are in agreement with an analogous heterogeneous distribution of aortic foam cells in cholesterol‐fed rabbits Citation46,47. In contradiction to the MRI results with αvβ3‐integrin‐targeted nanoparticles, delayed contrast enhancement of the vessel wall with Gd‐DTPA revealed no significant difference between cholesterol‐fed and control diet rabbits. Thus, non‐specific contrast agents seem insensitive to the subtle vascular changes associated with the early development of atherosclerosis.

Figure 4. A magnetic resonance imaging(MRI) application at 1.5 T using αvβ3‐integrin‐targeted, paramagnetic perfluorocarbon nanoparticles in a hyperlipidaemic rabbits to detect angiogenesis within the aortic wall. Per cent enhancement maps (false‐coloured from blue to red) are illustrated from individual aortic segments at renal artery (A), mid‐aorta (B), and diaphragm (C) 2 hours after infusion of the αvβ3‐integrin‐targeted nanoparticles. (With permission from Winter et al. 2003 Citation45.)

Figure 4. A magnetic resonance imaging(MRI) application at 1.5 T using αvβ3‐integrin‐targeted, paramagnetic perfluorocarbon nanoparticles in a hyperlipidaemic rabbits to detect angiogenesis within the aortic wall. Per cent enhancement maps (false‐coloured from blue to red) are illustrated from individual aortic segments at renal artery (A), mid‐aorta (B), and diaphragm (C) 2 hours after infusion of the αvβ3‐integrin‐targeted nanoparticles. (With permission from Winter et al. 2003 Citation45.)

In addition to the detection of neovascularization in early atherosclerotic lesions, it has been demonstrated that αvβ3‐integrin‐targeted nanoparticles can also be used to specifically and locally deliver potent pharmaceutical agents, such as fumagillin, to elicit a marked antiangiogenic effect with even a single dose Citation44. Atherosclerotic rabbits were treated with αvβ3‐integrin‐targeted nanoparticles including 0 or 0.2 mole % fumagillin. The enhancement of the MRI signal, averaged over all imaged slices from the renal artery to the diaphragm at 2 hours after injection, was used as an integrated, quantitative assessment of the atherosclerotic burden. This was noted identical at baseline for animals treated with or without fumagillin. One week later, reassessment of residual aortic angiogenic activity with αvβ3‐integrin‐targeted paramagnetic nanoparticles (with no drug) revealed that MR signal enhancement in rabbits given αvβ3‐integrin‐targeted nanoparticles including fumagillin was markedly reduced whereas the MR signal from the neovasculature in control animals was unchanged. Thus, it has been postulated that αvβ3‐integrin‐targeted nanoparticles are an entity that might be used for specific detection, treatment and follow‐up of early atherosclerotic lesions Citation44.

Macrophages—ultrasmall particles of iron oxide

Iron oxides are superparamagnetic materials with potentially huge effects on the relaxation contrast of protons. A wide variety of iron oxide‐based nanoparticles have been developed that differ in hydrodynamic particle size and surface coating material Citation44. In general, these particles are categorized into superparamagnetic iron oxides (SPIOs) (diameter from 50 to 500 nm) and ultrasmall superparamagnetic iron oxides (USPIOs) (diameter <50 nm), the size of which has an important effect on their physicochemical and pharmacokinetic properties. USPIOs provide adequate circulation time for tissue penetration (the half‐life of USPIOs in human circulation is ∼30 hours) but they also require significant time, usually 24 hours, for sufficient background signal clearance. Because these agents often create an extended image void, the proximate anatomy surrounding key targeted points of interest may be obscured, particularly in high‐resolution MRI. This opposes the behaviour of paramagnetic contrast agents that can create rapid contrast enhancement signal which also allows for subtle tissue contrast features to be clearly discerned at high spatial resolutions.

Contrast agents which specifically identify components of vulnerable plaque are of considerable interest. Morphological characteristics of vulnerability are a predominance of macrophages, ulceration, intraplaque haemorrhage, and thrombosis Citation3. Angiogenic vessels within atherosclerotic plaques or inflammatory allografts permit extravasation of USPIOs, which are phagocytosed by resident tissue macrophages. The accumulated iron oxide is readily visualized in MRI as regions of signal void, which grossly correlate spatially with active atherosclerosis Citation48.

In an MRI study of rabbits, conventional extracellular contrast agent Gd‐DOTA (gadoterate, Dotarem) failed to reveal any abnormality in either the hyperlipidaemic or control rabbits in the aortic lumen before administration of USPIOs. In case of hyperlipidaemic rabbits, 5 days after the administration of USPIOs, the aortic wall exhibited marked irregularities in MRI that was not observed in controls Citation46. Histopathological analysis showed uptake of Fe‐particles in the plaque macrophages of the hyperlipidaemic rabbits but not in the controls. Fe‐particles are known to cause susceptibility effects and this can explain signal intensity irregularities in the aortic wall in the MRI of hyperlipidaemic rabbits. Interestingly, visual examination of the aortic walls of hyperlipidaemic as well as control rabbits did not reveal any appreciable irregularities. Thus, accumulation of USPIOs in macrophages in the aortic wall seems to provide delicate structural information on atherosclerotic processes beyond visual inspection. These observations may have some clinical relevance since increased macrophage activity is associated with active inflammation, a marker of vulnerable plaque Citation3,Citation49.

In a retrospective analysis in bladder or prostate cancer patients who had originally received USPIOs for staging lymph node metastases, signal loss in the arterial wall was observed in 7 of 19 patients, indicating some accumulation of USPIOs in human atherosclerotic plaques Citation50. In another study, in which MRI was performed before and after administration of USPIOs on 11 symptomatic patients, scheduled for carotid endarterectomy, histology and electron microscopy analyses of the plaques showed USPIOs primarily in macrophages within the plaques in 10 of 11 patients Citation48. Histology showed USPIOs in macrophages of carotid plaques in 75% of the ruptured and rupture‐prone lesions and in 7% of the stable lesions. Of the patients with USPIO uptake, significant signal decrease in the post‐USPIO MRI were observed in the vessel wall in 54% of the quadrants after 24 hours. These results indicate that the tested USPIOs accumulate predominantly in macrophages in ruptured and rupture‐prone human atherosclerotic lesions and that this can induce significant signal changes in in vivo MRI 24 hours after intravenous administration of USPIOs. Thus, USPIO‐enhanced MRI seems to be a promising way to identify high‐risk plaques.

In human studies the dosages of USPIOs have been noticeably smaller than in animal studies. As a result, the observed signal intensity reductions are relatively minor calling for particular attention on image analysis and reproducibility Citation51. In addition, the predictive value of USPIO‐enhanced plaque imaging to detect abnormality associated with increased stroke risk has to be confirmed in a prospective study. Nevertheless, the current evidence suggests that the USPIO‐enhanced imaging has a considerable potential for becoming a part of clinical routine in the assessment of patients with carotid atherosclerosis. The use of USPIOs has clear pathophysiological background. The histopathological and experimental studies have also confirmed the capability of USPIOs to indicate regional inflammation using clinical scanners.

Macrophages—recombinant high‐density‐lipoprotein‐like nanoparticles

A quite common problem with many available paramagnetic MR contrast agent constructs is that they are not capable of delivering a sufficiently large amount of gadolinium ions to the tissue in order to induce detectable changes in the MRI contrast. Also, some of the MR contrast agents may be too large to have free access to biochemical epitopes within the vascular subendothelium of atherosclerotic plaques. Recently a high‐density lipoprotein (HDL)‐like nanoparticle contrast agent, that selectively targets atherosclerotic plaques, has been introduced Citation52. The HDL particles are the key players in reverse cholesterol transport and have several advantages: a small size (diameter of 7–12 nm); protein components that are endogenous, biodegradable, and do not trigger immunoreactions; and the particles are not recognized by the reticuloendothelial system. Furthermore, HDL‐like particles can easily be reconstituted and they can carry a considerable contrast agent pay‐load.

Thus, reconstituted HDL particles (rHDL) of approximately 9 nm diameter were prepared containing 15–20 molecules of Gd‐DTPA‐DMPE (1,2‐dimyristoyl‐sn‐glycero‐3‐phosphoethanolamine diethylenetriaminepentaacetic acid), a phospholipid‐based contrast agent, and also a fluorescent phospholipid with a green emission, NBD‐DPPE (1,2‐dipalmitoyl‐sn‐glycero‐3‐phosphoethanolamine‐N‐(7‐nitro‐2,1,3‐benzoxadiazol‐4‐yl)), for confocal fluorescence microscopy Citation52. The general structure of the rHDL particles is shown in . In hyperlipidaemic apoE‐knockout mice, sequential MRI showed that Gd‐DTPA‐DMPE localized predominantly at the atherosclerotic plaque by 24 h after injection (see ). Furthermore, by 48 h after rHDL injection, the intensity of the plaque decreased to a value similar to that observed for the plaque immediately after injection. Importantly, the enhancement was also related to plaque composition: the more cellular the content was (as judged by histology), the more intense was the signal. At 24 h post‐injection, when MRI of the rHDL‐like contrast agent showed its maximum intensity in plaques, aortas were removed and imaged by confocal fluorescence microscopy. The fluorescence was located mainly in the intimal layer, where the lipids accumulate and a small number of cells had internalized and retained the fluorescence. Histology characterization identified these cells primarily as macrophages.

Figure 5. An application of a high‐density lipoprotein (HDL)‐like nanoparticle contrast agent for macrophage‐targeted in vivo magnetic resonance imaging (MRI). Phospholipid‐based contrast agent Gd‐DTPA‐DMPE and NBD‐DPPE‐labelled phospholipid for fluorescence confocal microscopy were used for the reconstruction of HDL. The MR images of an apolipoprotein E knockout mouse at 1.5 T illustrate the pre‐ and 24 h post‐injection of contrast agent at a rHDL dose of 4.36 μmol/kg. White arrows point to the abdominal aorta; the insets denote a magnification of the aorta region. (Modified from Frias et al. 2004 Citation52 with permission.)

Figure 5. An application of a high‐density lipoprotein (HDL)‐like nanoparticle contrast agent for macrophage‐targeted in vivo magnetic resonance imaging (MRI). Phospholipid‐based contrast agent Gd‐DTPA‐DMPE and NBD‐DPPE‐labelled phospholipid for fluorescence confocal microscopy were used for the reconstruction of HDL. The MR images of an apolipoprotein E knockout mouse at 1.5 T illustrate the pre‐ and 24 h post‐injection of contrast agent at a rHDL dose of 4.36 μmol/kg. White arrows point to the abdominal aorta; the insets denote a magnification of the aorta region. (Modified from Frias et al. 2004 Citation52 with permission.)

Fibrin—various approaches

Plaque rupture with subsequent thrombosis is a common underlying pathophysiology in ACS and stroke. Direct in vivo visualization of thrombi would thus be beneficial both for diagnosis and follow‐up of therapy. Therefore, there has recently been an active search for fibrin‐targeted MRI contrast agents as an aid for vulnerable plaque detection. The successful approaches include fibrin‐targeted paramagnetic perfluorocarbon nanoparticles Citation53 as well as fibrin‐binding gadolinium‐labelled peptides EP‐1873 Citation54 and EP‐1242 Citation55. In recent studies, a remarkable progression in the detection of the components of ruptured or highly vulnerable plaque has been achieved. It has been demonstrated that imaging with a Gd‐based fibrin‐binding contrast agent, EP‐2104R, can specifically detect thrombotic material with platelets and fibrin Citation56,57. In the aforementioned studies tissue contrast on images has been promising, suggesting the method to be sensitive enough to detect thrombus also in clinical practice. Imaging with EP‐2104R may provide new information on the aetiology and pathogenesis of recent thromboembolic events but its predictive value in case of thrombotic events still remains to be clarified. EP‐2104R is currently being tested in human subjects in phase I and phase II clinical trials Citation57. In addition, quantitative ‘magnetic resonance immunohistochemistry’ with the fibrin‐targeted perfluorocarbon nanoparticles has recently been illustrated in the case of an ex vivo human carotid endarterectomy sample (), showing the potential use of 19F imaging and spectroscopy for identifying unstable atherosclerotic lesions which exhibit microdeposits of fibrin indicative of the possibility for a future rupture Citation58.

Figure 6. Illustration of quantitative‘magnetic resonance immunohistochemistry’ with the fibrin‐targeted perfluorocarbon nanoparticles in the case of an ex vivo human carotid endarterectomy sample: (a) An optical image of a 5‐mm cross‐section of a human carotid endarterectomy sample. This section showed moderate luminal narrowing as well as several atherosclerotic lesions. (b) A 19F projection image acquired at 4.7 T through the entire carotid artery sample shows high signal along the lumen due to nanoparticles bound to fibrin. (c) A concentration map of bound nanoparticles in the carotid sample. (With permission from Morawski et al. 2004 Citation58.)

Figure 6. Illustration of quantitative‘magnetic resonance immunohistochemistry’ with the fibrin‐targeted perfluorocarbon nanoparticles in the case of an ex vivo human carotid endarterectomy sample: (a) An optical image of a 5‐mm cross‐section of a human carotid endarterectomy sample. This section showed moderate luminal narrowing as well as several atherosclerotic lesions. (b) A 19F projection image acquired at 4.7 T through the entire carotid artery sample shows high signal along the lumen due to nanoparticles bound to fibrin. (c) A concentration map of bound nanoparticles in the carotid sample. (With permission from Morawski et al. 2004 Citation58.)

Miscellaneous

The new field of molecular targeted MRI is in an active phase of continuous development, and the role of nanotechnology in detecting the various phases of atherothrombosis is rapidly expanding Citation59. In addition to the abovementioned applications there are several emerging techniques that are also likely to find their way to the studies of atherothrombosis. These include in vivo MRI visualization of gene expression and detection of stem cells Citation60. Contrast agents targeted to endothelial cell adhesion molecules as well as to enzymatic and proteolytic processes in the extracellular matrix are also likely to benefit atherosclerosis research Citation1. Moreover, ‘smart’ agents (that undergo a physicochemical change due to their intended molecular interaction with the target) and the holistic antibody‐mediated targeting of a certain disease stage‐related characteristic vascular proteins (applying the phage display method) are fascinating options likely to induce the development of molecular specific ways for the imaging of atherothrombotic processes Citation1,Citation60. It is also currently envisioned that the site‐directed delivery of therapeutic substances, concomitantly with the detection of early atherosclerotic complications, may well have a substantial impact on clinical cardiology in the near future Citation59.

Magnetic resonance spectroscopy

Lipoprotein subclasses by 1H MRS of plasma

In the mid‐1990s two research groups independently developed 1H MRS‐based lipoprotein lipid quantification methodology Citation61,62, and later on several groups have been assessing this option Citation63. One of the original approaches Citation61 was developed for a commercial assay, named NMR LipoProfile® by LipoScience Inc. According to the company it has performed over one million NMR LipoProfile® tests (http://www.liposcience.com). In contrast to other existing methods of lipoprotein subclass quantification, involving both physical separations of the subclasses (e.g., ultracentrifugation) and separate determinations of subclass specific lipid values (e.g., enzymatic measurements of cholesterol), the 1H MRS method relies on a single spectrum of a plasma sample and subsequent mathematical deconvolution of the subclass specific information. Recent clinical applications of the MRS‐based methodology have evidently shown its usefulness in biomedical research Citation5.

Is 1H MRS of serum able to diagnose coronary heart disease?

It has been suggested that pattern recognition techniques applied to 1H MR spectra of human serum could diagnose not only the presence, but also the severity, of coronary heart disease (CHD) Citation64. Results were reported that more than 90% of subjects with stenosis of all three major coronary vessels were distinguished from subjects with angiographically normal coronary arteries, with specificity greater than 90%. According to the current understanding of the complexity of atherothrombosis and the array of clinical data on lipoprotein lipids and their relation to CHD, the above indication is quite unexpected. Particularly, since conventional 1H MRS characteristics were used resulting in spectra that many independent research groups have shown to reflect mostly variations in the concentrations of lipoprotein subclasses, the variations of which are not statistically distinctive for diagnostic purposes Citation15. Also the study group of patients was very limited and not indicative of clinical patient material which is likely to explain the fortuitous conclusion concerning 1H MRS‐based CHD diagnosis Citation64. (A note added in proof: A report to support these conclusions has recently been appeared stating that 1H MRS analysis of plasma is only a weak predictor of CAD (Kirschenlohr HL, Griffin JL, Clarke SC, Rhydwen R, Grace AA, Schofield PM, et al. Proton NMR analysis of plasma is a weak predictor of coronary artery disease. Nat Med. 2006;12:705–10.).)

On the other hand, 1H MRS of plasma has been shown to detect a proatherogenic state in healthy, non‐diabetic subjects who subsequently develop diabetes Citation5. Furthermore, it has recently been indicated that an overall measure combining information from seven lipids (total, HDL, LDL and VLDL cholesterol, triglycerides, apolipoprotein A‐I, and apolipoprotein B) is a superior predictor of CHD risk compared with conventional lipid measures Citation65—since the 1H MR spectrum of plasma is currently known to contain not only these data but specific information on lipoprotein subclasses and on an array of other metabolites Citation15—it would be unexpected if the total 1H MR spectrum of plasma would not lead to good estimation and classification of atherosclerosis risks. Thus, as also suggested in , the potential usage of in vitro1H MRS is in the risk evaluation of atherothrombotic events, the true clinical value of which also remains to be elucidated in extensive clinical studies.

Molecular characterization of plaque—1H and 13C MRS

The molecular characteristics of various atherosclerotic plaques have been studied using 1H and 13C MR spectroscopy ex vivoCitation66,67. Interestingly, 13C MRS of human arterial plaques revealed the relation of decreased polyunsaturation for increased stenosis Citation68. In the light of recent evidence, that apoptosis would be an indicator of atherothrombotic processes Citation69 and that in vivo1H MRS can detect apoptosis‐related accumulation of polyunsaturated fatty acids Citation70, MRS studies might be useful in figuring out the relation of apoptosis to the development of atherothrombosis and to plaque vulnerability in vivo. In relation to animal models of atherosclerosis it is good to note that 1H MRS has shown clear molecular differences between rabbit and human plaques Citation71.

The lipid components of atherosclerotic plaques are abundant in liquid‐like cholesterol esters (CEs) derived largely from apolipoprotein B‐100 containing lipoproteins Citation72. Exploiting this, a study demonstrating the usefulness of MR image‐guided, single‐voxel ex vivo1H MRS to characterize lipid‐rich and lipid‐poor regions of atherosclerotic plaque (see ) has just appeared Citation73. This procedure may show the way for non‐invasive detection and quantification of CEs in atherosclerotic plaques in vivo since preliminary work at 3.0 T on human carotid artery atherosclerosis has given promising results Citation73. Another recently established methodology, based on in vivo1H MRS with cardiac and respiratory gating, for myocardial lipid quantification in humans, supports the future rise of cardiac applications of in vivo1H MRS Citation74. It is also anticipated that in addition to traditional individual signal quantifications, a holistic approach of MR metabonomics will find its way to in vivo MRS.

Figure 7. Demonstration of the magnetic resonance(MR) image‐guided, single‐voxel ex vivo1H magnetic resonance spectroscopy (MRS) to characterize lipid‐rich and lipid‐poor regions of atherosclerotic plaque. Spectra elicited from 1‐mm3 voxels are illustrated with insets showing the parametric map and voxel position for reference. Panels A and C illustrate the intense water signal without water suppression. Weak lipid signals are evident in panel B only after vertical expansion (inset). In contrast, strong lipid signals (consistent with cholesterol esters) are seen in panel D. (With permission from Ruberg et al. 2006 Citation73.)

Figure 7. Demonstration of the magnetic resonance(MR) image‐guided, single‐voxel ex vivo1H magnetic resonance spectroscopy (MRS) to characterize lipid‐rich and lipid‐poor regions of atherosclerotic plaque. Spectra elicited from 1‐mm3 voxels are illustrated with insets showing the parametric map and voxel position for reference. Panels A and C illustrate the intense water signal without water suppression. Weak lipid signals are evident in panel B only after vertical expansion (inset). In contrast, strong lipid signals (consistent with cholesterol esters) are seen in panel D. (With permission from Ruberg et al. 2006 Citation73.)

Combined MR schemes for individual diagnostics—a future strategy?

It would be advantageous to detect molecular and cellular processes related to the early stages of developing atherosclerosis. This would clinically facilitate early individual primary prevention and also give a personal rationale to comply with life‐style modifications and potential drug therapies. The recent applications of MR in atherothrombosis research, as indicated in this review, suggest the potential usage of in vitro MRS for risk assessment and of in vivo MRI for direct detection of plaque composition and vulnerability. The scheme presented in can be seen as one option to elucidate the potential of MR in detecting individual intermediate atherothrombotic end‐points and utilizing their prognostic value before the occurrence of a definite end‐point. Molecular targeted MRI also shows notable possibilities for the non‐invasive detection of early atherothrombotic processes at various pre‐clinical stages of atherosclerosis. The field of MR technologies, in combination with nanotechnology and bioinformatics, is progressing rapidly. Consequently, it is tempting to envision that combinatorial MRI and MRS schemes could be operational at the clinic in the near future.

Acknowledgements

Dr Juhana Hakumäki is acknowledged for fruitful discussions about molecular imaging and MRI technology. This study was supported by the Academy of Finland.

References

  • Choudhury R. P., Fuster V., Fayad Z. A. Molecular, cellular and functional imaging of atherothrombosis. Nat Rev Drug Disc 2004; 3: 913–25
  • Pentikäinen M. O., Öörni K., Ala‐Korpela M., Kovanen P. T. Modified LDL—trigger of atherosclerosis and inflammation in the arterial intima. J Intern Med 2000; 247: 359–70
  • Fuster V., Moreno P. R., Fayad Z. A., Corti R., Badimon J. J. Atherothrombosis and high‐risk plaque. Part I: Evolving concepts. J Am Coll Cardiol 2005; 46: 937–54
  • Opie L. H., Commerford P. J., Gersh B. J. Controversies in stable coronary artery disease. Lancet 2006; 367: 69–78
  • Festa A., Williams K., Hanley A. J., Otvos J. D., Goff D. C., Wagenknecht L. E., et al. Nuclear magnetic resonance lipoprotein abnormalities in prediabetic subjects in the Insulin Resistance Atherosclerosis Study. Circulation 2005; 111: 3465–72
  • Naghavi M., Libby P., Falk E., Casscells S. W., Litovsky S., Rumberger J., et al. From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: Part I. Circulation 2003; 108: 1664–72
  • Naghavi M., Libby P., Falk E., Casscells S. W., Litovsky S., Rumberger J., et al. From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: Part II. Circulation 2003; 108: 1772–8
  • Fuster V. Editorial: The evolving role of CT and MRI in atherothrombotic evaluation and management. Nat Clin Pract Cardiovasc Med 2005; 2: 323
  • Kim W. Y., Danias P. G., Stuber M., Flamm S. D., Plein S., Nagel E., et al. Coronary magnetic resonance angiography for the detection of coronary stenoses. N Engl J Med 2001; 345: 1863–9
  • Glagov S., Weisenberg E., Zarins C. K., Stankunavicius R., Kolettis G. J. Compensatory enlargement of human atherosclerotic coronary arteries. N Engl J Med 1987; 316: 1371–5
  • Little W. C., Constantinescu M., Applegate R. J., Kutcher M. A., Burrows M. T., Kahl F. R., et al. Can coronary angiography predict the site of a subsequent myocardial infarction in patients with mild‐to‐moderate coronary artery disease?. Circulation 1988; 78: 1157–66
  • Lipinski M. J., Fuster V., Fisher E. A., Fayad Z. A. Technology insight: targeting of biological molecules for evaluation of high‐risk atherosclerotic plaques with magnetic resonance imaging. Nat Clin Pract Cardiovasc Med 2004; 1: 48–55
  • Winslow R. D., Mehta D., Fuster V. Sudden cardiac death: mechanisms, therapies and challenges. Nat Clin Pract Cardiovasc Med 2005; 2: 352–60
  • Raggi P., Taylor A., Fayad Z., O'Leary D., Nissen S., Rader D., et al. Atherosclerotic plaque imaging: contemporary role in preventive cardiology. Arch Intern Med 2005; 165: 2345–53
  • Ala‐Korpela M. 1H NMR spectroscopy of human blood plasma. Progr Nucl Magn Reson Spectr 1995; 27: 475–554
  • Lardo A. C., Fayad Z. A., Chronos N. A. F., Fuster V., editors. Cardiovascular magnetic resonance. Established and emerging applications. Martin Dunitz, London, New York 2003
  • Nagel E., van Rossum A. C., Fleck E., editors. Cardiovascular magnetic resonance. Steinkopff Verlag, Darmstadt 2004
  • Mollet N. R., Cademartiri F., van Mieghem C. A., Runza G., McFadden E. P., Baks T., et al. High‐resolution spiral computed tomography coronary angiography in patients referred for diagnostic conventional coronary angiography. Circulation 2005; 112: 2318–23
  • Sakuma H., Ichikawa Y., Suzawa N., Hirano T., Makino K., Koyama N., et al. Assessment of coronary arteries with total study time of less than 30 minutes by using whole‐heart coronary MR angiography. Radiology 2005; 237: 316–21
  • Fuster V., Fayad Z. A., Moreno P. R., Poon M., Corti R., Badimon J. J. Atherothrombosis and high‐risk plaque. Part II: Approaches by noninvasive computed tomographic/magnetic resonance imaging. J Am Coll Cardiol 2005; 46: 1209–18
  • Itskovich V. V., Samber D. D., Mani V., Aguinaldo J. G., Fallon J. T., Tang C. Y., et al. Quantification of human atherosclerotic plaques using spatially enhanced cluster analysis of multicontrast‐weighted magnetic resonance images. Magn Reson Med 2004; 52: 515–23
  • Cappendijk V. C., Cleutjens K. B., Kessels A. G., Heeneman S., Schurink G. W., Welten R. J., et al. Assessment of human atherosclerotic carotid plaque components with multisequence MR imaging: initial experience. Radiology 2005; 234: 487–92
  • Viereck J., Ruberg F. L., Qiao Y., Perez A. S., Detwiller K., Johnstone M., et al. MRI of atherothrombosis associated with plaque rupture. Arterioscler Thromb Vasc Biol 2005; 25: 240–5
  • Choudhury R. P., Aguinaldo J. G., Rong J. X., Kulak J. L., Kulak A. R., Reis E. D., et al. Atherosclerotic lesions in genetically modified mice quantified in vivo by non‐invasive high‐resolution magnetic resonance microscopy. Atherosclerosis 2002; 162: 315–21
  • McAteer M. A., Schneider J. E., Clarke K., Neubauer S., Channon K. M., Choudhury R. P. Quantification and 3D reconstruction of atherosclerotic plaque components in apolipoprotein E knockout mice using ex vivo high‐resolution MRI. Arterioscler Thromb Vasc Biol 2004; 24: 2384–90
  • Fayad Z. A., Fuster V., Fallon J. T., Jayasundera T., Worthley S. G., Helft G., et al. Noninvasive in vivo human coronary artery lumen and wall imaging using black‐blood magnetic resonance imaging. Circulation 2000; 102: 506–10
  • Kim W. Y., Stuber M., Bornert P., Kissinger K. V., Manning W. J., Botnar R. M. 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: 296–9
  • Jaffer F. A., O'Donnell C. J., Larson M. G., Chan S. K., Kissinger K. V., Kupka M. J., et al. Age and sex distribution of subclinical aortic atherosclerosis: a magnetic resonance imaging examination of the Framingham Heart Study. Arterioscler Thromb Vasc Biol 2002; 22: 849–54
  • Wiesmann F., Petersen S. E., Leeson P. M., Francis J. M., Robson M. D., Wang Q., et al. Global impairment of brachial, carotid, and aortic vascular function in young smokers: direct quantification by high‐resolution magnetic resonance imaging. J Am Coll Cardiol 2004; 44: 2056–64
  • Yuan C., Zhang S. X., Polissar N. L., Echelard D., Ortiz G., Davis J. W., et al. Identification of fibrous cap rupture with magnetic resonance imaging is highly associated with recent transient ischemic attack or stroke. Circulation 2002; 105: 181–5
  • European Carotid Surgery Trialists' Collaborative Group. Randomised trial of endarterectomy for recently symptomatic carotid stenosis: final results of the MRC European Carotid Surgery Trial (ECST). Lancet 1998; 351: 1379–87
  • Corti R., Fuster V., Fayad Z. A., Worthley S. G., Helft G., Smith D., et al. Lipid lowering by simvastatin induces regression of human atherosclerotic lesions. Two years' follow‐up by high‐resolution noninvasive magnetic resonance imaging. Circulation 2002; 106: 2884–7
  • Yonemura A., Momiyama Y., Fayad Z. A., Ayaori M., Ohmori R., Higashi K., et al. Effect of lipid‐lowering therapy with atorvastatin on atherosclerotic aortic plaques detected by noninvasive magnetic resonance imaging. J Am Coll Cardiol 2005; 45: 733–42
  • Taniguchi H., Momiyama Y., Fayad Z. A., Ohmori R., Ashida K., Kihara T., et al. In vivo magnetic resonance evaluation of associations between aortic atherosclerosis and both risk factors and coronary artery disease in patients referred for coronary angiography. Am Heart J 2004; 148: 137–43
  • Hegyi L., Hockings P. D., Benson M. G., Busza A. L., Overend P., Grimsditch D. C., et al. Short term arterial remodelling in the aortae of cholesterol fed New Zealand white rabbits shown in vivo by high‐resolution magnetic resonance imaging—implications for human pathology. Pathol Oncol Res 2004; 10: 159–65
  • Viles‐Gonzalez J. F., Fuster V., Corti R., Valdiviezo C., Hutter R., Corda S., et al. Atherosclerosis regression and TP receptor inhibition: effect of S18886 on plaque size and composition—a magnetic resonance imaging study. Eur Heart J 2005; 26: 1557–61
  • Choudhury R. P., Fuster V., Badimon J. J., Fisher E. A., Fayad Z. A. MRI and characterization of atherosclerotic plaque emerging applications and molecular imaging. Arterioscler Thromb Vasc Biol 2002; 22: 1065–74
  • Lombardi M., Aquaro G., Favilli B. Contrast media in cardiovascular magnetic resonance. Curr Pharm Des 2005; 11: 2151–61
  • Kramer C. M., Cerilli L. A., Hagspiel K., DiMaria J. M., Epstein F. H., Kern J. A. Magnetic resonance imaging identifies the fibrous cap in atherosclerotic abdominal aortic aneurysm. Circulation 2004; 109: 1016–21
  • Yuan C., Kerwin W. S., Ferguson M. S., Polissar N., Zhang S., Cai J., et al. Contrast‐enhanced high resolution MRI for atherosclerotic carotid artery tissue characterization. J Magn Reson Imaging 2002; 15: 62–7
  • Barkhausen J., Ebert W., Heyer C., Debatin J. F., Weinmann H. J. Detection of atherosclerotic plaque with Gadofluorine‐enhanced magnetic resonance imaging. Circulation 2003; 108: 605–9
  • Sirol M., Itskovich V. V., Mani V., Aguinaldo J. G. S., Fallon J. T., Misselwitz B., et al. Lipid‐rich atherosclerotic plaques detected by gadofluorine‐enhanced in vivo magnetic resonance imaging. Circulation 2004; 109: 2890–6
  • Cai J., Hatsukami T. S., Ferguson M. S., Kerwin W. S., Saam T., Chu B., et al. In vivo quantitative measurement of intact fibrous cap and lipid‐rich necrotic core size in atherosclerotic carotid plaque. Comparison of high‐resolution, contrast‐enhanced magnetic resonance imaging and histology. Circulation 2005; 112: 3437–44
  • Lanza G. M., Winter P. M., Caruthers S. D., Morawski A. M., Schmieder A. H., Crowder K. C., et al. Magnetic resonance molecular imaging with nanoparticles. J Nucl Cardiol 2004; 11: 733–43
  • Winter P. M., Morawski A. M., Caruthers S. D., Fuhrhop R. W., Zhang H., Williams T. A., et al. Molecular imaging of angiogenesis in early‐stage atherosclerosis with αvβ3‐integrin‐targeted nanoparticles. Circulation 2003; 108: 2270–4
  • Ruehm S. G., Corot C., Vogt P., Kolb S., Debatin J. F. Magnetic resonance imaging of atherosclerotic plaque with ultrasmall superparamagnetic particles of iron oxide in hyperlipidemic rabbits. Circulation 2001; 103: 415–22
  • Colley C. S., Kazarian S. G., Weinberg P. D., Lever M. J. Spectroscopic imaging of arteries and atherosclerotic plaques. Biopolymers 2004; 74: 328–35
  • Kooi M. E., Cappendijk V. C., Cleutjens K. B., Kessels A. G., Kitslaar P. J., Borgers M., et al. Accumulation of ultrasmall superparamagnetic particles of iron oxide in human atherosclerotic plaques can be detected by in vivo magnetic resonance imaging. Circulation 2003; 107: 2453–8
  • Cyrus T., Winter P. M., Caruthers S. D., Wickline S. A., Lanza G. M. Magnetic resonance nanoparticles for cardiovascular molecular imaging and therapy. Expert Rev Cardiovasc Ther 2005; 3: 705–15
  • Schmitz S. A., Taupitz M., Wagner S., Wolf K. J., Beyersdorff D., Hamm B. Magnetic resonance imaging of atherosclerotic plaques using superparamagnetic iron oxide particles. J Magn Reson Imaging 2001; 14: 355–61
  • Trivedi R. A., U‐King‐Im J. M., Graves M. J., Cross J. J., Horsley J., Goddard M. J., et al. In vivo detection of macrophages in human carotid atheroma: temporal dependence of ultrasmall superparamagnetic particles of iron oxide‐enhanced MRI. Stroke 2004; 35: 1631–5
  • Frias J. C., Williams K. J., Fisher E. A., Fayad Z. A. Recombinant HDL‐like nanoparticles: a specific contrast agent for MRI of atherosclerotic plaques. J Am Chem Soc 2004; 126: 16316–7
  • Flacke S., Fischer S., Scott M. J., Fuhrhop R. J., Allen J. S., McLean M., et al. Novel MRI contrast agent for molecular imaging of fibrin: implications for detecting vulnerable plaques. Circulation 2001; 104: 1280–5
  • Botnar R. M., Perez A. S., Witte S., Wiethoff A. J., Laredo J., Hamilton J., et al. In vivo molecular imaging of acute and subacute thrombosis using a fibrin‐binding magnetic resonance imaging contrast agent. Circulation 2004; 109: 2023–9
  • Sirol M., Aguinaldo J. G., Graham P. B., Weisskoff R., Lauffer R., Mizsei G., et al. Fibrin‐targeted contrast agent for improvement of in vivo acute thrombus detection with magnetic resonance imaging. Atherosclerosis 2005; 182: 79–85
  • Botnar R. M., Buecker A., Wiethoff A. J., Parsons E. C., Jr., Katoh M., Katsimaglis G., et al. In vivo magnetic resonance imaging of coronary thrombosis using a fibrin‐binding molecular magnetic resonance contrast agent. Circulation 2004; 110: 1463–6
  • Sirol M., Fuster V., Badimon J. J., Fallon J. T., Moreno P. R., Toussaint J. F., et al. Chronic thrombus detection with in vivo magnetic resonance imaging and a fibrin‐targeted contrast agent. Circulation 2005; 112: 1594–600
  • Morawski A. M., Winter P. M., Yu X., Fuhrhop R. W., Scott M. J., Hockett F., et al. Quantitative “magnetic resonance immunohistochemistry” with ligand‐targeted 19F nanoparticles. Magn Reson Med 2004; 52: 1255–62
  • Wickline S. A., Neubauer A. M., Winter P., Caruthers S., Lanza G. Applications of nanotechnology to atherosclerosis, thrombosis, and vascular biology. Arterioscler Thromb Vasc Biol 2006; 26: 435–41
  • Jaffer F. A., Weissleder R. Seeing within: molecular imaging of the cardiovascular system. Circ Res 2004; 94: 433–45
  • Otvos J. D., Jeyarajah E. J., Bennett D. W., Krauss R. M. Development of a proton nuclear magnetic resonance spectroscopic method for determining plasma lipoprotein concentrations and subspecies distributions from a single, rapid measurement. Clin Chem 1992; 38: 1632–8
  • Ala‐Korpela M., Korhonen A., Keisala J., Hörkkö S., Korpi P., Ingman L. P., et al. 1H NMR‐based absolute quantitation of human lipoproteins and their lipid contents directly from plasma. J Lipid Res 1994; 35: 2292–304
  • Bathen T. F., Krane J., Engan T., Bjerve K. S., Axelson D. Quantification of plasma lipids and apolipoproteins by use of proton NMR spectroscopy, multivariate and neural network analysis. NMR Biomed 2000; 13: 271–88
  • Brindle J. T., Antti H., Holmes E., Tranter G., Nicholson J. K., Bethell H. W., et al. Rapid and noninvasive diagnosis of the presence and severity of coronary heart disease using 1H‐NMR‐based metabonomics. Nat Med 2002; 8: 1439–44
  • Everett C. J., Mainous A. G 3rd., Koopman R. J., Diaz V. A. Predicting coronary heart disease risk using multiple lipid measures. Am J Cardiol 2005; 95: 986–8
  • Toussaint J. F., Pachot‐Clouard M., Kantor H. L. Tissue characterization of atherosclerotic plaque vulnerability by nuclear magnetic resonance. J Cardiovasc Magn Reson 2000; 2: 225–32
  • Peng S., Guo W., Morrisett J. D., Johnstone M. T., Hamilton J. A. Quantification of cholesteryl esters in human and rabbit atherosclerotic plaques by magic‐angle spinning 13C‐NMR. Arterioscler Thromb Vasc Biol 2000; 20: 2682–8
  • Toussaint J. F., Southern J. F., Fuster V., Kantor H. L. 13C‐NMR spectroscopy of human atherosclerotic lesions. Relation between fatty acid saturation, cholesteryl ester content, and luminal obstruction. Arterioscler Thromb 1994; 14: 1951–7
  • Stoneman V. E., Bennett M. R. Role of apoptosis in atherosclerosis and its therapeutic implications. Clin Sci (Lond) 2004; 107: 343–54
  • Hakumäki J. M., Poptani H., Sandmair A. M., Ylä‐Herttuala S., Kauppinen R. A. 1H MRS detects polyunsaturated fatty acid accumulation during gene therapy of glioma: implications for the in vivo detection of apoptosis. Nat Med 1999; 5: 1323–7
  • Trouard T. P., Altbach M. I., Hunter G. C., Eskelson C. D., Gmitro A. F. MRI and NMR spectroscopy of the lipids of atherosclerotic plaque in rabbits and humans. Magn Reson Med 1997; 38: 19–26
  • Öörni K., Posio P., Ala‐Korpela M., Jauhiainen M., Kovanen P. T. Sphingomyelinase induces aggregation and fusion of small very low‐density lipoprotein and intermediate‐density lipoprotein particles and increases their retention to human arterial proteoglycans. Arterioscler Thromb Vasc Biol 2005; 25: 1678–83
  • Ruberg F. L., Viereck J., Phinikaridou A., Qiao Y., Loscalzo J., Hamilton J. A. Identification of cholesteryl esters in human carotid atherosclerosis by ex vivo image‐guided proton magnetic resonance spectroscopy. J Lipid Res 2006; 47: 310–7
  • Szczepaniak L. S., Dobbins R. L., Metzger G. J., Sartoni‐D'Ambrosia G., Arbique D., Vongpatanasin W., et al. Myocardial triglycerides and systolic function in humans: in vivo evaluation by localized proton spectroscopy and cardiac imaging. Magn Reson Med 2003; 49: 417–23

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.