ISSN: 1885-5857 Impact factor 2023 7.2
Vol. 59. Num. 1.
Pages 57-66 (January 2006)

Assessment of Progression and Regression of Coronary Atherosclerosis by Intravascular Ultrasound. A New Paradigm Shift?

Evaluación de la progresión y la regresión de la aterosclerosis coronaria mediante ecografía intravascular. ¿Un nuevo cambio de paradigma?

Stephen J NichollsaIlke SipahiaE Murat Tuzcua

Options

Intravascular ultrasound (IVUS) has emerged as a powerful imaging modality for the assessment of the arterial wall and it's response to the accumulation of atherosclerotic plaque. The ability to image the coronary arteries on a serial basis provides a unique opportunity for IVUS to monitor the impact that potential anti-atherosclerotic strategies exert on plaque burden. As a result, studies incorporating serial IVUS imaging as the primary endpoint have demonstrated that therapeutic strategies that modify LDL, HDL and blood pressure in patients with established coronary artery disease can have a profound impact on the progression of atherosclerotic plaque. This heralds a paradigm shift that emphasizes plaque regression as a potential target in the development of preventive strategies.

Keywords

Coronary artery disease
Plaque progression/regression
Coronary atherosclerosis
Intravascular ultrasound
Coronary imaging

INTRODUCTION

Atherosclerotic cardiovascular disease is a growing global public health challenge. Despite the advances in the treatment of clinically manifest coronary artery disease, recurrences are common. Most importantly, initiation of treatment after a clinical event is usually a therapy too late. Accordingly, the search continues to identify strategies that effectively reduce vascular risk. Demonstration of a reduction of clinical events, in response to particular therapies, remains the sine qua non of proof of benefit. However, assessing the impact of potential preventive strategies must be performed on the background of a combination of multiple agents with proven efficacy. High rates of concomitant administration of aspirin, beta-blockers, and statins, in addition to aggressive control of blood pressure and glucose, will result in a substantial lowering of event rates in placebo arms. As a result, it will become necessary to enrol increasingly large numbers of patients, followed for many years to achieve an adequate event rate in order to assess the therapeutic efficacy of experimental agents. The emergence of imaging modalities that characterize the arterial wall provide a unique opportunity to monitor changes in the size and morphology of atherosclerotic plaque, in response to the administration of potential anti-atherosclerotic therapies. Studies using such an imaging modality may provide valuable preliminary information for the design of much larger randomised controlled trials.

EFFECT OF INTERVENTIONS ON ESTABLISHED ATHEROSCLEROSIS

A number of lipid lowering agents reduce the incidence of clinical events in studies of both primary and secondary prevention.1-9 The precise mechanism that confers this benefit remains uncertain. Various groups have attempted to determine whether the reduction in clinical events is due to a reduction in atherosclerotic burden. Demonstrating plaque regression, in response to an intervention, has remained relatively elusive. This has stimulated considerable interest in the concept that the clinical benefit related to the use of these interventions is due largely to resulting changes in plaque composition rather than size. While this may be the case, there is currently no established method of assessing the changes in plaque characteristics in humans.

Coronary angiography is the clinical gold standard for the detection of significant luminal stenoses. It plays a pivotal role in clinical decision making, with regard to the implementation of a range of revascularization strategies. However, its ability to predict ischemic events is limited. Several groups have reported that the majority of myocardial infarctions occur in a vascular territory supplied by a culprit lesion, deemed to be only mildly stenotic at the time of angiography.10-12 However, it has become increasingly recognized that while coronary angiography is a valuable technique for imaging of the luminal border, it's ability to identify the total extent of atherosclerosis is suboptimal.13 This has led to the development of modalities that are more able to accurately image atherosclerotic plaque. Intravascular ultrasound (IVUS) has emerged as a technique that provides a unique opportunity to characterize the response of the arterial wall to serial changes in the accumulation of atherosclerotic plaque.

INTRAVASCULAR ULTRASOUND

IVUS involves the insertion of a catheter, whose tip contains a high frequency ultrasound transducer, within the epicardial coronary arteries. It can be safely performed at the time of diagnostic coronary angiography. IVUS generates high resolution tomographic cross-sectional images of the arterial wall, plaque and lumen. IVUS can be used in the interventional setting to guide decision making, with regard to the management of specific lesions. In addition, continuous imaging during withdrawal of the catheter generates a series of tomographic images that allow for a systematic assessment of atheroma burden. Pullbacks are performed manually or by connecting the catheter to a motorized device, which operates at a constant rate, typically 0.5 mm/s.

The resulting images provide information regarding the extent and composition of atherosclerotic plaque. Using planimetry it is possible to determine the area occupied by the external elastic lamina (EEM) and the lumen. Plaque area can then be calculated as the difference between EEM and lumen areas14 (Figure 1). With the generation of a series of consecutive images, resulting from the pullback of the catheter, the plaque areas from these tomographic images can be summated to estimate the volumetric burden of atheroma in the segment of artery studied. Most groups have typically measured images spaced exactly 1-mm apart for volumetric analysis. IVUS also provides an assessment of the presence, extent and location of calcification. The ability to perform IVUS studies within the same arterial segment at different time points highlights its potential to assess the natural progression of the atherosclerotic process and the impact of various anti-atherosclerotic interventions (Figure 2).

Figure 1. Cross-sectional tomographic image of a coronary artery obtained at IVUS pullback (left panel). Plaque area (yellow) is depicted as the area between the leading edges of the EEM and lumen (right panel).

Figure 2. Representative cross-sectional IVUS images demonstrating plaque progression (top panels) and regression (bottom panels) at matched sites that were studied at baseline (left panels) and follow-up (right panels).

IVUS HIGHLIGHTS THE NATURAL HISTORY OF ATHEROGENESIS

IVUS has provided a number of important insights into the response of the arterial wall to the deposition of atherosclerotic plaque. The ability to image the entire vascular wall allows for an accurate determination of plaque burden. IVUS has confirmed that atherosclerosis is a much more diffuse process than would be suggested by the examination of coronary angiograms.15 In addition, IVUS studies have demonstrated that this chronic process begins early in life. In a study of 262 patients that underwent cardiac transplantation, IVUS examinations were performed of the donor coronary arteries shortly following the transplant procedure. Significant atheroma, defined by a mean maximal plaque thickness greater than 0.5 mm, was found in 1 in 6 apparently healthy hearts from teenage donors. The prevalence of significant atheroma increased dramatically with age.16 These results confirm previous reports of the early appearance of macroscopic atheroma in necropsy studies17,18 and population studies using carotid intimal medial thickness as a surrogate measure of coronary atherosclerosis.19,20

It has become apparent that the vessel wall is not a passive player in the atherosclerotic process. Rather, the wall undergoes substantial changes in size and structure, in response to plaque accumulation. On the basis of necropsy studies, Glagov and colleagues described arterial remodeling as expansion of the internal elastic lamina, in response to the presence of atheroma.21 These changes, subsequently confirmed by IVUS studies,22,23 initially preserve lumen dimensions. It is therefore possible for the arterial wall to harbor a substantial amount of atheroma before it causes any detectable obstruction on angiography. Further studies have demonstrated that the direction of arterial remodeling correlates with the mode of clinical presentation. Culprit lesions in the setting of acute coronary syndromes are more likely to be associated with expansive remodeling, while constrictive remodeling correlates with stable angina pectoris.24

IVUS has identified the presence of multiple ruptured plaques within the coronary arteries in patients with a history of acute coronary syndrome.25,26 Plaque rupture has been found to be present, not only in the culprit vessel, but throughout the coronary arterial tree. This supports the concept that atherosclerosis and it's ischemic complications are part of a systemic and not focal process.27,28 It also confirms that the overwhelming majority of episodes of plaque rupture are not accompanied by clinical ischemia. This suggests that a combination of factors influence whether breakdown of the fibrous cap results in thrombosis and luminal occlusion.29

IVUS has also provided important insights into the natural history of nonatherosclerotic processes, such as transplant vasculopathy. IVUS is a sensitive approach for the detection of early neointimal thickening.30,31 Using serial assessments, IVUS has documented the incidence and subsequent rate of progression of these changes and the impact of interventions.32,33 As a result, serial IVUS imaging has become the clinical gold standard for the routine surveillance of the coronary arteries in heart transplant recipients.34

USE OF IVUS TO ASSESS THE IMPACT OF INTERVENTIONS ON THE NATURAL HISTORY OF PROGRESSION

The ability of IVUS to be performed on a serial basis and to quantify the extent of atherosclerotic plaque or neointimal thickening provides an exciting opportunity to assess the impact of therapeutic interventions. Several reports have emerged that describe the impact of strategies that modify plasma lipids, blood pressure, and the immunological mediators of transplant vasculopathy.

IMPACT OF HDL ELEVATION ON PLAQUE BURDEN

Despite the overwhelming evidence supporting a protective role of high density lipoproteins (HDL) in population35 and animal36-39 studies, data demonstrating a benefit from promoting HDL in humans is limited. It was recently reported that infusing reconstituted HDL particles containing apolipoprotein A-I Milano, a variant of apo A-I, promoted the rapid progression of coronary atheroma in humans.40 Forty seven patients, who underwent coronary angiography and IVUS within 2 weeks of an acute coronary syndrome were randomised to receive weekly intravenous infusions of either placebo or recombinant apo A-I Milano/phospholipid complexes (ETC-216) at a protein dose of 15 mg/kg or 45 mg/kg for 5 weeks. A repeat IVUS study was performed within 2 weeks of the final infusion. The combined treatment group, representing patients that received either low or high dose ETC-216, demonstrated a 4.2% reduction in atheroma volume. The degree of regression was greatest in the most severely diseased 10-mm subsegments. This result extends the findings that carriers of the apo A-I Milano variant appear to be relatively protected from coronary heart disease, despite low HDL levels,41 and that similar infusions in animal models exert a beneficial influence on lesion size and composition.42-44 Further, it extends the findings, that the combination of niacin and a statin, with resulting HDL elevation, promoted atherosclerotic regression in angiographic studies45 and halted progression of carotid intimal medial thickness.46 This small study is an important proof of concept that interventions that promote HDL have the potential to have a dramatic impact on coronary atheroma. It calls for the design of large scale studies to further investigate the potential impact that promoting HDL has on coronary atheroma and the incidence of clinical events.

IMPACT OF LIPID LOWERING ON PLAQUE BURDEN

It is well established that LDL lowering with statins reduces clinical events.1-6 However, it remains uncertain whether a LDL threshold exists, below which incremental clinical benefit is limited. This has stimulated considerable debate in the development of lipid lowering guidelines. Serial IVUS studies have documented the response of coronary atheroma to intensive lipid lowering strategies. Early reports of atherosclerotic regression were reported, using both quantitative coronary angiography and IVUS, following LDL apheresis in patients with familial hypercholesterolemia that was refractory to conventional medical therapy.47,48 Numerous angiographic studies have demonstrated that lipid lowering with statins slowed the rate of progression of atheroma in comparison with placebo.49-53 Recent IVUS studies have directly addressed the issue whether the degree of LDL lowering with statin therapy influences coronary atheroma.

The Reversal of Atherosclerosis with Aggressive Lipid Lowering (REVERSAL) study54 randomised 502 patients with coronary artery disease and an LDL-C level between 125 mg/dL and 210 mg/dL to receive either a moderate lipid lowering strategy with pravastatin 40 mg daily or an intensive lipid lowering strategy with atorvastatin 80 mg daily for 18 months. Mean LDL-C was reduced to 79 mg/dL and 110 mg/dL with atorvastatin and pravastatin respectively. In addition the strategies differed markedly in their ability to lower CRP. CRP decreased by 36.4% and 5.2% in atorvastatin and pravastatin treated patients respectively. Serial IVUS demonstrated that atheroma volume increased by 2.7% with pravastatin, indicating net progression. In contrast, treatment with atorvastatin resulted in no significant change of atheroma volume compared with baseline. Further, it was demonstrated that both strategies promoted regression in the 10-mm segments that were determined to harbor the greatest plaque burden at baseline. Overall, this result suggested that use of an intensive lipid lowering strategy has the potential to alter the natural history of progression of coronary atheroma. This corresponds with the recently reported Treating to New Targets (TNT) study which demonstrated that incremental LDL lowering with high dose compared with low dose atorvastatin was associated with a 22% reduction in the combination of clinical end points.

The beneficial effect of statin therapy on plaque burden was recently supported by the finding that in a small cohort of 40 males with hypercholesterolemia and ischemic heart disease that 3 months of a lipid-lowering diet followed by simvastatin 40 mg daily resulted in a 42.6% reduction in LDL and associated 6.3% reduction in plaque volume, consistent with plaque regression.55

While there appeared to be a continuous relationship between the change in LDL and change in plaque progression with both strategies in REVERSAL, at any level of LDL-C the regression line was lower for atorvastatin. It appeared that the lower progression rate with atorvastatin was equivalent to an additional 20% LDL-C lowering with pravastatin and suggested strongly that the beneficial properties of atorvastatin may extend beyond lipid lowering. Subsequent analysis demonstrated a correlation between the degree of CRP lowering and rate of progression of plaque, with subjects receiving the largest reduction in CRP demonstrating evidence of atheroma regression.56 The incremental benefit of intensive lipid lowering and its correlation with the degree of CRP reduction supported the recently reported benefit of high dose atorvastatin compared with pravastatin therapy on clinical event rates in patients with an acute coronary syndrome in the Pravastatin or Atorvastatin Evaluation and Infection Therapy (PROVE-IT) study.57,58

LDL lowering was demonstrated to have a beneficial impact on plaque burden when commenced in the setting of a recent acute coronary syndrome. In the ESTABLISH study59 70 patients with ST elevation myocardial infarction, who proceeded to percutaneous intervention of the culprit lesion were randomized to receive atorvastatin 20 mg daily or conventional treatment with a lipid lowering diet for 6 months. A cholesterol absorption inhibitor could be initiated in the conventional arm if their LDL remained greater than 150 mg/dL. Forty eight of the subjects underwent serial IVUS examinations either proximal or distal to the intervention site at baseline and follow-up. Atorvastatin treatment was associated with a 41.7% reduction in LDL levels and promoted plaque regression, as evidenced by a 13.1% reduction in plaque volume. In contrast, LDL did not change in the conventional treatment group, which demonstrated an 8.7% increase in plaque volume. The percentage change in plaque volume correlated with both the LDL level at follow-up and the percent reduction in LDL. The result of this short term study is parallel to the finding in the Myocardial Ischemia Reduction with Aggressive Cholesterol Lowering (MIRACL) study60 which enrolled patients with acute coronary syndromes. In this study high dose atorvastatin was associated with a significant 16% reduction in clinical events within 16 weeks of an acute coronary syndrome.

IMPACT OF BLOOD PRESSURE LOWERING ON PLAQUE BURDEN

Clinical IVUS studies have also assessed the impact of targeting risk factors beyond plasma lipoproteins. There is currently no consensus view with regard to optimal blood pressure management in normotensive patients with established coronary artery disease. The recently reported Comparison of Amlodipine versus Enalapril to Limit Occurrences of Thrombosis (CAMELOT) study61 was designed to assess the influence that blood pressure reduction had on clinical events and plaque burden in normotensive subjects. A total of 1991 patients with coronary artery disease and a diastolic blood pressure less than 100 mm Hg, with or without treatment, were randomized to receive either amlodipine 10 mg daily, enalapril 20 mg daily, or placebo for 24 months. Treatment with amlodipine resulted in a 31% reduction in the composite incidence of clinical events compared with placebo. This benefit resulted largely from a 27% reduction in coronary revascularization and 42% reduction in hospitalization for angina. Two hundred and seventy four of these subjects participated in an IVUS substudy, which demonstrated plaque progression with placebo and no change in atherosclerotic burden with administration of amlodipine. When patients with blood pressure at baseline greater than the mean were considered (a prespecified analysis), the difference between the amlodipine and placebo groups became statistically significant. It was also noted that the effects of the treatment arms on clinical events corresponded with their effect on plaque burden.

USE OF SERIAL IVUS TO MONITOR PROGRESSION OF OTHER VASCULAR PATHOLOGIES

IVUS has provided a number of insights into the in vivo response of the arterial wall to angioplasty. In particular, it has established that restenosis following percutaneous non-stent interventions results from a combination of arterial recoil, remodeling and the development of neointimal hyperplasia.62 In addition to assessing the impact of bare metal and drug eluting stents,63,64 serial IVUS examinations have been employed to investigate the clinical utility of a number of experimental strategies that potentially inhibit neointima formation. In the multivitamins and probucol study group, 317 patients were randomised to receive the antioxidant probucol, multivitamins, the combination or placebo 30 days prior to coronary angioplasty and for a period of 6 months after. Serial IVUS performed in a subset of 94 patients confirmed that restenosis resulted primarily from inadequate remodeling in the setting of neointima formation, which was prevented by treatment with probucol.65 It was demonstrated that a cross-sectional narrowing less than 67% on IVUS performed immediately following angioplasty predicted a low rate of restenosis in patients treated with probucol at 6 months.66 Further, the experimental anti-inflammatory and antioxidant agent AGI-1067 was recently demonstrated to prevent restenosis following angioplasty, in the absence of QTc prolongation seen with probucol.67

As IVUS has emerged as the gold standard for the clinical surveillance of the development and progression of coronary artery vasculopathy in heart transplant recipients it provides a unique opportunity to assess the efficacy of strategies designed to inhibit this process. The onset of transplant vasculopathy portends a poor clinical outcome and is the leading indication for repeat cardiac transplantation.68 Recent reports have found that the progression of coronary vasculopathy predicts the long-term incidence of clinical events.69,70 Unfortunately there are few therapeutic options that have been demonstrated to have any impact on this process.71 It has become increasingly apparent that a cascade of immunological events promotes the formation of neointimal hyperplasia.72 As a result, immunological mediators have received increasing attention as potential targets.

Everolimus is a novel proliferation inhibitor and immunosuppressant.73 Serial IVUS was used to demonstrate the ability of everolimus to prevent the development of vasculopathy.74 six hundred and thrity four primary heart transplant recipients were randomised to receive 1.5 mg of everolimus, 3 mg of everolimus or 1-3 mg of azathioprine in combination with standard medical therapy. Coronary artery IVUS studies were performed within 6 weeks of transplantation and at 12 month follow-up. Compared with the azathioprine group, the incidence of vasculopathy was reduced by 33% and 42% with low and high dose everolimus respectively. Further, everolimus inhibited the increase in average maximal intimal thickness by 60%-70%. This benefit was associated with a 22%-42% reduction in the composite clinical endpoint of death, graft loss or retransplantation, loss to follow-up, acute rejection grade 3A or rejection with hemodynamic compromise. This result reflects an important ability of serial in vivo IVUS imaging to correlate the impact of therapeutic interventions on the incidence of both arterial vasculopathy and clinical events.

Although it is not a proof, all of the data from trials using IVUS and clinical information support the concept that IVUS can be used as a reliable representative of clinical outcome.

NEED TO DEVELOP STRATEGIES TO MONITOR PLAQUE ACTIVITY

It has become increasingly recognized that the composition of atherosclerotic plaque, in addition to its total burden, is an important determinant of its translation to clinical ischemia. Several groups have reported that myocardial infarction occurs predominantly in the vascular territory supplied by a culprit lesion, that is only mild to moderately stenotic on coronary angiography.10-12 Pathologic studies have demonstrated that macrophage rich plaques are more likely to undergo erosion or rupture of the fibrous cap, the typical precipitant of ischemic events.28,75 In support, it has been reported that systemic levels of CRP predict outcome in both healthy subjects76 and in the setting of acute coronary syndromes.77 Immense interest has therefore focused on the development of imaging modalities that monitor the activity of atherosclerotic plaque. A number of techniques have been utilized in the research setting to characterize plaque composition. While IVUS provides an excellent assessment of atherosclerotic burden, its ability to characterize plaque components is relatively limited to a broad distinction between echolucent (lipidic), echodense (fibrous), and calcific components. Although the relationship between a plaque's histological and ultrasonic characteristics are inconsistent.14 Recent developments of IVUS that have focussed on the analysis of radiofrequency backscatter allow for the characterization of plaque components, with good histopathological correlation.78 Spectral analysis of this backscattered data has been demonstrated ex vivo to accurately identify the size and composition of different components of atherosclerotic plaque in human coronary arteries. As a result, this modality has become available for use in the clinical setting to identify lipid rich, and therefore potentially vulnerable, lesions. Optical coherence tomography (OCT), which measures reflected light, rather than ultrasound, has been demonstrated to image atheroma macrophage activity with high resolution.79,80 In addition, the ability of catheters to quantify plaque temperature81,82 and compressibility81,83 has the potential to identify inflamed and lipidic regions. The major limitation of each of these strategies is their need for invasive cardiac catheterisation. The ideal approach would involve a non-invasive assessment of the coronary arteries. Magnetic resonance has been reported to characterize plaque components.84,85 However, an MRI technique for clinical use is not yet available. Computerized tomography (CT), in contrast, is limited at this point in time to the assessment of the extent of coronary calcification, detection of luminal stenoses and characterization of arterial remodelling in response to plaque.86-88 There are efforts to assess the arterial wall characteristics by using Hounsfeld units. The combination of CT with positron emission tomographic (PET) imaging, which has been demonstrated to accurately assess plaque macrophage activity,89 is promising. The further developments of these imaging techniques will provide an ideal opportunity to assess the serial response of plaque composition, and therefore vulnerability, in response to a number of therapeutic interventions in vivo.

CONCLUSION

The development of IVUS has enhanced our understanding of the factors that influence the progressive accumulation of atherosclerotic plaque. The ability to perform IVUS on a serial basis provides a unique opportunity to characterize changes in atheroma in response to various pharmacologic strategies. It has become apparent that aggressive modification of traditional cardiovascular risk factors can have a profound impact on the natural history of plaque progression. This benefit appears to complement the ability of these interventions to prevent clinical events. The ability to identify strategies that promote the regression of atheroma, rather than just halting progression or reducing plaque vulnerability, represents a major paradigm shift in cardiovascular protection. As a result, it will become increasingly important to incorporate serial assessment of plaque burden as a major endpoint in the design of future clinical studies that focus on the development of emerging strategies to further reduce cardiovascular risk.


Correspondence: Dr E. Murat Tuzcu.
The Cleveland Clinic Foundation. Department of Cardiovascular Medicine/F15
9500 Euclid Avenue, Cleveland OH 44195, USA.
E-mail: tuzcue@ccf.org

Bibliography
[1]
Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S)..
Lancet, (1994), 344 pp. 1383-9
[2]
Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels..
The Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group..
N Engl J Med, (1998), 339 pp. 1349-57
[3]
MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20 536 high-risk individuals: a randomised placebo-controlled trial..
[4]
Downs JR, Clearfield M, Weis S, Whitney E, Shapiro DR, Beere PA, et al..
Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFcaps/Texcaps. AirForce/Texas Coronary Atherosclerosis Prevention Study..
JAMA, (1998), 279 pp. 1615-22
[5]
Sacks FM, Pfefer MA, Moye LA, Rouleau JL, Rutherford JD, Cole TG, et al..
The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events Trial investigators..
N Engl J Med, (1996), 335 pp. 1001-9
[6]
Shepherd J, Cobbe SM, Ford I, Isles CG, Lorimer AR, MacFarlane PW, et al..
Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. West of Scotland Coronary Prevention Study Group..
N Engl J Med, (1995), 333 pp. 1301-7
[7]
Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Elam MB, et al..
Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group..
N Engl J Med, (1999), 341 pp. 410-8
[8]
Frick MH, Elo O, Haapa K, Heinonen OP, Heinsalmi P, Helo P, et al..
Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle aged men with dyslipidemia. Safety of treatment, changes in risk factors and incidence of coronary heart disease..
N Engl J Med, (1987), 317 pp. 1237-45
[9]
Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G..
Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators..
N Engl J Med, (2000), 342 pp. 145-53
[10]
Ambrose JA, Tannenbaum MA, Alexopoulos D, Hjemdahl-Monsen CE, Leavy J, Weiss M, et al..
Angiographic progression of coronary artery disease and the development of myocardial infarction..
J Am Coll Cardiol, (1988), 12 pp. 56-62
[11]
Giroud D, Li JM, Urban P, Meier B, Rutishauer W..
Relation of the site of acute myocardial infarction to the most severe coronary arterial stenosis at prior angiography..
Am J Cardiol, (1992), 69 pp. 729-32
[12]
Little WC, Constantinescu M, Applegate RJ, Kutcher MA, Burrows MT, Kahl FR, 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 pp. 1157-66
[13]
Topol EJ, Nissen SE..
Our preoccupation with coronary luminology. The dissociation between clinical and angiographic findings in ischemic heart disease..
Circulation, (1995), 92 pp. 2333-42
[14]
Mintz GS, Nissen SE, Anderson WD, Bailey SR, Erbel R, Fitzgerald PJ, et al..
American College of Cardiology Clinical Expert Consensus Document on Standards for Acquisition, Measurement and Reporting of Intravascular Ultrasound Studies (IVUS). A report of the American College of Cardiology Task Force on Clinical Expert Consensus Documents..
J Am Coll Cardiol, (2001), 37 pp. 1478-92
[15]
Atherosclerosis in angiographically
[16]
Tuzcu EM, Kapadia SR, Tutar E, Ziada KM, Hobbs RE, McCarthy PM, et al..
High prevalence of coronary atherosclerosis in asymptomatic teenagers and young adults: evidence from intravascular ultrasound..
Circulation, (2001), 103 pp. 2705-10
[17]
Berenson GS, Wattigney WA, Tracy RE, Newman WP 3r.d, Srinivasan SR, Webber LS, et al..
Atherosclerosis of the aorta and coronary arteries and cardiovascular risk factors in persons aged 6 to 30 years and studied at necropsy (The Bogalusa Heart Study)..
Am J Cardiol, (1992), 70 pp. 851-8
[18]
Relationship of atherosclerosis in young men to serum lipoprotein cholesterol concentrations and smoking..
A preliminary report from the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group..
JAMA, (1990), 264 pp. 3018-24
[19]
Knoflach M, Kiechl S, Kind M, Said M, Sief R, Gisinger M, et al..
Cardiovascular risk factors and atherosclerosis in young males: ARMY study (Atherosclerosis Risk-Factors in Male Youngsters)..
[20]
Urbina EM, Srinivasan SR, Tang R, Bond MG, Kieltyka L, Berenson GS..
Impact of multiple coronary risk factors on the intima-media thickness of different segments of carotid artery in healthy young adults (The Bogalusa Heart Study)..
Am J Cardiol, (2002), 90 pp. 953-8
[21]
Glagov S, Weisenberg E, Zarins CK, Stankunavicius R, Kolettis GJ..
Compensatory enlargement of human atherosclerotic coronary arteries..
N Engl J Med, (1987), 316 pp. 1371-5
[22]
Pasterkamp G, Wensing PJ, Post MJ, Hillen B, Mali WP, Borst C..
Paradoxical arterial wall shrinkage may contribute to luminal narrowing of human atherosclerotic femoral arteries..
Circulation, (1995), 91 pp. 1444-9
[23]
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 pp. 2570-7
[24]
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 pp. 598-603
[25]
Rioufol G, Finet G, Ginon I, Andre-Fouet X, Rossi R, Vialle E, et al..
Multiple atherosclerotic plaque rupture in acute coronary syndrome: a three-vessel intravascular ultrasound study..
Circulation, (2002), 106 pp. 804-8
[26]
Schoenhagen P, Stone GW, Nissen SE, Grines CL, Griffin J, Clemson BS, et al..
Coronary plaque morphology and frequency of ulceration distant from culprit lesions in patients with unstable and stable presentation..
Arterioscler Thromb Vasc Biol, (2003), 23 pp. 1895-900
[27]
Lutgens E, van Suylen RJ, Faber BC, Gijbels MJ, Eurlings PM, Bijnens AP, et al..
Atherosclerotic plaque rupture:local or systemic process? Arterioscler Thromb Vasc Biol, (2003), 23 pp. 2123-30
[28]
Ross R..
Atherosclerosis--an inflammatory disease..
N Engl J Med, (1999), 340 pp. 115-26
[29]
Naghavi M, Libby P, Falk E, Casscells SW, 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 pp. 1664-72
[30]
Pinto FJ, Chenzbraun A, Botas J, Valantine HA, St Goar FG, Alderman EL, et al..
Feasibility of serial intracoronary ultrasound imaging for assessment of progression of intimal proliferation in cardiac transplant recipients..
Circulation, (1994), 90 pp. 2348-55
[31]
Jimenez J, Kapadia SR, Yamani MH, Platt L, Hobbs RE, Rincon G, et al..
Cellular rejection and rate of progression of transplant vasculopathy: a 3-year serial intravascular ultrasound study..
J Heart Lung Transplant, (2001), 20 pp. 393-8
[32]
Tuzcu EM, De Franco AC, Goormastic M, Hobbs RE, Rincon G, Bott-Silverman C, et al..
Dichotomous pattern of coronary atherosclerosis 1 to 9 years after transplantation: insights from systematic intravascular ultrasound imaging..
J Am Coll Cardiol, (1996), 27 pp. 839-46
[33]
Yeung AC, Davis SF, Hauptman PJ, Kobashigawa JA, Miller LW, Valantine HA, et al..
Incidence and progression of transplant coronary artery disease over 1 year: results of a multicenter trial with use of intravascular ultrasound. Multicenter Intravascular Ultrasound Transplant Study Group..
J Heart Lung Transplant, (1995), 14 pp. S215-220
[34]
Kapadia SR, Nissen SE, Tuzcu EM..
Impact of intravascular ultrasound in understanding transplant coronary artery disease..
Curr Opin Cardiol, (1999), 14 pp. 140-50
[35]
Gordon T, Castelli WP, Hjortland MC, Kannel WB, Dawber TR..
High density lipoprotein as a protective factor against coronary heart disease. The Framingham Study..
Am J Med, (1977), 62 pp. 707-14
[36]
Badimon JJ, Badimon L, Galvez A, Dische R, Fuster V..
High density lipoprotein plasma fractions inhibit aortic fatty streaks in cholesterol-fed rabbits..
Laboratory Investigation, (1989), 60 pp. 455-61
[37]
Badimon JJ, Badimon L, Fuster V..
Regression of atherosclerotic lesions by high density lipoprotein plasma fraction in the cholesterol-fed rabbit..
J Clin Invest, (1990), 85 pp. 1234-41
[38]
Rong JX, Li J, Reis ED, Choudhury RP, Dansky HM, Elmalem VI, et al..
Elevating high-density lipoprotein cholesterol in apolipoprotein E-deficient mice remodels advanced atherosclerotic lesions by decreasing macrophage and increasing smooth muscle cell content..
Circulation, (2001), 104 pp. 2447-52
[39]
Plump AS, Scott CJ, Breslow JL..
Human apolipoprotein A-I gene expression increases high density lipoprotein and suppresses atherosclerosis in the apolipoprotein E-deficient mouse..
Proc Nat Acad Sci (USA), (1994), 91 pp. 9607-11
[40]
Nissen SE, Tsunoda T, Tuzcu EM, Schoenhagen P, Cooper CJ, Yasin M, et al..
Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial..
JAMA, (2003), 290 pp. 2292-300
[41]
Franceschini G, Sirtori CR, Capurso A 2n.d, Weisgraber KH, Mahley RW..
A-IMilano apoprotein. Decreased high density lipoprotein cholesterol levels with significant lipoprotein modifications and without clinical atherosclerosis in an Italian family..
J Clin Invest, (1980), 66 pp. 892-900
[42]
Chiesa G, Monteggia E, Marchesi M, Lorenzon P, Laucello M, Lorusso V, et al..
Recombinant apolipoprotein A-IMilano infusion into rabbit carotid artery rapidly removes lipid from fatty streaks..
Circ Res, (2002), 90 pp. 974-80
[43]
Shah PK, Nilsson J, Kaul S, Fishbein MC, Ageland H, Hamsten A, et al..
Effects of recombinant apolipoprotein A-I(Milano) on aortic atherosclerosis in apolipoprotein E-deficient mice..
Circulation, (1998), 97 pp. 780-5
[44]
Shah PK, Yano J, Reyes O, Chyu KY, Kaul S, Bisgaier CL, et al..
High-dose recombinant apolipoprotein A-IMilano mobilizes tissue cholesterol and rapidly reduces plaque lipid and macrophage content in apolipoprotein E-deficient mice..
Circulation, (2001), 103 pp. 3047-50
[45]
Brown BG, Zhao X-Q, Chait A, Fisher LD, Cheung MC, Morse JS, et al..
Simvastatin and niacin, antioxidant vitamins, or the combination for the prevention of coronary disease..
N Engl J Med, (2001), 345 pp. 1583-92
[46]
Taylor AJ, Sullenberger LE, Lee HJ, Lee JK, Grace KA..
Arterial Biology for the Investigation of the Treatment Effects of Reducing Cholesterol (ARBITER) 2: a double-blind, placebo-controlled study of extended-release niacin on atherosclerosis progression in secondary prevention patients treated with statins..
[47]
Matsuzaki M, Hiramori K, Imaizumi T, Kitabatake A, Hishida H, Nomura M, et al..
Intravascular ultrasound evaluation of coronary plaque regression by low density lipoprotein-apheresis in familial hypercholesterolemia: the Low Density Lipoprotein-Apheresis Coronary Morphology and Reserve Trial (LACMART)..
J Am Coll Cardiol, (2002), 40 pp. 220-7
[48]
Koga N, Iwata Y, Yamamoto A..
Angiographic and pathological studies on regression of coronary atherosclerosis of FH patients who received LDL-apheresis treatment..
Artif Organs, (1992), 16 pp. 171-6
[49]
Pitt B, Mancini GB, Ellis SG, Rosman HS, Park JS, McGovern ME..
Pravastatin limitation of atherosclerosis in the coronary arteries (PLAC I): reduction in atherosclerosis progression and clinical events. PLAC I investigation..
J Am Coll Cardiol, (1995), 26 pp. 1133-9
[50]
Jukema JW, Bruschke AV, van Boven AJ, Reiber JH, Bal ET, Zwinderman AH, et al..
Effects of lipid lowering by pravastatin on progression and regression of coronary artery disease in symptomatic men with normal to moderately elevated serum cholesterol levels. The Regression Growth Evaluation Statin Study (REGRESS)..
Circulation, (1995), 91 pp. 2528-40
[51]
Blankenhorn DH, Azen SP, Kramsch DM, Mack WJ, Cashin-Hemphill L, Hodis HN, et al..
Coronary angiographic changes with lovastatin therapy. The Monitored Atherosclerosis Regression Study (MARS). The MARS Research Group..
Ann Intern Med, (1993), 119 pp. 969-76
[52]
Arntz HR, Agrawal R, Wunderlich W, Schnitzer L, Stern R, Fischer F, et al..
Beneficial effects of pravastatin (+/-colestyramine/niacin) initiated immediately after a coronary event (the randomized Lipid-Coronary Artery Disease [L-CAD] Study)..
Am J Cardiol, (2000), 86 pp. 1293-8
[53]
Effect of simvastatin on coronary atheroma: the Multicentre Anti-Atheroma Study (MAAS)..
Lancet, (1994), 344 pp. 633-8
[54]
Nissen SE, Tuzcu EM, Schoenhagen P, Brown BG, Ganz P, Vogel RA, et al; REVERSAL Investigators..
Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial..
JAMA, (2004), 291 pp. 1071-80
[55]
Jensen LO, Thayssen P, Pedersen KE, Stender S, Haghfelt T..
Regression of coronary atherosclerosis by simvastatin: a serial intravascular ultrasound study..
[56]
Nissen SE, Tuzcu EM, Schoenhagen P, Crowe T, Sasiela WJ, Tsai J, et al; Reversal of Atherosclerosis with Aggressive Lipid Lowering (REVERSAL) Investigators..
Statin therapy, LDL cholesterol, C-reactive protein, and coronary artery disease..
N Engl J Med, (2005), 352 pp. 29-38
[57]
Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL, Belder R, et al; Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 Investigators..
Intensive versus moderate lipid lowering with statins after acute coronary syndromes..
N Engl J Med, (2004), 350 pp. 1495-504
[58]
Ridker PM, Cannon CP, Morrow D, et al..
C-reactive protein levels and outcomes after statin therapy. N Engl J Me, (2005), 352 pp. 20-8
[59]
Okazaki S, Yokoyama T, Miyauchi K, Rifai N, Rose LM, McCabe CH, et al; Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 (PROVE IT-TIMI 22) Investigators..
Early statin treatment in patients with acute coronary syndrome: demonstration of the beneficial effect on atherosclerotic lesions by serial volumetric intravascular ultrasound analysis during half a year after coronary event: the ESTABLISH Study..
[60]
Schwartz GG, Olsson AG, Ezekowitz MD, Ganz P, Oliver MF, Waters D, et al; Myocardial Ischemia Reduction with Aggressive Cholesterol Lowering (MIRACL) Study Investigators..
Effects of atorvastatin on early recurrent ischemic events in acute coronary syndromes: the MIRACL study: a randomized controlled trial..
JAMA, (2001), 285 pp. 1711-8
[61]
Nissen SE, Tuzcu EM, Libby P, Thompson PD, Ghali M, Garza D, et al..
Effect of antihypertensive agents on cardiovascular events in patients with coronary disease and normal blood pressure: the CAMELOT study: a randomized controlled trial..
JAMA, (2004), 292 pp. 2217-25
[62]
Mintz GS, Kent KM, Pichard AD, Popma JJ, Satler LF, Leon MB..
Intravascular ultrasound insights into mechanisms of stenosis formation and restenosis..
Cardiol Clin, (1997), 15 pp. 17-29
[63]
Fuessl RT, Hoepp HW, Sechtem U..
Intravascular ultrasonography in the evaluation of results of coronary angioplasty and stenting..
Curr Opin Cardiol, (1999), 14 pp. 471-9
[64]
Serruys PW, Degertekin M, Tanabe K, Russell ME, Guagliumi G, Webb J, et al..
Vascular responses at proximal and distal edges of paclitaxel-eluting stents: serial intravascular ultrasound analysis from the TAXUS II trial..
[65]
Cote G, Tardif JC, Lesperance J, Lambert J, Bourassa M, Bonan R, et al..
Effects of probucol on vascular remodeling after coronary angioplasty. Multivitamins and Protocol Study Group..
Circulation, (1999), 99 pp. 30-5
[66]
Tardif JC, Cote G, Lesperance J, Gosselin G, Joyal M, de Guise P, et al..
Impact of residual plaque burden after balloon angioplasty in the MultiVitamins and Probucol (MVP) trial..
Can J Cardiol, (2001), 17 pp. 49-55
[67]
Tardif JC, Gregoire J, Schwartz L, Title L, Laramee L, Reeves F, et al; Canadian Antioxidant Restenosis Trial (CART-1) Investigators..
Effects of AGI-1067 and probucol after percutaneous coronary interventions..
Circulation, (2003), 107 pp. 552-8
[68]
Worldwide thoracic organ transplantation: a report from the UNOS/ISHLT International Registry for Thoracic Organ Transplantation. Clin Transpl. 1999:35-49.
[69]
Kobashigawa JA, Tobis JM, Starling RC, Tuzcu EM, Smith AL, Valantine HA, et al..
Intravascular ultrasound validation study among heart transplant recipients. Outcomes after 5 years..
J Am Coll Cardiol, (2005), 45 pp. 1532-7
[70]
Tuzcu EM, Kapadia SR, Sachar R, Ziada KM, Crowe TD, Feng J, et al..
Intravascular ultrasound evidence of angiographically silent progression in coronary atherosclerosis predicts long-term morbidity and mortality after cardiac transplantation..
J Am Coll Cardiol, (2005), 45 pp. 1538-42
[71]
Kobashigawa JA, Katznelson S, Laks H, Johnson JA, Yeatman L, Wang XM, et al..
Effect of pravastatin on outcomes after cardiac transplantation..
N Engl J Med, (1995), 333 pp. 621-7
[72]
Pinney SP, Mancini D..
Cardiac allograft vasculopathy: advances in understanding its pathophysiology, prevention, and treatment..
Curr Opin Cardiol, (2004), 19 pp. 170-6
[73]
Schuler W, Sedrani R, Cottens S, Haberlin B, Schulz M, Schuurman HJ, et al..
SDZ RAD, a new rapamycin derivative: pharmacological properties in vitro and in vivo..
Transplantation, (1997), 64 pp. 36-42
[74]
Eisen HJ, Tuzcu EM, Dorent R, Kobashigawa J, Mancini D, Valantine-von Kaeppler HA, et al..
Everolimus for the prevention of allograft rejection and vasculopathy in cardiac-transplant recipients..
N Engl J Med, (2003), 349 pp. 847-58
[75]
Falk E, Shah PK, Fuster V..
Coronary plaque disruption..
Circulation, (1995), 92 pp. 657-71
[76]
Ridker PM, Rifai N, Rose L, Buring JE, Cook NR..
Comparison of C-reactive protein and low-density lipoprotein cholesterol levels in the prediction of first cardiovascular events..
N Engl J Med, (2002), 347 pp. 1557-65
[77]
Lindahl B, Toss H, Siegbahn A, Venge P, Wallentin L..
Markers of myocardial damage and inflammation in relation to long-term mortality in unstable coronary artery disease. FRISC Study Group. Fragmin during Instability in Coronary Artery Disease..
N Engl J Med, (2000), 343 pp. 1139-47
[78]
Nair A, Kuban BD, Tuzcu EM, Schoenhagen P, Nissen SE, Vince DG..
Coronary plaque classification with intravascular ultrasound radiofrequency data analysis..
Circulation, (2002), 106 pp. 2200-6
[79]
MacNeill BD, Jang IK, Bouma BE, Iftimia N, Takano M, Yabushita H, et al..
Focal and multi-focal plaque macrophage distributions in patients with acute and stable presentations of coronary artery disease..
J Am Coll Cardiol, (2004), 44 pp. 972-9
[80]
Tearney GJ, Yabushita H, Houser SL, Aretz HT, Jang IK, Schlendorf KH, et al..
Quantification of macrophage content in atherosclerotic plaques by optical coherence tomography..
Circulation, (2003), 107 pp. 113-9
[81]
MacNeill BD, Lowe HC, Takano M, Fuster V, Jang IK..
Intravascular modalities for detection of vulnerable plaque: current status..
Arterioscler Thromb Vasc Biol, (2003), 23 pp. 1333-42
[82]
Verheye S, de Meyer GR, Van Langenhove G, Knaapen MW, Kockx MM..
In vivo temperature heterogeneity of atherosclerotic plaques is determined by plaque composition..
Circulation, (2002), 105 pp. 1596-601
[83]
Schaar JA, de Korte CL, Mastik F, Strijder C, Pasterkamp G, Boersma E, et al..
Characterizing vulnerable plaque features with intravascular elastography..
Circulation, (2003), 108 pp. 2636-41
[84]
Saam T, Ferguson MS, Yarnykh VL, Takaya N, Xu D, Polissar NL, et al..
Quantitative evaluation of carotid plaque composition by in vivo MRI..
Arterioscler Thromb Vasc Biol, (2005), 25 pp. 234-9
[85]
Choudhury RP, Fuster V, Badimon JJ, Fisher EA, Fayad ZA..
MRI and characterization of atherosclerotic plaque: emerging applications and molecular imaging..
Arterioscler Thromb Vasc Biol, (2002), 22 pp. 1065-74
[86]
Achenbach S, Daniel WG..
Imaging of coronary atherosclerosis using computed tomography: current status and future directions..
Curr Atheroscler Rep, (2004), 6 pp. 213-8
[87]
Schoenhagen P, Halliburton SS, Stillman AE, Kuzmiak SA, Nissen SE, Tuzcu EM, et al..
Noninvasive imaging of coronary arteries: current and future role of multi-detector row CT..
Radiology, (2004), 232 pp. 7-17
[88]
Schoepf UJ, Becker CR, Ohnesorge BM, Yucel EK..
CT of coronary artery disease..
Radiology, (2004), 232 pp. 18-37
[89]
Rudd JH, Warburton EA, Fryer TD, Jones HA, Clark JC, Antoun N, et al..
Imaging atherosclerotic plaque inflammation with [18F]-fluorodeoxyglucose positron emission tomography..
Circulation, (2002), 105 pp. 2708-11
Are you a healthcare professional authorized to prescribe or dispense medications?