Abstract

Intravascular imaging has been often used over the recent years to examine the efficacy of emerging therapies targeting plaque evolution. Serial intravascular ultrasound, optical coherence tomography, or near-infrared spectroscopy-intravascular ultrasound studies have allowed us to evaluate the effects of different therapies on plaque burden and morphology, providing unique mechanistic insights about the mode of action of these treatments. Plaque burden reduction, a decrease in necrotic core component or macrophage accumulation—which has been associated with inflammation—and an increase in fibrous cap thickness over fibroatheromas have been used as surrogate endpoints to assess the value of several drugs in inhibiting plaque evolution and improving clinical outcomes. However, some reports have demonstrated weak associations between the effects of novel treatments on coronary atheroma and composition and their prognostic implications. This review examines the value of invasive imaging in assessing pharmacotherapies targeting atherosclerosis. It summarizes the findings of serial intravascular imaging studies assessing the effects of different drugs on atheroma burden and morphology and compares them with the results of large-scale trials evaluating their impact on clinical outcome. Furthermore, it highlights the limited efficacy of established intravascular imaging surrogate endpoints in predicting the prognostic value of these pharmacotherapies and introduces alternative imaging endpoints based on multimodality/hybrid intravascular imaging that may enable more accurate assessment of the athero-protective and prognostic effects of emerging therapies.

1. Introduction

Coronary artery disease (CAD) is the most common cause of death in the developed world, is associated with increased morbidity, and has devastating economic consequences in Europe and in the USA. Therefore, an effort has been made to understand the pathophysiological mechanisms that regulate plaque progression and to develop effective therapies that will inhibit atherosclerosis evolution, improve quality of life, and prolong life expectancy in patients who suffer from CAD.1

Intravascular imaging, which enables detailed assessment of plaque pathology albeit with some limitations (Figure 1), has been used to examine the effect of these therapies on plaque burden (PB) and it has provided unique insights into the effects of these drugs on plaque morphology.2–4 In contrast to outcome trials that require large numbers of patients to prove prognostic benefit, intravascular imaging studies use imaging-based surrogate endpoints such as changes in per cent atheroma volume (PAV) or composition to evaluate their efficacy on plaque progression using a smaller number of patients and at a lower cost.5 Imaging-based studies have been performed to investigate the mechanisms of action of emerging therapies and provide proof of their athero-protective effect that would justify the conduction of large outcome studies, or to complement ongoing outcome studies with mechanistical insights on in vivo modes of action. Nevertheless, some reports demonstrated only a weak association between changes in PB or its characteristics and clinical outcomes, questioning the value of imaging-based endpoints in assessing the efficacy of novel treatments (see Supplementary material online, Figure S1).

Advantages and limitations of the clinically available intravascular imaging modalities for assessing vessel wall pathology. The table at the bottom of the figure provides a quantification of the efficacy of each modality in detecting different tissue types using histology as the reference standard. The symbol (−) indicates that the modality is unable to detect the specific characteristic. Conversely, the symbols (+), (++), and (+++) indicate weak, moderate, and excellent ability of the modality to detect a plaque characteristic, respectively. White arrows indicate neo-vessels, cholesterol crystal, erupted calcified nodule, thin fibrous cap and macrophages infiltration in cross-sectional OCT images. Asterisks indicate calcific tissue on IVUS (upper panel) and OCT (lower panel) and evidence of intraluminal thrombus on OCT. A red arc defines the lipid necrotic core in NIRS-IVUS image. The superimposed light blue colour indicates plaque burden and positive remodelling on IVUS, while the ivory colour defines fibrous tissue on OCT. IVUS, intravascular ultrasound; NIRS, near-infrared spectroscopy; OCT, optical coherence tomography; RF, radiofrequency.
Figure 1

Advantages and limitations of the clinically available intravascular imaging modalities for assessing vessel wall pathology. The table at the bottom of the figure provides a quantification of the efficacy of each modality in detecting different tissue types using histology as the reference standard. The symbol (−) indicates that the modality is unable to detect the specific characteristic. Conversely, the symbols (+), (++), and (+++) indicate weak, moderate, and excellent ability of the modality to detect a plaque characteristic, respectively. White arrows indicate neo-vessels, cholesterol crystal, erupted calcified nodule, thin fibrous cap and macrophages infiltration in cross-sectional OCT images. Asterisks indicate calcific tissue on IVUS (upper panel) and OCT (lower panel) and evidence of intraluminal thrombus on OCT. A red arc defines the lipid necrotic core in NIRS-IVUS image. The superimposed light blue colour indicates plaque burden and positive remodelling on IVUS, while the ivory colour defines fibrous tissue on OCT. IVUS, intravascular ultrasound; NIRS, near-infrared spectroscopy; OCT, optical coherence tomography; RF, radiofrequency.

The aim of this review is to present the findings of intravascular imaging studies evaluating the efficacy of different drugs, summarize the results of clinical trials that tested their prognostic value, and discuss the advantages and limitations of invasive imaging endpoints in predicting the potency of these therapies in reducing cardiovascular event rates.

2. The rationale behind intravascular imaging for assessing the efficacy of novel therapies targeting atherosclerosis

The use of intravascular imaging-based surrogate endpoints to examine the value of novel pharmacotherapies in improving outcome relies on the premise that specific plaque characteristics, which can be assessed by intravascular imaging, are associated with a risk of subsequent major adverse cardiovascular events (MACE).

Cumulative data have shown that PB and its change provide useful prognostic information and identification of patients at risk of future events. A metanalysis including 4137 patients recruited in 6 clinical trials showed that baseline PAV and its change at 18–24 months follow-up were independent predictors of MACE.6

The above findings were also confirmed in prospective studies reporting patient-level results. In the European Collaborative Project on Inflammation and Vascular Wall Remodeling in Atherosclerosis-Intravascular Ultrasound (ATHEROREMO-IVUS) study that included 581 patients undergoing single vessel virtual histology (VH)-intravascular ultrasound (IVUS), patients with PB ≥ 70% and a thin-cap fibroatheroma (TCFA) phenotype were at high-risk of developing MACE.7 The prognostic value of plaque morphology was also shown in the ATHEROREMO-near-infrared spectroscopy (NIRS),8 the lipid-rich plaque (LRP)9 and the Providing Regional Observations to Study Predictors of Events in the Coronary Tree (PROSPECT) II10 studies which demonstrated that increased lipid component detected by NIRS was associated with worse prognosis on a patient-level analysis (Figure 2).

Design and outcome of studies assessing the efficacy of intravascular imaging in predicting events. ACS, acute coronary syndrome; AS, area stenosis; CFD, computational fluid dynamics; DS, diameter stenosis; FCT, fibrous cap thickness; IB, integrated backscatter; IVUS, intravascular ultrasound; LCBI, lipid core burden index; maxLCBI4mm, maximum LCBI in 4 mm segment; MI, myocardial infarction; MLA, minimum lumen area; NIRS, near-infrared spectroscopy; NPV, negative predictive value; OCT, optical coherence tomography; PAV, per cent atheroma volume; PB, plaque burden; PCI, percutaneous coronary intervention; PPV, positive predictive value; RI, remodelling index; TCFA, thin-cap fibroatheroma; UA, unstable angina; VH, virtual histology; WSS, wall shear stress.
Figure 2

Design and outcome of studies assessing the efficacy of intravascular imaging in predicting events. ACS, acute coronary syndrome; AS, area stenosis; CFD, computational fluid dynamics; DS, diameter stenosis; FCT, fibrous cap thickness; IB, integrated backscatter; IVUS, intravascular ultrasound; LCBI, lipid core burden index; maxLCBI4mm, maximum LCBI in 4 mm segment; MI, myocardial infarction; MLA, minimum lumen area; NIRS, near-infrared spectroscopy; NPV, negative predictive value; OCT, optical coherence tomography; PAV, per cent atheroma volume; PB, plaque burden; PCI, percutaneous coronary intervention; PPV, positive predictive value; RI, remodelling index; TCFA, thin-cap fibroatheroma; UA, unstable angina; VH, virtual histology; WSS, wall shear stress.

Prospective large-scale studies which investigated clinical endpoints on a lesion-level analysis also provided relevant data on the value of intravascular imaging in identifying vulnerable plaques. The PROSPECT,11 the VH-IVUS in vulnerable atherosclerosis (VIVA),12 the Prediction of Progression of Coronary Artery Disease and Clinical Outcome Using Vascular Profiling of Shear Stress and Wall Morphology (PREDICTION),13 and the PROSPECT II10 have underscored the prognostic implications of PB and composition and highlighted the efficacy of intravascular imaging in detecting plaques that were prone to progress and cause events. In addition, the relationship between optical coherence tomography (OCT) coronary plaque morphology and clinical outcome (CLIMA)14 was the first study demonstrating that the thickness of fibrous cap over LRP and vascular inflammation indicated by the presence of macrophages on OCT imaging were independent predictors of MACE (Figure 2).

Based on the findings of the above studies and the evidence from histology reports showing that culprit lesions have a specific phenotype,15–17 it has been hypothesized that a decrease in PB, necrotic core and vascular inflammation as well as an increase in fibrous cap thickness (FCT) over LRP indicate plaque passivation and thus these variables can be used as surrogate endpoints to predict the efficacy of emerging therapies targeting atherosclerosis in reducing cardiovascular events.5

3. Efficacy of drug therapies in modifying plaque size and composition and improving outcomes

In this section, we focused our attention on pharmacotherapies, introduced to inhibit plaque evolution, that have been tested in intravascular imaging-based studies (see Supplementary material online, Tables S1 and S2) and/or outcome trials (see Supplementary material online, Table S3) in secondary prevention of CAD.

3.1 Statins

Statins target hepatocytes and are selective, competitive inhibitors of hydroxymethylglutaryl-CoA (HMG-CoA) reductase, a key regulator of cholesterol biosynthesis (see Supplementary material online, Figure S2). The reduction in intracellular cholesterol production causes upregulation of hepatic LDL receptors which decreases levels of circulating LDL as well as oxidized LDL within the arterial intima thwarting the inflammatory cascade that promotes monocyte recruitment and foam cell formation, the initial and key step in atherogenesis. Furthermore, statins have cholesterol-independent cardiovascular protective effects that include reduction of oxidative stress and platelet aggregation, vascular tone improvement (by increasing nitric oxide synthesis and reducing smooth muscle cell activation and proliferation), plaque stabilization (promoting macrocalcification and increase in FCT), as well as systemic and local anti-inflammatory effects [i.e. they reduce C-reactive protein (CRP), tumour necrosis factor (TNF) alpha, interleukin (IL)-1beta, and leucocyte-endothelial adhesion].18

The prognostic implications of statin therapy are well established and seem to be associated with the reduction in LDL-C induced by these drugs (see Supplementary material online, Figure S3). A meta-analysis including 21 statin trials and more than 129 000 patients showed that at every 1.0 mmol/L reduction in LDL-C there is a 22% reduction in cardiovascular events and a 10% reduction in all-cause mortality.19 In addition, numerous intravascular imaging studies have attempted to provide mechanistic insights and examine the effects of statin therapy on plaque morphology and burden.

3.1.1 Rosuvastatin

The ASTEROID (A Study to Evaluate the Effect of Rosuvastatin on Intravascular Ultrasound-Derived Coronary Atheroma Burden),20 a single-arm observational study, and the SATURN (Study of Coronary Atheroma by Intravascular Ultrasound: Effect of Rosuvastatin Versus Atorvastatin),21 an appropriately powered randomized control trial, showed that treatment with rosuvastatin 40 mg daily led to a marginal but statistical significant reduction in PAV (−0.98%, P < 0.001 and −1.22%, P < 0.001, respectively) on IVUS imaging at 2-year follow-up. An exploratory sub-analysis of the SATURN—including 71 patients undergoing serial VH-IVUS—demonstrated no changes in the lipid and fibrotic tissue components but an increase in the calcific and a reduction in the fibrofatty tissue burden at follow-up.22

The IBIS-4 (The fourth Integrated Biomarker and Imaging Study)4 confirmed that rosuvastatin 40 mg reduces PAV by −0.9% (P = 0.007) in patients with ST-elevation myocardial infarction (STEMI), while VH-IVUS analysis showed an increase in the calcific burden and a reduction of the fibrotic tissue component but no change in the plaque phenotype at follow-up. The OCT-sub-study of IBIS-4 demonstrated that this drug regimen may promote plaque stabilization increasing FCT over LRP by +24.4 μm (P = 0.008) and reducing macrophages accumulation at 13-month follow-up (see Supplementary material online, Table S2).3 An advantage of the IBIS-4 study is the fact that it studied the entire coronary tree including the culprit and non-culprit vessels with both VH-IVUS and OCT at two-time points to identify changes in plaque morphology; however, its main limitation is the lack of a control group that would allow us to test the superiority of this regimen over low-dose statin therapy.

Conversely, the IBIS-3 (The third Integrated Biomarker and Imaging Study)23 showed that rosuvastatin 40 mg did not change the necrotic core volume at 6 or 12-month follow-up (see Supplementary material online, Table S2). This was an observational single-arm study that aimed to recruit 300 patients but managed to enrol 241 patients. From these patients, 164 had evaluable serial VH-IVUS imaging, while 103 matched baseline and follow-up NIRS. The study was underpowered for the primary endpoint that was the change in the necrotic core volume at follow-up assessed by VH-IVUS. The secondary endpoint of the study was the change in the lipid core burden index (LCBI) of the studied segment assessed by NIRS, and the authors found no difference in the LCBI at follow-up; however, for this endpoint there was no power calculation. For the above reasons, the IBIS-3 findings should be interpreted with caution.

Multimodality IVUS and NIRS imaging was also used in the YELLOW (Reduction in Yellow Plaque by Aggressive Lipid LOWering Therapy) study24 to evaluate the short-term implications (6–8 week follow-up) of rosuvastatin 40 mg in flow-limiting lesions. Patients treated with this regimen exhibited a higher reduction in maxLCBI4 mm compared to the control group, but there were no changes in PB at follow-up (see Supplementary material online, Table S1). The small number of recruited patients, differences in plaque composition between groups and the need for NIRS and IVUS co-registration at two-time points raised concerns about the validity of the reported results. Furthermore, these findings were not confirmed in the YELLOW II study25 which had a similar design and demonstrated no difference in the maxLCBI4mm between baseline and follow-up. However, in the YELLOW II also serial OCT was used showing a significant increase in minimum FCT and decrease in the incidence of TCFA (see Supplementary material online, Table S2), but it was an exploratory analysis because the study was not powered for these endpoints.

Finally, the STABLE (Statin and Atheroma Vulnerability Evaluation) study26 which randomized patients with non-flow-limiting disease to high or moderate-dose of rosuvastatin showed a similar reduction in both groups in the per cent necrotic core volume and in the incidence of VH-IVUS-defined TCFA at 1-year follow-up (see Supplementary material online, Table S1). However, the study included only 225 patients with serial intravascular imaging instead of 276 and, thus, it was underpowered for the primary endpoint—that was the change in VH-defined per cent compositional volume within the target segment from baseline to follow-up—and for the secondary endpoint—defined as the change in per cent compositional volume at follow-up between the two treatment groups.

Studies investigating the prognostic implications of rosuvastatin in secondary prevention of CAD are limited and were performed only in patients with chronic heart failure. Two randomized studies27,28 including patients suffering from ischaemic or non-ischaemic heart failure failed to demonstrate a prognostic benefit of rosuvastatin in this cohort (see Supplementary material online, Table S3); however, a patient-level metanalysis29 of these two studies demonstrated that treatment with rosuvastatin is associated with a lower incidence of myocardial infarction (MI) in patients suffering from ischaemic heart disease (HR 0.81, 95% CI 0.66–0.99, P < 0.05); therefore, it is expected that rosuvastatin will be equally effective as the other statins in preventing MACE in patients with CAD.

3.1.2 Atorvastatin

Several studies have underscored the beneficial effects of intensive or moderate-dose atorvastatin therapy on PB. In the REVERSAL (Reversal of Atherosclerosis with Aggressive Lipid Lowering) study30 atorvastatin 80 mg daily inhibited disease progression, whereas there was plaque progression on IVUS in the pravastatin group at 18-month follow-up (total atheroma volume, TAV: −0.4 vs. +2.7%, P = 0.02). Conversely, there was no difference in PAV changes between the rosuvastatin and atorvastatin group in the SATURN 21 where atorvastatin 80 mg daily reduced PAV by −0.99% (P < 0.001) at follow-up.

Small randomized studies demonstrated that even low-dose atorvastatin may have beneficial effects on atheroma burden assessed by IVUS in patients with acute coronary syndrome (ACS)31 and in those with mild coronary atherosclerotic lesions (see Supplementary material online, Table S1).32

The Effect of Atorvastatin Therapy on Fibrous Cap Thickness in Coronary Atherosclerotic Plaque as Assessed by Optical Coherence Tomography (EASY-FIT) was the only prospective and properly powered study that compared the implications of two atorvastatin doses (20 mg vs. 5 mg daily) on FCT. This study showed that the higher atorvastatin dose significantly increased FCT at 12-month follow-up leading to plaque stabilization (69% vs. 17%, P < 0.001).33

The prognostic value of atorvastatin therapy in patients with CAD was first tested in the PROVE IT-TIMI22 (Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22) trial34 that randomized 4162 patients who suffered an ACS to atorvastatin 80 mg or pravastatin 40 mg and described a lower MACE rate in the atorvastatin arm (22.4% vs. 26.3%, P = 0.005) at 2-year follow-up. In patients with stable CAD, the TNT (Treating to New Targets) trial35 demonstrated that 80 mg/day of atorvastatin was more effective than 10 mg/day in reducing the risk of death from CAD, non-fatal MI, resuscitation after cardiac arrest or stroke over a median follow-up of 4.9 years (see Supplementary material online, Table S3).

3.1.3 Pravastatin and pitavastatin

The effects of pravastatin and pitavastatin on PB and composition have also been studied.

In the REVERSAL trial,30 treatment with pravastatin 40 mg was associated with an increase in TAV at follow-up as previously documented. The JAPAN-ACS (Japan Assessment of Pitavastatin and Atorvastatin in Acute Coronary Syndrome) study demonstrated that treatment with pitavastatin 4 mg/day was associated with a significant reduction in PAV in the treated culprit vessel at 8–12 month follow-up with no significant differences compared to a low-dose atorvastatin regimen.31 The TRUTH36 (treatment with statin on atheroma regression evaluated by IVUS with VH) was a small prospective study that randomized Japanese patients to pitavastatin or pravastatin and showed that both statin regimens modified plaque composition by reducing the fibrofatty and increasing the calcified plaque component assessed by VH-IVUS at 8-month follow-up.

The CARE (Cholesterol and Recurrent Events) trial examined the prognostic value of pravastatin in secondary prevention and showed that pravastatin 40 mg/day was more effective than placebo in reducing fatal and non-fatal coronary events (24% relative risk reduction, 95% CI: 9–36%; P = 0.003) in patients with ACS who had normal cholesterol.37 Similarly, the LIPID study (long-term intervention with pravastatin in ischaemic disease) showed that pravastatin reduced cardiovascular events and all-cause mortality compared to placebo in patients who had a previous ACS (see Supplementary material online, Table S3).38 Pitavastatin does not have an indication in secondary prevention, but it is approved for the treatment of primary hyperlipidaemia.

3.2 Ezetimibe plus statin

Ezetimibe is a lipid-lowering drug that targets the Niemann–Pick C1-like 1 protein and localizes in the brush border of the small intestinal enterocytes reducing the uptake of cholesterol into the enterocytes (see Supplementary material online, Figure S2) and its overall delivery to the liver, thereby promoting the synthesis of LDL receptors with a subsequent reduction of serum LDL-C. Apart from this systemic effect, recent studies suggested that ezetimibe also inhibits macrophages migration by decreasing VCAM-1, MCP-1, and TNF-α levels and reduces ROS levels that are instigators of plaque progression.39

When added to statins, ezetimibe reduces LDL-C levels by −22.3% compared to placebo40; therefore, ezetimibe is recommended as an add-on therapy in patients who do not reach the LDL-C goal with the maximum tolerated dose of statin.41

The PRECISE-IVUS (Plaque Regression With Cholesterol Absorption Inhibitor or Synthesis Inhibitor Evaluated by Intravascular Ultrasound) is the largest study investigating the effect of ezetimibe on PB; it randomized 246 patients to atorvastatin 10 mg plus ezetimibe 10 mg daily or atorvastatin alone and demonstrated a greater PAV regression (−1.4% vs. −0.3%, P = 0.001) in the dual-therapy group.42 A recently published meta-analysis pooling data from the PRECISE-IVUS study and five smaller studies comprising 583 patients in total confirmed that the combination of ezetimibe and statin therapy was more effective than statin monotherapy in reducing atheroma volume.43 Small scale studies examining the role of combined ezetimibe and statin therapy on plaque composition have consistently demonstrated that the addition of ezetimibe has no significant effect on the changes of plaque characteristics at follow-up (see Supplementary material online, Table S1).

The clinical benefit of combining ezetimibe and a statin was shown for the first time in the IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial)44 which randomized 18 144 patients admitted with ACS to simvastatin 40 mg plus ezetimibe 10 mg once a day or simvastatin 40 mg monotherapy. A 6.4% relative risk reduction in the dual-therapy group was noted for the composite endpoint of cardiovascular death, non-fatal MI, unstable angina requiring rehospitalization, coronary revascularization or non-fatal stroke at 7-year follow-up (32.7% vs. 34.7%, P = 0.016). A similar trend was also reported in the HIJ-PROPER trial (Heart Institute of Japan Proper level of lipid lOwering with Pitavastatin and Ezetimibe in acute coRonary syndrome) that included 1734 patients with ACS and dyslipidaemia who were randomized to dual therapy with ezetimibe 10 mg and pitavastatin 2 mg daily or pitavastatin monotherapy; however, the difference in the event rate between groups was not statically significant as the study was underpowered for the primary endpoint (see Supplementary material online, Table S3).45

3.3 Proprotein convertase subtilisin-kexin type 9 inhibitors

Proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors prevent the degradation of LDL receptors which in turn increase their number on the surface of hepatocytes and promote LDL clearance (see Supplementary material online, Figure S2); these drugs can reduce LDL-C by 45–60% if used alone or in combination with a statin.46,47 PCSK9 inhibitors may also have pleiotropic effects as it appears that PCSK9 is expressed by various cell types involved in atherosclerosis and it affects endothelial function, promotes smooth muscle cell migration, and exerts paracrine function on macrophages in the intima increasing the expression of pro-inflammatory cytokines and modifying the uptake of oxidized LDL.48

The GLAGOV (Global Assessment of Plaque reGression with a PCSK9 antibOdy as Measured by intraVascular Ultrasound)2 was the first trial that examined the effect of PCSK9 inhibitors on PB. The included patients were randomized to evolocumab plus high or moderate-intensity statin or statin monotherapy and had serial IVUS imaging in a single vessel with non-significant stenosis at baseline and at 76-week follow-up. In this study, the PAV decreased by −0.95% (P < 0.001) in the combined therapy group, while it remained unchanged in the statin monotherapy arm. A pre-specified sub-study of GLAGOV49 which included patients who had concomitant VH-IVUS imaging showed no differences in changes in plaque composition between groups (see Supplementary material online, Table S1).

Conversely, the ODYSSEY-J IVUS (Evaluation of Effect of Alirocumab on Coronary Atheroma Volume in Japanese Patients Hospitalized for Acute Coronary Syndrome With Hypercholesterolemia) trial failed to demonstrate any difference in the changes in PAV or TAV between patients treated with alirocumab and a statin (rosuvastatin ≥5 mg/day or atorvastatin ≥10 mg/day) and those receiving statin monotherapy at 36-week follow-up.50 However, this study had significant limitations: power calculation assumed a large % change difference in the normalized TAV between the two groups that led to the recruitment of a small number of patients; IVUS imaging was performed in both culprit and non-culprit vessels where it is likely the TAV to be different and thus introduce bias; ezetimibe was added in the control group in 40% of the patients during the follow-up period.

A recent randomized study51 including only 48 patients showed a reduction in the lipid index and macrophages grade and an increase in the FCT assessed by OCT in patients treated with alirocumab compared to those receiving statin, whereas an observational report52 of 53 patients showed a reduction in the maxLCBI4mm on NIRS-IVUS imaging in patients treated with PCSK9 inhibitors compared to those being on statin monotherapy. However, in the former study no power calculation was performed, while the latter included two different PCSK9 inhibitors and was not a randomized study. Therefore, both reports should be regarded as exploratory analyses and their findings require confirmation in the two large appropriately powered randomized control studies that are currently ongoing. The Imaging of Coronary Plaques in Subjects Treated With Evolocumab (HUYGENS; NCT03570697)53 trial aims to assess the effect of treatment with evolocumab on FCT in 164 patients admitted with an non-STEMI, while in the Vascular Effects of Alirocumab in Acute MI-Patients (PACMAN-AMI; NCT03067844)54 study serial NIRS-IVUS and OCT imaging has been performed in 300 patients with acute MI to assess the effect of alirocumab on plaque volume, lipid burden, and FCT.

Large outcomes trials reported that PCSK9 inhibitors combined with a statin therapy decrease the risk of MACE. In the FOURIER (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk) trial, evolocumab reduced the rate of MI (3.4% vs. 4.6%, P < 0.001) and coronary revascularization (5.5% vs. 7%, P < 0.001) compared to placebo in statin-treated patients at a median follow-up of 2.2 years, without reducing cardiovascular mortality.55 Similarly, the ODYSSEY OUTCOMES (Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab) trial that included patients with an ACS showed that the addition of alicorumab to high-dose statin therapy reduces the incidence of recurrent cardiovascular events at 2.8-year follow-up (see Supplementary material online, Table S3).56

3.4 Drugs increasing HDL cholesterol

3.4.1 HDL mimetics

HDL cholesterols have been a target for drug development because of their anti-atherogenic properties. Several HDL mimetic drugs have been developed by combining peptides and proteins with varying structures, all of which bind lipids found in HDL. Infusion of reconstituted HDL directly increases the number of functional HDL particles in the circulation and thus cholesterol efflux capacity,57 in other words it promotes the extraction of cholesterol from donor cells located in peripheral tissues—such as macrophages—and its transportation to the liver (see Supplementary material online, Figure S2).

HDL mimetic agents have attracted attention after animal studies highlighting their athero-protective effects.58,59 Over the last 20 years three different HDL mimetics (ETC-126, now called MDCO-216, CER-001, and CSL-111) have been evaluated in randomized control trials which enrolled patients with history of ACS and used invasive imaging surrogate endpoints to assess their value in inhibiting plaque progression. Apart from the first study that showed that 5-week ETC-216 infusion reduces PB on IVUS imaging,60 all the other trials demonstrated a neutral effect of HDL mimetics on PB (see Supplementary material online, Table S1).61–64 A possible explanation of this paradox is the small number of patients included in the first report that did not allow to draw safe conclusion; in addition, patients recruited in recent studies received contemporary treatment for atherosclerotic disease and had normal HDL-cholesterol levels which may limit the effect of HDL mimetics on plaque pathobiology. Whether infusion of HDL mimetics is effective in the context of strongly reduced HDL-cholesterol levels and impaired cholesterol efflux capacity remains unknown.

There is no current evidence on the role of HDL mimetics in preventing cardiovascular events. The efficacy and safety of CSL-112 in patients after an ACS is currently under investigation in the ApoA-I Event Reducing in Ischaemic Syndromes II (AEGIS II, NCT03473223) trial.65

3.4.2 Cholesterylester transfer protein inhibitors

Cholesterylester transfer protein (CETP) is a hydrophobic glycoprotein that is synthesized mainly in the liver and regulates the exchange of lipids between different lipoprotein particles. This process leads to a net mass transfer of cholesterol esters and triglycerides from non-atherogenic HDLs to ApoB100-containing lipoproteins such as very low-density lipoproteins (VLDLs) and LDLs that are pro-atherogenic (see Supplementary material online, Figure S2). Inhibition of this pathway eventually increases the content of cholesterol in HDL particles and the formation of larger HDL particles that are catabolized slower than the normal HDL.66

The role of these agents in modifying plaque size has been evaluated in the Investigation of Lipid Level Management Using Coronary Ultrasound to Assess Reduction of Atherosclerosis by CETP Inhibition and HDL Elevation (ILLUSTRATE) trial67 which randomized patients with evidence of mild to moderate CAD to atorvastatin alone and atorvastatin in addition to torcetrapib 60 mg daily and showed no differences in PAV changes between the two groups after 24 months (see Supplementary material online, Table S1).

Interestingly, the Investigation of Lipid Level Management to Understand its Impact in Atherosclerotic Events (ILLUMINATE) trial68 demonstrated that torcetrapib in association with atorvastatin significantly increased the risk of MACE and all-cause mortality compared to atorvastatin in patients with history of type 2 diabetes mellitus or previous cardiovascular disease. Conversely, anacetrapib—another CETP inhibitor—demonstrated promising results in reducing MACE,69 while evacetrapib had a neutral effect on MACE70 when compared to placebo in patients with cardiovascular disease (see Supplementary material online, Table S3). A possible explanation of the prognostic benefit of anacetrapib is the fact that, in contrast to the other CETP inhibitors, this medication not only enhances reverse LDL transport, but also reduces apolipoprotein b levels. On the other hand, evacetrapib and torcetrapib increase apoA1 in HDL subspecies containing apoC3 and other HDL subspecies associated with increased risk of CAD; these mechanisms may explain their lack of clinical benefits although they raise HDL.71

4. Anti-inflammatory drugs

Apart from lipid-lowering drugs that have an established role in reducing MACE in patients with CAD, recent evidence indicates that aggressive inhibition of inflammation may also improve prognosis in this population.72 Several anti-inflammatory drugs have been introduced to inhibit vascular inflammation; the lipoprotein-associated phospholipase A2 (Lp-PLA2) inhibitor darapladib was the first drug that has been evaluated in both intravascular imaging and large-scale outcome trials.

4.1 Darapladib

Darapladib is a direct inhibitor of the Lp-PLA2 which is a calcium-independent enzyme secreted by the inflammatory cells (including monocyte-derived macrophages, T cells, and mast cells), circulates in plasma in its active form and it is primarily bound to LDL-cholesterol. Lp-PLA2 is involved in the metabolism of oxidized LDL (see Supplementary material online, Figure S2) generating potent pro-inflammatory mediators that contribute to plaque development, progression, and destabilization by promoting foam cell formation, endothelial dysfunction, and apoptosis.73–75 Histological data76 showed that the concentration of Lp-PLA2 protein is increased in TCFAs compared to smaller and more stable plaques, and there is evidence that raised Lp-PLA2 plasma levels are associated with coronary events.77

In the Integrated Biomarker and Imaging Study-2 (IBIS-2), 12-month therapy with darapladib had no effect on TAV but it appeared to inhibit necrotic core progression, resulting in a significant difference in the changes of the necrotic core volume between patients treated with darapladip and the control group at follow-up (see Supplementary material online, Table S1).78 Of note, the IBIS-2 study was not powered to assess changes in TAV and composition which were secondary endpoints of the study but to examine the effect of darapladib on plaque deformability estimated by palpography, a modality that later was proven unreliable in assessing plaque vulnerability.79

The effects of darapladip on clinical outcomes were examined in the STABILITY (Stabilization of Atherosclerotic Plaque by Initiation of Darapladib Therapy) trial which included 15 828 patients with stable CAD randomized to darapladib or placebo who were followed up for 3.7 years.80 Darapladib did not reduce the primary composite endpoint of cardiovascular death, MI, or stroke, but decreased the rate of major and total coronary events. Similarly, the SOLID-TIMI 52 (Stabilization of Plaque Using Darapladib-Thrombolysis in Myocardial Infarction 52) trial that included 13 026 patients admitted with ACS showed no prognostic benefit of darapladib at 3-year follow-up (see Supplementary material online, Table S3).81

4.1.1 Methotrexate

Methotrexate is a chemotherapy agent and immune-system suppressant that inhibits the enzyme dihydrofolate reductase essential for nucleotide synthesis. There is evidence that patients with chronic inflammatory diseases such as rheumatoid or psoriatic arthritis treated with low-dose methotrexate had fewer cardiovascular events than patients who received other therapies or placebo.82,83 Methotrexate appears to suppress YAP (yes-associated protein 1) activation that leads to a reduction in the levels of inflammatory factors (e.g. IL-6, connective tissue growth factor) and adhesion molecules; and through this mechanism, it may inhibit atherosclerotic disease progression.84

The National Institutes of Health-sponsored CIRT (Cardiovascular Inflammation Reduction Trial) that included 4786 patients who had stable atherosclerosis with diabetes mellitus or metabolic syndrome reported that low-dose methotrexate did not reduce MACE (see Supplementary material online, Table S3).85 A possible explanation of this finding is the fact that low-dose methotrexate also failed to reduce the plasma levels of IL-1β, IL-6, or CRP.

4.1.2 Canakinumab

Canakinumab is a recombinant human monoclonal antibody that has anti-inflammatory effects by selectively inhibiting IL-1β receptor binding. IL-1β is released following activation of the NLRP3 inflammasome and plays a central role in the systemic inflammatory response by increasing the production of IL-6 by various cell types and driving its signalling pathway.86 IL-6 mediates the acute phase response by stimulating the liver to produce proteins for host defences, but also promotes thrombosis and inhibits fibrinolysis. IL-1β has also direct pro-atherogenic effects as it increases the expression of leucocyte adhesion molecules and thrombogenic mediators and promotes smooth muscle cells proliferation and endothelial cells activation.87,88

The CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcome Study)89 demonstrated that in patients with previous MI and evidence of ongoing sub-clinical inflammation—defined by a high-sensitivity CRP ≥2 mg/L—inhibition of IL-1β with canakinumab at a dose of 150 mg every 3 months was associated with a 15% reduction in MACE regardless of lipid-level lowering (see Supplementary material online, Table S3). A subsequent analysis of the CANTOS trial found that random allocation to canakinumab compared to placebo reduced the total number of serious cardiovascular events during a median follow-up of 3.7 years irrespective of the administrated dose.90 However, this drug did not have any effect on the all-cause mortality compared to placebo due to a higher rate of fatal infections.89

4.1.3 Colchicine

Colchicine is a unique anti-inflammatory agent with broad cellular effects. It is accumulated in leucocytes, and by binding to microtubules and interfering with their function this drug modulates the expression of cytokines and interleukins, and the ability of neutrophils to marginate, ingress, aggregate, express superoxide, release neutrophil extracellular traps, and interact with platelets.91

In a serial angiography and IVUS study, Deftereos et al.92 demonstrated that colchicine was associated with a lower incidence of binary restenosis due to a reduction in the normalized neointima volume in diabetic patients treated with bare metal stents. However, there is lack of evidence on the effect of colchicine in preventing coronary plaque evolution in native segments.

So far, four independent randomized controlled trials have evaluated the effect of colchicine in patients with acute and chronic coronary syndromes (see Supplementary material online, Table S3). The Low-Dose Colchicine (LoDoCo) study,93 which included patients with stable CAD, demonstrated a reduction in the incidence of cardiovascular events in patients treated with colchicine; however, this was an open-label trial involving only 532 patients. The Low-Dose Colchicine 2 (LoDoCo2)94—a randomized, controlled, double-blind, event-driven trial—enrolled 5522 patients who were randomized to placebo or colchicine 0.5 mg once daily and demonstrated that colchicine prevents cardiovascular events. Similarly, the Colchicine Cardiovascular Outcomes Trial (COLCOT)95 showed a reduction in the incidence of MACE by −23% in patients with ACS at 2-year follow-up. Nevertheless, in contrast to the LoDoCo and COLCOT trials, the Australian Colchicine in Patients with Acute Coronary Syndrome (COPS)96 trial demonstrated that colchicine did not reduce cardiovascular events at 1-year follow-up and there was a trend toward a higher rate of all-cause mortality in the colchicine group compared to placebo; however, this study was underpowered to assess the effect of colchicine on clinical outcome. A recent metanalysis97 including 11 816 patients with CAD showed that colchicine reduced MACE rate as well as the risk of MI, stroke, and coronary revascularization compared to placebo, with no significant differences in all-cause mortality or cardiovascular death between the two groups.

The above studies were important to highlight the value of colchicine in secondary prevention but also had significant limitations. None of them used clinical or biological markers of inflammation for the selection of participants, cholesterol levels or blood pressure at enrolment were not reported and they recruited predominantly male patients. In the coming years, the CLEAR SYNERGY and the COLCARDIO trial (ACTRN12616000400460) will provide additional evidence on the efficacy and long-term safety of colchicine in patients with ACS.

5. Other therapies

5.1 Antihypertensive agents

Several antihypertensive agents, in particular angiotensin-converting enzyme (ACE) inhibitors and calcium channel blockers, have been tested in intravascular imaging-based studies to investigate their role in inhibiting plaque progression. The rationale behind their potential benefits in inhibiting atherosclerosis might be explained by the pleiotropic effects of these drugs in addition to blood pressure reduction. For instance, ACE inhibitors downregulate the pro-atherogenic effects induced by angiotensin-II that increases oxidative stress, expression of inflammatory cytokines, and adhesion molecules modulating endothelial function as well as cellular migration and proliferation.98 Calcium channel blockers may affect smooth muscle cells proliferation and migration, increase lipid resistance to oxidative stress, and improve endothelial function by inhibiting apoptosis and modulating nitric oxide expression.99

The Comparison of Amlodipine vs. Enalapril to Limit Occurrences of Thrombosis (CAMELOT) study showed that neither amlodipine nor enalapril reduced PAV after 2 years of treatment.100 In the PERindopril's Prospective Effect on Coronary aTherosclerosis by Angiography and IntraVascular Ultrasound Evaluation (PERSPECTIVE) study,101 no difference was noted between perindopril and placebo in the changes in plaque area at 3-year follow-up. Similar findings were reported by the Effect of Nifedipine on Coronary Endothelial Function and Plaque Formation in Patients With Coronary Artery Disease (ENCORE II) study where nifedipine did not reduce PAV compared to placebo during a follow-up of 18–24 months (see Supplementary material online, Table S1).102 It has to be stressed, however, that IVUS-based endpoints were not the primary outcomes of these studies and thus none of them was powered to detect differences in PB.

In line with the intravascular imaging studies, calcium channel blockers do not seem to improve outcomes in patients with CAD. In the PREVENT (Prospective Randomized Evaluation of the Vascular Effects of Norvasc Trial) study amlodipine 5–10 mg did not reduce MACE over a 3-year follow-up period, despite reducing hospital admissions for unstable angina and coronary revascularization.103 Similarly, the ACTION (A Coronary disease Trial Investigating Outcome with Nifedipine) study showed no prognostic benefit of treatment with nifedipine in patients with stable CAD.104 Conversely, the HOPE (Heart Outcomes Prevention Evaluation)105 and the EUROPA (EUropean Trial on Reduction of Cardiac Events with Perindopril in stable CAD)106 trials which evaluated the prognostic benefit of treatment with ramipril and perindopril, respectively, showed that both drugs improve outcomes in patients with established CAD. On the other hand, in the Prevention of Events with Angiotensin-Converting Enzyme inhibition (PEACE) trial, trandolapril did not improve prognosis in patients with stable CAD and preserved left ventricular systolic function (see Supplementary material online, Table S3).107 This paradox can be explained by the fact that ACE inhibitors are not equally effective against cardiovascular disease; perindopril may be superior to trandolapril in this setting as it has a beneficial effect on endothelial function.108,109

5.1.1 Antidiabetic drugs

The implications of oral glucose-lowering agents on PB have been investigated in IVUS-based studies. In the Pioglitazone Effect on Regression of Intravascular Sonographic Coronary Obstruction Prospective Evaluation (PERISCOPE) study,110 pioglitazone was superior to glimepiride in inhibiting plaque progression (PAV: −0.16% vs. +0.73%, P = 0.02), whereas the Assessment on the Prevention of Progression by Rosiglitazone on Atherosclerosis in Diabetes Patients With Cardiovascular History (APPROACH) study111 showed no difference between the effect of rosiglitazone and glipizide on PAV (see Supplementary material online, Table S1).

A meta-analysis demonstrated that pioglitazone reduces the risk of MACE, stroke, and MI in patients with previous history of cardiovascular disease, but it also increases the risk of heart failure.112 In contrast, in a sub-analysis of the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial rosiglitazone did not affect MACE in patients with type 2 diabetes mellitus and established CAD.113 Similarly, the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS) study showed that sitaglipitin was not associated with a higher risk of MACE and hospitalization for heart failure when added to usual care in patients with type 2 diabetes mellitus and cardiovascular disease (see Supplementary material online, Table S3).114

6. Discussion

From the above studies, it is apparent that the pharmacotherapies which have been associated with a prognostic benefit in large-scale outcome trials have a minimal but consistent effect on PB among appropriately powered intravascular imaging studies, while there is limited data about their implications on plaque composition. Studies assessing the effects of emerging therapies on necrotic core component, FCT, and macrophage accumulation have included small numbers of patients or have not been powered for these endpoints; therefore, further research is needed towards this direction to fully explore the potential of these intravascular imaging-based surrogate endpoints in predicting the prognostic value of novel therapies.

Although there is a consistency between intravascular imaging and outcome studies—i.e. drugs that reduce PB are associated with prognostic benefit—it seems that the changes in PB are disproportionately lower than the reduction of events reported in clinical studies. This could be due to differences in inclusion criteria between studies, but also to the fact that an increased PB does not necessarily indicate a high-risk lesion. In the PROSPECT study,11 PB ≥70% was the strongest predictor of MACE but it had a low positive predictive value for events (9.5%) in a lesion-level analysis.

Plaque features such as lipid burden, thin fibrous cap over lipid tissue, and macrophage accumulation have been associated with plaque vulnerability and future events. For instance, in the PROSPECT II study10 PB and composition assessed by NIRS-IVUS provided complementary information and these variables were independent predictors of MACE. Similar were the findings of the CLIMA study14 where patients with lesions characterized by small lumen area, increased lipid component, a thin fibrous cap over the lipid tissue and macrophages infiltration were at higher risk of suffering future events.

It is likely that pharmacotherapies targeting coronary plaque evolution may change not only PB but also its composition and reduce vascular inflammation leading to plaque passivation.

6.1 Multimodality–hybrid imaging for more accurate assessment of plaque phenotype changes

Over the recent years, there has been a shift toward the use of multimodality imaging to better assess vessel wall response to treatment with novel therapies.3,4,25,115 However, this approach has also significant limitations. Firstly, all the existing intravascular imaging modalities have limited efficacy in assessing some high-risk plaque features, such as its composition, the presence of macrophages, FCT, cholesterol crystals, and neo-vessels (Figure 1). Secondly, the co-registration of the imaging data acquired by two different catheters is a challenging and tedious process that is prone to errors. Advances in hybrid intravascular imaging and the design of catheters with multiple imaging probes for simultaneous data acquisition are expected the overcome this limitation enabling a complete and comprehensive assessment of plaque composition and biology.115

Moreover, without prospective evidence from large-scale multimodality intravascular imaging studies, there are no established scores that combine different plaque features to allow more accurate quantification of plaque stability. Finally, some of the current intravascular imaging studies focus on the changes in PB and composition in the entire studied segment. This approach averages changes in PB in disease-free and atherosclerotic sub-segments, whereas effect of pharmacotherapies may be more intense on vulnerable plaques; therefore, this approach is likely to underestimate their effects on plaque stabilization.4,24 Future studies focusing on plaque changes at lesion-level will allow more representative assessment of therapies on plaque vulnerability and will reduce the cost of studies as they will require recruitment of a smaller number of patients to meet their primary endpoints since high-risk patients have multiple plaques in the coronary tree.11,116

6.2 Implications of pharmacotherapies on local hemodynamic forces

Cumulative data have highlighted the role of endothelial shear stress in vulnerable plaque formation and destabilization showing that low shear stress promotes the formation of high-risk plaques, whereas high shear stress appears to activate mechano-transduction pathways that lead to their destabilization.117,118 The PREDICTION13 and ad hoc analyses of the PROSPECT study119,120 have recently shown that shear stress may be an independent predictor of plaque vulnerability and has a higher accuracy than IVUS or VH-IVUS-derived variables in predicting MACE. Similarly, plaque axial and longitudinal stress also provided useful prognostic information to identify lesions at risk (Figure 3).120,121 Minor changes in PB and composition noted in intravascular imaging studies may have a detrimental effect on shear stress distribution. In the IBIS 4 study,4 high-dose rosuvastatin therapy did not change lumen area at follow-up in ‘segment-level analysis’, but increased lumen area in the 10 mm most diseased segment by +2.5%. This change is expected to affect shear stress distribution and reduce mean shear stress in this segment by approximately -3.6% if there is no change in coronary flow. Likewise, a minor decrease in PB, remodelling index, and a necrotic core burden and especially an increase in the FCT may increase the minimum stress required to cause fibrous cap destabilization and plaque rupture.122 It is essential, therefore, to focus on lesion-level analysis, use multimodality imaging to thoroughly and meticulously assess the effects of novel athero-protective medications on plaque morphology and examine their implications on its physiology that determines plaque evolution and vulnerability.123

Case examples highlighting the prognostic value of the local hemodynamic forces in predicting events. Panel (A) illustrates the angiographic image of a lesion located in the mid left circumflex that caused an event at 13-month follow-up (D). VH-IVUS imaging showed a moderate lesion with a minimum lumen area of 2.94 mm2 (indicated with a white circle on angiography, the corresponding VH-IVUS cross-section is shown in the white inset), a plaque burden of 70.5% and a TCFA phenotype. Blood flow simulation analysis demonstrated high wall shear stress (WSS) at the throat of the lesion with the maximum predominant WSS estimated at 6.94 Pa (B), while plaque structural stress (PSS) analysis also showed increased plaque stress (C) estimated at 123 kPa (maximum PSS location on coronary angiography is indicated with a light blue circle, the corresponding VH-IVUS cross-section is shown in the light blue inset). Panel (E) portrays the angiographic image of a moderate lesion located in the right coronary artery that remained quiescent at 13-month follow-up (H). VH-IVUS imaging showed a plaque with a TCFA phenotype, minimum lumen area of 3.75 mm2 (its location in coronary angiography is shown with a white circle and the VH-IVUS image in the white inset) and similar plaque burden (78.6%) compared to the previous lesion. However, in this occasion, the maximum predominant WSS was normal (3.73 Pa, F), while PSS (the location in angiography is shown with a light blue circle and the corresponding VH-IVUS frame in the light blue inset) was lower (71 kPa, G) suggesting an athero-protective hemodynamic environment that inhibited atherosclerotic disease progression.
Figure 3

Case examples highlighting the prognostic value of the local hemodynamic forces in predicting events. Panel (A) illustrates the angiographic image of a lesion located in the mid left circumflex that caused an event at 13-month follow-up (D). VH-IVUS imaging showed a moderate lesion with a minimum lumen area of 2.94 mm2 (indicated with a white circle on angiography, the corresponding VH-IVUS cross-section is shown in the white inset), a plaque burden of 70.5% and a TCFA phenotype. Blood flow simulation analysis demonstrated high wall shear stress (WSS) at the throat of the lesion with the maximum predominant WSS estimated at 6.94 Pa (B), while plaque structural stress (PSS) analysis also showed increased plaque stress (C) estimated at 123 kPa (maximum PSS location on coronary angiography is indicated with a light blue circle, the corresponding VH-IVUS cross-section is shown in the light blue inset). Panel (E) portrays the angiographic image of a moderate lesion located in the right coronary artery that remained quiescent at 13-month follow-up (H). VH-IVUS imaging showed a plaque with a TCFA phenotype, minimum lumen area of 3.75 mm2 (its location in coronary angiography is shown with a white circle and the VH-IVUS image in the white inset) and similar plaque burden (78.6%) compared to the previous lesion. However, in this occasion, the maximum predominant WSS was normal (3.73 Pa, F), while PSS (the location in angiography is shown with a light blue circle and the corresponding VH-IVUS frame in the light blue inset) was lower (71 kPa, G) suggesting an athero-protective hemodynamic environment that inhibited atherosclerotic disease progression.

6.3 Pleiotropic effects of drugs targeting atherosclerosis

In addition to plaque anatomy and pathophysiology, atherosclerotic evolution also depends on systemic factors such as blood viscosity, platelet activity, fibrinogen levels, and the interplay between coagulation and fibrinolytic system which regulate thrombus formation.124,125 These pathways determine the clinical consequences of plaque rupture as well as the effects of plaque erosion or the eruption of a calcific nodule that constitute common causes of acute coronary events.16 Some of the tested pharmacotherapies have pleiotropic effects affecting plaque evolution and also endothelial function, platelet reactivity, and vascular inflammation. For instance, statins not only increase the number of intimal smooth muscle cells and the expression of type I procollagen, but also decrease the proliferation and activation of macrophages as well as tissue factor expression in animal models.126,127 Similarly, experimental studies have shown that anti-PCKS9 antibodies reduce macrophages infiltration within aortic plaques and increase the endothelial progenitor and circulating angiogenic cells.128 These changes may play a key role in promoting plaque healing following plaque rupture and thus reducing the risk of ACS.129 Finally, lipid-lowering drugs by reducing cholesterol levels seem also to induce collagen synthesis which is important not only for the passivation of high-risk lesions but also for the healing of ruptured or eroded plaques.3,130,131 It is apparent that intravascular imaging studies cannot assess all these pleiotropic effects of novel therapies on plaque biology and their prognostic implications. Therefore, outcome studies should be always considered as the ultimate test for examining the potency of novel drugs.

7. Conclusions

The changes in PB which has been the traditional intravascular imaging endpoint for assessing the efficacy of pharmacotherapies targeting atherosclerosis have a consistent but limited efficacy in predicting their prognostic benefit. While this may reflect the pleiotropic effects of some of these drugs, it also indicates that PB alone does not accurately reflect plaque vulnerability. Future studies evaluating the effects of novel drugs on plaque characteristics are expected to utilize serial multimodality/hybrid imaging to assess more accurately plaque morphology and incorporate physiological endpoints such as shear stress and plaque stress.

These studies are anticipated to enrich our understanding, provide mechanistic insights about the effect of drugs on atherosclerotic evolution, and fully explore the potential of intravascular imaging in predicting their effect on clinical outcomes.

Supplementary material

Supplementary material is available at Cardiovascular Research online.

Funding

V.T. and C.V.B. are funded by the National Institute of Health Research (NIHR) Barts Biomedical Research Centre. M.R.B. is funded by the British Heart Foundation (BHF) grants RG71070 and RG84554, the NIHR Cambridge Biomedical Research Centre, and the Cambridge BHF Centre for Research Excellence [RE/18/1/34212]. S.Z.G. is funded by the BHF Clinical Research Fellowship FS/19/66/34658.

Data availability

The data underlying this article are available in the article and in its online supplementary material.

References

1

Hegele
RA
,
Tsimikas
S
.
Lipid-lowering agents
.
Circ Res
2019
;
124
:
386
404
.

2

Nicholls
SJ
,
Puri
R
,
Anderson
T
,
Ballantyne
CM
,
Cho
L
,
Kastelein
JJ
,
Koenig
W
,
Somaratne
R
,
Kassahun
H
,
Yang
J
,
Wasserman
SM
,
Scott
R
,
Ungi
I
,
Podolec
J
,
Ophuis
AO
,
Cornel
JH
,
Borgman
M
,
Brennan
DM
,
Nissen
SE
.
Effect of evolocumab on progression of coronary disease in statin-treated patients: the glagov randomized clinical trial
.
JAMA
2016
;
316
:
2373
2384
.

3

Raber
L
,
Koskinas
KC
,
Yamaji
K
,
Taniwaki
M
,
Roffi
M
,
Holmvang
L
,
Garcia Garcia
HM
,
Zanchin
T
,
Maldonado
R
,
Moschovitis
A
,
Pedrazzini
G
,
Zaugg
S
,
Dijkstra
J
,
Matter
CM
,
Serruys
PW
,
Luscher
TF
,
Kelbaek
H
,
Karagiannis
A
,
Radu
MD
,
Windecker
S
.
Changes in coronary plaque composition in patients with acute myocardial infarction treated with high-intensity statin therapy (IBIS-4): a serial optical coherence tomography study
.
JACC Cardiovasc Imaging
2019
;
12
:
1518
1528
.

4

Raber
L
,
Taniwaki
M
,
Zaugg
S
,
Kelbaek
H
,
Roffi
M
,
Holmvang
L
,
Noble
S
,
Pedrazzini
G
,
Moschovitis
A
,
Luscher
TF
,
Matter
CM
,
Serruys
PW
,
Juni
P
,
Garcia-Garcia
HM
,
Windecker
S
,
Investigators
IT
.
Effect of high-intensity statin therapy on atherosclerosis in non-infarct-related coronary arteries (IBIS-4): a serial intravascular ultrasonography study
.
Eur Heart J
2015
;
36
:
490
500
.

5

Bose
D
,
von Birgelen
C
,
Erbel
R
.
Intravascular ultrasound for the evaluation of therapies targeting coronary atherosclerosis
.
J Am Coll Cardiol
2007
;
49
:
925
932
.

6

Nicholls
SJ
,
Hsu
A
,
Wolski
K
,
Hu
B
,
Bayturan
O
,
Lavoie
A
,
Uno
K
,
Tuzcu
EM
,
Nissen
SE
.
Intravascular ultrasound-derived measures of coronary atherosclerotic plaque burden and clinical outcome
.
J Am Coll Cardiol
2010
;
55
:
2399
2407
.

7

Cheng
JM
,
Garcia-Garcia
HM
,
de Boer
SP
,
Kardys
I
,
Heo
JH
,
Akkerhuis
KM
,
Oemrawsingh
RM
,
van Domburg
RT
,
Ligthart
J
,
Witberg
KT
,
Regar
E
,
Serruys
PW
,
van Geuns
RJ
,
Boersma
E
.
In vivo detection of high-risk coronary plaques by radiofrequency intravascular ultrasound and cardiovascular outcome: results of the ATHEROREMO-IVUS study
.
Eur Heart J
2014
;
35
:
639
647
.

8

Oemrawsingh
RM
,
Cheng
JM
,
Garcia-Garcia
HM
,
van Geuns
RJ
,
de Boer
SP
,
Simsek
C
,
Kardys
I
,
Lenzen
MJ
,
van Domburg
RT
,
Regar
E
,
Serruys
PW
,
Akkerhuis
KM
,
Boersma
E
,
Investigators
A-N
.
Near-infrared spectroscopy predicts cardiovascular outcome in patients with coronary artery disease
.
J Am Coll Cardiol
2014
;
64
:
2510
2518
.

9

Waksman
R
,
Di Mario
C
,
Torguson
R
,
Ali
ZA
,
Singh
V
,
Skinner
WH
,
Artis
AK
,
Cate
TT
,
Powers
E
,
Kim
C
,
Regar
E
,
Wong
SC
,
Lewis
S
,
Wykrzykowska
J
,
Dube
S
,
Kazziha
S
,
van der Ent
M
,
Shah
P
,
Craig
PE
,
Zou
Q
,
Kolm
P
,
Brewer
HB
,
Garcia-Garcia
HM
,
Samady
H
,
Tobis
J
,
Zainea
M
,
Leimbach
W
,
Lee
D
,
Lalonde
T
,
Skinner
W
,
Villa
A
,
Liberman
H
,
Younis
G
,
de Silva
R
,
Diaz
M
,
Tami
L
,
Hodgson
J
,
Raveendran
G
,
Goswami
N
,
Arias
J
,
Lovitz
L
,
Carida
R
II
,
Potluri
S
,
Prati
F
,
Erglis
A
,
Pop
A
,
McEntegart
M
,
Hudec
M
,
Rangasetty
U
,
Newby
D
,
Investigators LRP
.
Identification of patients and plaques vulnerable to future coronary events with near-infrared spectroscopy intravascular ultrasound imaging: a prospective, cohort study
.
Lancet
2019
;
394
:
1629
1637
.

10

Erlinge
D
,
Maehara
A
,
Ben-Yehuda
O
,
Botker
HE
,
Maeng
M
,
Kjoller-Hansen
L
,
Engstrom
T
,
Matsumura
M
,
Crowley
A
,
Dressler
O
,
Mintz
GS
,
Frobert
O
,
Persson
J
,
Wiseth
R
,
Larsen
AI
,
Okkels Jensen
L
,
Nordrehaug
JE
,
Bleie
O
,
Omerovic
E
,
Held
C
,
James
SK
,
Ali
ZA
,
Muller
JE
,
Stone
GW
,
Ahlehoff
O
,
Amin
A
,
Angerås
O
,
Appikonda
P
,
Balachandran
S
,
Barvik
S
,
Bendix
K
,
Bertilsson
M
,
Boden
U
,
Bogale
N
,
Bonarjee
V
,
Calais
F
,
Carlsson
J
,
Carstensen
S
,
Christersson
C
,
Christiansen
EH
,
Corral
M
,
De Backer
O
,
Dhaha
U
,
Dworeck
C
,
Eggers
K
,
Elfström
C
,
Ellert
J
,
Eriksen
E
,
Fallesen
C
,
Forsman
M
,
Fransson
H
,
Gaballa
M
,
Gacki
M
,
Götberg
M
,
Hagström
L
,
Hallberg
T
,
Hambraeus
K
,
Haraldsson
I
,
Harnek
J
,
Havndrup
O
,
Hegbom
K
,
Heigert
M
,
Helqvist
S
,
Herstad
J
,
Hijazi
Z
,
Holmvang
L
,
Ioanes
D
,
Iqbal
A
,
Iversen
A
,
Jacobson
J
,
Jakobsen
L
,
Jankovic
I
,
Jensen
U
,
Jensevik
K
,
Johnston
N
,
Jonasson
TF
,
Jørgensen
E
,
Joshi
F
,
Kajermo
U
,
Kåver
F
,
Kelbæk
H
,
Kellerth
T
,
Kish
M
,
Koenig
W
,
Koul
S
,
Lagerqvist
B
,
Larsson
B
,
Lassen
JF
,
Leiren
O
,
Li
Z
,
Lidell
C
,
Linder
R
,
Lindstaedt
M
,
Lindström
G
,
Liu
S
,
Løland
KH
,
Lønborg
J
,
Márton
L
,
Mir-Akbari
H
,
Mohamed
S
,
Odenstedt
J
,
Ogne
C
,
Oldgren
J
,
Olivecrona
G
,
Östlund-Papadogeorgos
N
,
Ottesen
M
,
Packer
E
,
Palmquist
ÅM
,
Paracha
Q
,
Pedersen
F
,
Petursson
P
,
Råmunddal
T
,
Rotevatn
S
,
Sanchez
R
,
Sarno
G
,
Saunamäki
KI
,
Scherstén
F
,
Serruys
PW
,
Sjögren
I
,
Sørensen
R
,
Srdanovic
I
,
Subhani
Z
,
Svensson
E
,
Thuesen
A
,
Tijssen
J
,
Tilsted
H-H
,
Tödt
T
,
Trovik
T
,
Våga
BI
,
Varenhorst
C
,
Veien
K
,
Vestman
E
,
Völz
S
,
Wallentin
L
,
Wykrzykowska
J
,
Zagozdzon
L
,
Zamfir
M
,
Zedigh
C
,
Zhong
H
,
Zhou
Z
,
Investigators PROSPECT II
.
Identification of vulnerable plaques and patients by intracoronary near-infrared spectroscopy and ultrasound (PROSPECT II): a prospective natural history study
.
Lancet
2021
;
397
:
985
995
.

11

Stone
GW
,
Maehara
A
,
Lansky
AJ
,
de Bruyne
B
,
Cristea
E
,
Mintz
GS
,
Mehran
R
,
McPherson
J
,
Farhat
N
,
Marso
SP
,
Parise
H
,
Templin
B
,
White
R
,
Zhang
Z
,
Serruys
PW
,
Investigators PROSPECT
.
A prospective natural-history study of coronary atherosclerosis
.
N Engl J Med
2011
;
364
:
226
235
.

12

Calvert
PA
,
Obaid
DR
,
O'Sullivan
M
,
Shapiro
LM
,
McNab
D
,
Densem
CG
,
Schofield
PM
,
Braganza
D
,
Clarke
SC
,
Ray
KK
,
West
NE
,
Bennett
MR
.
Association between IVUS findings and adverse outcomes in patients with coronary artery disease
.
JACC Cardiovasc Imaging
2011
;
4
:
894
901
.

13

Stone
PH
,
Saito
S
,
Takahashi
S
,
Makita
Y
,
Nakamura
S
,
Kawasaki
T
,
Takahashi
A
,
Katsuki
T
,
Nakamura
S
,
Namiki
A
,
Hirohata
A
,
Matsumura
T
,
Yamazaki
S
,
Yokoi
H
,
Tanaka
S
,
Otsuji
S
,
Yoshimachi
F
,
Honye
J
,
Harwood
D
,
Reitman
M
,
Coskun
AU
,
Papafaklis
MI
,
Feldman
CL
,
Investigators
P
.
Prediction of progression of coronary artery disease and clinical outcomes using vascular profiling of endothelial shear stress and arterial plaque characteristics: the PREDICTION Study
.
Circulation
2012
;
126
:
172
181
.

14

Prati
F
,
Romagnoli
E
,
Gatto
L
,
La Manna
A
,
Burzotta
F
,
Ozaki
Y
,
Marco
V
,
Boi
A
,
Fineschi
M
,
Fabbiocchi
F
,
Taglieri
N
,
Niccoli
G
,
Trani
C
,
Versaci
F
,
Calligaris
G
,
Ruscica
G
,
Di Giorgio
A
,
Vergallo
R
,
Albertucci
M
,
Biondi-Zoccai
G
,
Tamburino
C
,
Crea
F
,
Alfonso
F
,
Arbustini
E
.
Relationship between coronary plaque morphology of the left anterior descending artery and 12 months clinical outcome: the CLIMA study
.
Eur Heart J
2020
;
41
:
383
391
.

15

Abela
GS
,
Aziz
K
,
Vedre
A
,
Pathak
DR
,
Talbott
JD
,
Dejong
J
.
Effect of cholesterol crystals on plaques and intima in arteries of patients with acute coronary and cerebrovascular syndromes
.
Am J Cardiol
2009
;
103
:
959
968
.

16

Burke
AP
,
Farb
A
,
Malcom
GT
,
Liang
YH
,
Smialek
J
,
Virmani
R
.
Coronary risk factors and plaque morphology in men with coronary disease who died suddenly
.
N Engl J Med
1997
;
336
:
1276
1282
.

17

Naghavi
M
,
Libby
P
,
Falk
E
,
Casscells
SW
,
Litovsky
S
,
Rumberger
J
,
Badimon
JJ
,
Stefanadis
C
,
Moreno
P
,
Pasterkamp
G
,
Fayad
Z
,
Stone
PH
,
Waxman
S
,
Raggi
P
,
Madjid
M
,
Zarrabi
A
,
Burke
A
,
Yuan
C
,
Fitzgerald
PJ
,
Siscovick
DS
,
de Korte
CL
,
Aikawa
M
,
Juhani Airaksinen
KE
,
Assmann
G
,
Becker
CR
,
Chesebro
JH
,
Farb
A
,
Galis
ZS
,
Jackson
C
,
Jang
IK
,
Koenig
W
,
Lodder
RA
,
March
K
,
Demirovic
J
,
Navab
M
,
Priori
SG
,
Rekhter
MD
,
Bahr
R
,
Grundy
SM
,
Mehran
R
,
Colombo
A
,
Boerwinkle
E
,
Ballantyne
C
,
Insull
W
Jr
,
Schwartz
RS
,
Vogel
R
,
Serruys
PW
,
Hansson
GK
,
Faxon
DP
,
Kaul
S
,
Drexler
H
,
Greenland
P
,
Muller
JE
,
Virmani
R
,
Ridker
PM
,
Zipes
DP
,
Shah
PK
,
Willerson
JT
.
From vulnerable plaque to vulnerable patient: a call for new definitions and risk assessment strategies: part I
.
Circulation
2003
;
108
:
1664
1672
.

18

Almeida
SO
,
Budoff
M
.
Effect of statins on atherosclerotic plaque
.
Trends Cardiovasc Med
2019
;
29
:
451
455
.

19

Cholesterol Treatment Trialists Collaboration
,
Baigent
C
,
Blackwell
L
,
Emberson
J
,
Holland
LE
,
Reith
C
,
Bhala
N
,
Peto
R
,
Barnes
EH
,
Keech
A
,
Simes
J
,
Collins
R
.
Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials
.
Lancet
2010
;
376
:
1670
1681
.

20

Nissen
SE
,
Nicholls
SJ
,
Sipahi
I
,
Libby
P
,
Raichlen
JS
,
Ballantyne
CM
,
Davignon
J
,
Erbel
R
,
Fruchart
JC
,
Tardif
JC
,
Schoenhagen
P
,
Crowe
T
,
Cain
V
,
Wolski
K
,
Goormastic
M
,
Tuzcu
EM
,
Investigators ASTEROID
.
Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial
.
JAMA
2006
;
295
:
1556
1565
.

21

Nicholls
SJ
,
Ballantyne
CM
,
Barter
PJ
,
Chapman
MJ
,
Erbel
RM
,
Libby
P
,
Raichlen
JS
,
Uno
K
,
Borgman
M
,
Wolski
K
,
Nissen
SE
.
Effect of two intensive statin regimens on progression of coronary disease
.
N Engl J Med
2011
;
365
:
2078
2087
.

22

Puri
R
,
Libby
P
,
Nissen
SE
,
Wolski
K
,
Ballantyne
CM
,
Barter
PJ
,
Chapman
MJ
,
Erbel
R
,
Raichlen
JS
,
Uno
K
,
Kataoka
Y
,
Tuzcu
EM
,
Nicholls
SJ
.
Long-term effects of maximally intensive statin therapy on changes in coronary atheroma composition: insights from SATURN
.
Eur Heart J Cardiovasc Imaging
2014
;
15
:
380
388
.

23

Oemrawsingh
RM
,
Garcia-Garcia
HM
,
van Geuns
RJ
,
Lenzen
MJ
,
Simsek
C
,
de Boer
SP
,
Van Mieghem
NM
,
Regar
E
,
de Jaegere
PP
,
Akkerhuis
KM
,
Ligthart
JM
,
Zijlstra
F
,
Serruys
PW
,
Boersma
E
.
Integrated Biomarker and Imaging Study 3 (IBIS-3) to assess the ability of rosuvastatin to decrease necrotic core in coronary arteries
.
EuroIntervention
2016
;
12
:
734
739
.

24

Kini
AS
,
Baber
U
,
Kovacic
JC
,
Limaye
A
,
Ali
ZA
,
Sweeny
J
,
Maehara
A
,
Mehran
R
,
Dangas
G
,
Mintz
GS
,
Fuster
V
,
Narula
J
,
Sharma
SK
,
Moreno
PR
.
Changes in plaque lipid content after short-term intensive versus standard statin therapy: the YELLOW trial (reduction in yellow plaque by aggressive lipid-lowering therapy)
.
J Am Coll Cardiol
2013
;
62
:
21
29
.

25

Kini
AS
,
Vengrenyuk
Y
,
Shameer
K
,
Maehara
A
,
Purushothaman
M
,
Yoshimura
T
,
Matsumura
M
,
Aquino
M
,
Haider
N
,
Johnson
KW
,
Readhead
B
,
Kidd
BA
,
Feig
JE
,
Krishnan
P
,
Sweeny
J
,
Milind
M
,
Moreno
P
,
Mehran
R
,
Kovacic
JC
,
Baber
U
,
Dudley
JT
,
Narula
J
,
Sharma
S
.
Intracoronary imaging, cholesterol efflux, and transcriptomes after intensive statin treatment: the YELLOW II study
.
J Am Coll Cardiol
2017
;
69
:
628
640
.

26

Park
SJ
,
Kang
SJ
,
Ahn
JM
,
Chang
M
,
Yun
SC
,
Roh
JH
,
Lee
PH
,
Park
HW
,
Yoon
SH
,
Park
DW
,
Lee
SW
,
Kim
YH
,
Lee
CW
,
Mintz
GS
,
Han
KH
,
Park
SW
.
Effect of statin treatment on modifying plaque composition: a double-blind, randomized study
.
J Am Coll Cardiol
2016
;
67
:
1772
1783
.

27

Kjekshus
J
,
Apetrei
E
,
Barrios
V
,
Bohm
M
,
Cleland
JG
,
Cornel
JH
,
Dunselman
P
,
Fonseca
C
,
Goudev
A
,
Grande
P
,
Gullestad
L
,
Hjalmarson
A
,
Hradec
J
,
Janosi
A
,
Kamensky
G
,
Komajda
M
,
Korewicki
J
,
Kuusi
T
,
Mach
F
,
Mareev
V
,
McMurray
JJ
,
Ranjith
N
,
Schaufelberger
M
,
Vanhaecke
J
,
van Veldhuisen
DJ
,
Waagstein
F
,
Wedel
H
,
Wikstrand
J
,
Group C
.
Rosuvastatin in older patients with systolic heart failure
.
N Engl J Med
2007
;
357
:
2248
2261
.

28

Tavazzi
L
,
Maggioni
AP
,
Marchioli
R
,
Barlera
S
,
Franzosi
MG
,
Latini
R
,
Lucci
D
,
Nicolosi
GL
,
Porcu
M
,
Tognoni
G
,
GISSI-HF Investigators
.
Effect of rosuvastatin in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial
.
Lancet
2008
;
372
:
1231
1239
.

29

Feinstein
MJ
,
Jhund
P
,
Kang
J
,
Ning
H
,
Maggioni
A
,
Wikstrand
J
,
Kjekshus
J
,
Tavazzi
L
,
McMurray
J
,
Lloyd-Jones
DM
.
Do statins reduce the risk of myocardial infarction in patients with heart failure? A pooled individual-level reanalysis of CORONA and GISSI-HF
.
Eur J Heart Fail
2015
;
17
:
434
441
.

30

Nissen
SE
,
Tuzcu
EM
,
Schoenhagen
P
,
Brown
BG
,
Ganz
P
,
Vogel
RA
,
Crowe
T
,
Howard
G
,
Cooper
CJ
,
Brodie
B
,
Grines
CL
,
DeMaria
AN
,
REVERSAL Investigators
.
Effect of intensive compared with moderate lipid-lowering therapy on progression of coronary atherosclerosis: a randomized controlled trial
.
JAMA
2004
;
291
:
1071
1080
.

31

Hiro
T
,
Kimura
T
,
Morimoto
T
,
Miyauchi
K
,
Nakagawa
Y
,
Yamagishi
M
,
Ozaki
Y
,
Kimura
K
,
Saito
S
,
Yamaguchi
T
,
Daida
H
,
Matsuzaki
M
,
Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 Investigators
.
Effect of intensive statin therapy on regression of coronary atherosclerosis in patients with acute coronary syndrome: a multicenter randomized trial evaluated by volumetric intravascular ultrasound using pitavastatin versus atorvastatin (JAPAN-ACS [Japan assessment of pitavastatin and atorvastatin in acute coronary syndrome] study)
.
J Am Coll Cardiol
2009
;
54
:
293
302
.

32

Lee
CW
,
Kang
SJ
,
Ahn
JM
,
Song
HG
,
Lee
JY
,
Kim
WJ
,
Park
DW
,
Lee
SW
,
Kim
YH
,
Park
SW
,
Park
SJ
.
Comparison of effects of atorvastatin (20 mg) versus rosuvastatin (10 mg) therapy on mild coronary atherosclerotic plaques (from the ARTMAP trial)
.
Am J Cardiol
2012
;
109
:
1700
1704
.

33

Komukai
K
,
Kubo
T
,
Kitabata
H
,
Matsuo
Y
,
Ozaki
Y
,
Takarada
S
,
Okumoto
Y
,
Shiono
Y
,
Orii
M
,
Shimamura
K
,
Ueno
S
,
Yamano
T
,
Tanimoto
T
,
Ino
Y
,
Yamaguchi
T
,
Kumiko
H
,
Tanaka
A
,
Imanishi
T
,
Akagi
H
,
Akasaka
T
.
Effect of atorvastatin therapy on fibrous cap thickness in coronary atherosclerotic plaque as assessed by optical coherence tomography: the EASY-FIT study
.
J Am Coll Cardiol
2014
;
64
:
2207
2217
.

34

Cannon
CP
,
Braunwald
E
,
McCabe
CH
,
Rader
DJ
,
Rouleau
JL
,
Belder
R
,
Joyal
SV
,
Hill
KA
,
Pfeffer
MA
,
Skene
AM
,
Pravastatin or Atorvastatin Evaluation
,
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
:
1495
1504
.

35

LaRosa
JC
,
Grundy
SM
,
Waters
DD
,
Shear
C
,
Barter
P
,
Fruchart
JC
,
Gotto
AM
,
Greten
H
,
Kastelein
JJ
,
Shepherd
J
,
Wenger
NK
,
Treating to New Targets (TNT) Investigators
.
Intensive lipid lowering with atorvastatin in patients with stable coronary disease
.
N Engl J Med
2005
;
352
:
1425
1435
.

36

Nozue
T
,
Yamamoto
S
,
Tohyama
S
,
Umezawa
S
,
Kunishima
T
,
Sato
A
,
Miyake
S
,
Takeyama
Y
,
Morino
Y
,
Yamauchi
T
,
Muramatsu
T
,
Hibi
K
,
Sozu
T
,
Terashima
M
,
Michishita
I
.
Statin treatment for coronary artery plaque composition based on intravascular ultrasound radiofrequency data analysis
.
Am Heart J
2012
;
163
:
191
199.e1
.

37

Sacks
FM
,
Pfeffer
MA
,
Moye
LA
,
Rouleau
JL
,
Rutherford
JD
,
Cole
TG
,
Brown
L
,
Warnica
JW
,
Arnold
JM
,
Wun
CC
,
Davis
BR
,
Braunwald
E
.
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
:
1001
1009
.

38

Long-Term Intervention with Pravastatin in Ischaemic Disease Study Group
.
Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels
.
N Engl J Med
1998
;
339
:
1349
1357
.

39

Niedzielski
M
,
Broncel
M
,
Gorzelak-Pabis
P
,
Wozniak
E
.
New possible pharmacological targets for statins and ezetimibe
.
Biomed Pharmacother
2020
;
129
:
110388
.

40

Sudhop
T
,
Lutjohann
D
,
Kodal
A
,
Igel
M
,
Tribble
DL
,
Shah
S
,
Perevozskaya
I
,
von Bergmann
K
.
Inhibition of intestinal cholesterol absorption by ezetimibe in humans
.
Circulation
2002
;
106
:
1943
1948
.

41

Mach
F
,
Baigent
C
,
Catapano
AL
,
Koskinas
KC
,
Casula
M
,
Badimon
L
,
Chapman
MJ
,
De Backer
GG
,
Delgado
V
,
Ference
BA
,
Graham
IM
,
Halliday
A
,
Landmesser
U
,
Mihaylova
B
,
Pedersen
TR
,
Riccardi
G
,
Richter
DJ
,
Sabatine
MS
,
Taskinen
MR
,
Tokgozoglu
L
,
Wiklund
O
,
Mueller
C
,
Drexel
H
,
Aboyans
V
,
Corsini
A
,
Doehner
W
,
Farnier
M
,
Gigante
B
,
Kayikcioglu
M
,
Krstacic
G
,
Lambrinou
E
,
Lewis
BS
,
Masip
J
,
Moulin
P
,
Petersen
S
,
Petronio
AS
,
Piepoli
MF
,
Pintó
X
,
Räber
L
,
Ray
KK
,
Reiner
Ž
,
Riesen
WF
,
Roffi
M
,
Schmid
J-P
,
Shlyakhto
E
,
Simpson
IA
,
Stroes
E
,
Sudano
I
,
Tselepis
AD
,
Viigimaa
M
,
Vindis
C
,
Vonbank
A
,
Vrablik
M
,
Vrsalovic
M
,
Zamorano
JL
,
Collet
J-P
,
Koskinas
KC
,
Casula
M
,
Badimon
L
,
John Chapman
M
,
De Backer
GG
,
Delgado
V
,
Ference
BA
,
Graham
IM
,
Halliday
A
,
Landmesser
U
,
Mihaylova
B
,
Pedersen
TR
,
Riccardi
G
,
Richter
DJ
,
Sabatine
MS
,
Taskinen
M-R
,
Tokgozoglu
L
,
Wiklund
O
,
Windecker
S
,
Aboyans
V
,
Baigent
C
,
Collet
J-P
,
Dean
V
,
Delgado
V
,
Fitzsimons
D
,
Gale
CP
,
Grobbee
D
,
Halvorsen
S
,
Hindricks
G
,
Iung
B
,
Jüni
P
,
Katus
HA
,
Landmesser
U
,
Leclercq
C
,
Lettino
M
,
Lewis
BS
,
Merkely
B
,
Mueller
C
,
Petersen
S
,
Petronio
AS
,
Richter
DJ
,
Roffi
M
,
Shlyakhto
E
,
Simpson
IA
,
Sousa-Uva
M
,
Touyz
RM
,
Nibouche
D
,
Zelveian
PH
,
Siostrzonek
P
,
Najafov
R
,
van de Borne
P
,
Pojskic
B
,
Postadzhiyan
A
,
Kypris
L
,
Špinar
J
,
Larsen
ML
,
Eldin
HS
,
Viigimaa
M
,
Strandberg
TE
,
Ferrières
J
,
Agladze
R
,
Laufs
U
,
Rallidis
L
,
Bajnok
L
,
Gudjónsson
T
,
Maher
V
,
Henkin
Y
,
Gulizia
MM
,
Mussagaliyeva
A
,
Bajraktari
G
,
Kerimkulova
A
,
Latkovskis
G
,
Hamoui
O
,
Slapikas
R
,
Visser
L
,
Dingli
P
,
Ivanov
V
,
Boskovic
A
,
Nazzi
M
,
Visseren
F
,
Mitevska
I
,
Retterstøl
K
,
Jankowski
P
,
Fontes-Carvalho
R
,
Gaita
D
,
Ezhov
M
,
Foscoli
M
,
Giga
V
,
Pella
D
,
Fras
Z
,
de Isla
LP
,
Hagström
E
,
Lehmann
R
,
Abid
L
,
Ozdogan
O
,
Mitchenko
O
,
Patel
RS
,
Group ESCSD
.
2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk
.
Eur Heart J
2020
;
41
:
111
188
.

42

Tsujita
K
,
Sugiyama
S
,
Sumida
H
,
Shimomura
H
,
Yamashita
T
,
Yamanaga
K
,
Komura
N
,
Sakamoto
K
,
Oka
H
,
Nakao
K
,
Nakamura
S
,
Ishihara
M
,
Matsui
K
,
Sakaino
N
,
Nakamura
N
,
Yamamoto
N
,
Koide
S
,
Matsumura
T
,
Fujimoto
K
,
Tsunoda
R
,
Morikami
Y
,
Matsuyama
K
,
Oshima
S
,
Kaikita
K
,
Hokimoto
S
,
Ogawa
H
,
PRECISE–IVUS Investigators
.
Impact of dual lipid-lowering strategy with ezetimibe and atorvastatin on coronary plaque regression in patients with percutaneous coronary intervention
.
J Am Coll Cardiol
2015
;
66
:
495
507
.

43

Mirzaee
S
,
Thein
PM
,
Nogic
J
,
Nerlekar
N
,
Nasis
A
,
Brown
AJ
.
The effect of combined ezetimibe and statin therapy versus statin therapy alone on coronary plaque volume assessed by intravascular ultrasound: a systematic review and meta-analysis
.
J Clin Lipidol
2018
;
12
:
1133
1140.e15
.

44

Cannon
CP
,
Blazing
MA
,
Giugliano
RP
,
McCagg
A
,
White
JA
,
Theroux
P
,
Darius
H
,
Lewis
BS
,
Ophuis
TO
,
Jukema
JW
,
De Ferrari
GM
,
Ruzyllo
W
,
De Lucca
P
,
Im
K
,
Bohula
EA
,
Reist
C
,
Wiviott
SD
,
Tershakovec
AM
,
Musliner
TA
,
Braunwald
E
,
Califf
RM
,
IMPROVE-IT Investigators
.
Ezetimibe added to statin therapy after acute coronary syndromes
.
N Engl J Med
2015
;
372
:
2387
2397
.

45

Hagiwara
N
,
Kawada-Watanabe
E
,
Koyanagi
R
,
Arashi
H
,
Yamaguchi
J
,
Nakao
K
,
Tobaru
T
,
Tanaka
H
,
Oka
T
,
Endoh
Y
,
Saito
K
,
Uchida
T
,
Matsui
K
,
Ogawa
H
.
Low-density lipoprotein cholesterol targeting with pitavastatin + ezetimibe for patients with acute coronary syndrome and dyslipidaemia: the HIJ-PROPER study, a prospective, open-label, randomized trial
.
Eur Heart J
2017
;
38
:
2264
2276
.

46

Robinson
JG
,
Farnier
M
,
Krempf
M
,
Bergeron
J
,
Luc
G
,
Averna
M
,
Stroes
ES
,
Langslet
G
,
Raal
FJ
,
El Shahawy
M
,
Koren
MJ
,
Lepor
NE
,
Lorenzato
C
,
Pordy
R
,
Chaudhari
U
,
Kastelein
JJ
,
ODYSSEY LONG TERM Investigators
.
Efficacy and safety of alirocumab in reducing lipids and cardiovascular events
.
N Engl J Med
2015
;
372
:
1489
1499
.

47

Sabatine
MS
,
Giugliano
RP
,
Wiviott
SD
,
Raal
FJ
,
Blom
DJ
,
Robinson
J
,
Ballantyne
CM
,
Somaratne
R
,
Legg
J
,
Wasserman
SM
,
Scott
R
,
Koren
MJ
,
Stein
EA
.
Open-label study of long-term evaluation against LDLCI. Efficacy and safety of evolocumab in reducing lipids and cardiovascular events
.
N Engl J Med
2015
;
372
:
1500
1509
.

48

Ragusa
R
,
Basta
G
,
Neglia
D
,
De Caterina
R
,
Del Turco
S
,
Caselli
C
.
PCSK9 and atherosclerosis: looking beyond LDL regulation
.
Eur J Clin Invest
2021
;
51
:
e13459
.

49

Nicholls
SJ
,
Puri
R
,
Anderson
T
,
Ballantyne
CM
,
Cho
L
,
Kastelein
JJP
,
Koenig
W
,
Somaratne
R
,
Kassahun
H
,
Yang
J
,
Wasserman
SM
,
Honda
S
,
Shishikura
D
,
Scherer
DJ
,
Borgman
M
,
Brennan
DM
,
Wolski
K
,
Nissen
SE
.
Effect of evolocumab on coronary plaque composition
.
J Am Coll Cardiol
2018
;
72
:
2012
2021
.

50

Ako
J
,
Hibi
K
,
Tsujita
K
,
Hiro
T
,
Morino
Y
,
Kozuma
K
,
Shinke
T
,
Otake
H
,
Uno
K
,
Louie
MJ
,
Takagi
Y
,
Miyauchi
K
.
Effect of alirocumab on coronary atheroma volume in japanese patients with acute coronary syndrome- the ODYSSEY J-IVUS trial
.
Circ J
2019
;
83
:
2025
2033
.

51

Sugizaki
Y
,
Otake
H
,
Kawamori
H
,
Toba
T
,
Nagano
Y
,
Tsukiyama
Y
,
Yanaka
KI
,
Yamamoto
H
,
Nagasawa
A
,
Onishi
H
,
Takeshige
R
,
Nakano
S
,
Matsuoka
Y
,
Tanimura
K
,
Takahashi
Y
,
Fukuyama
Y
,
Shinke
T
,
Ishida
T
,
Hirata
KI
.
Adding alirocumab to rosuvastatin helps reduce the vulnerability of thin-cap fibroatheroma: an ALTAIR trial report
.
JACC Cardiovasc Imaging
2020
;
13
:
1452
1454
.

52

Ota
H
,
Omori
H
,
Kawasaki
M
,
Hirakawa
A
,
Matsuo
H
.
Clinical impact of PCSK9 inhibitor on stabilization and regression of lipid-rich coronary plaques: a near-infrared spectroscopy study
.
Eur Heart J Cardiovasc Imaging
2021
;
2
:
217
228
.

53

Nicholls
SJ
,
Nissen
SE
,
Prati
F
,
Windecker
S
,
Kataoka
Y
,
Puri
R
,
Hucko
T
,
Kassahun
H
,
Liao
J
,
Somaratne
R
,
Butters
J
,
Di Giovanni
G
,
Jones
S
,
Psaltis
PJ
.
Assessing the impact of PCSK9 inhibition on coronary plaque phenotype with optical coherence tomography: rationale and design of the randomized, placebo-controlled HUYGENS study
.
Cardiovasc Diagn Ther
2021
;
11
:
120
129
.

54

Zanchin
C
,
Koskinas
KC
,
Ueki
Y
,
Losdat
S
,
Haner
JD
,
Bar
S
,
Otsuka
T
,
Inderkum
A
,
Jensen
MRJ
,
Lonborg
J
,
Fahrni
G
,
Ondracek
AS
,
Daemen
J
,
van Geuns
RJ
,
Iglesias
JF
,
Matter
CM
,
Spirk
D
,
Juni
P
,
Mach
F
,
Heg
D
,
Engstrom
T
,
Lang
I
,
Windecker
S
,
Raber
L
.
Effects of the PCSK9 antibody alirocumab on coronary atherosclerosis in patients with acute myocardial infarction: a serial, multivessel, intravascular ultrasound, near-infrared spectroscopy and optical coherence tomography imaging study-Rationale and design of the PACMAN-AMI trial
.
Am Heart J
2021
;
238
:
33
44
.

55

Sabatine
MS
,
Giugliano
RP
,
Keech
AC
,
Honarpour
N
,
Wiviott
SD
,
Murphy
SA
,
Kuder
JF
,
Wang
H
,
Liu
T
,
Wasserman
SM
,
Sever
PS
,
Pedersen
TR
,
FOURIER Steering Committee and Investigators
.
Evolocumab and clinical outcomes in patients with cardiovascular disease
.
N Engl J Med
2017
;
376
:
1713
1722
.

56

Schwartz
GG
,
Steg
PG
,
Szarek
M
,
Bhatt
DL
,
Bittner
VA
,
Diaz
R
,
Edelberg
JM
,
Goodman
SG
,
Hanotin
C
,
Harrington
RA
,
Jukema
JW
,
Lecorps
G
,
Mahaffey
KW
,
Moryusef
A
,
Pordy
R
,
Quintero
K
,
Roe
MT
,
Sasiela
WJ
,
Tamby
JF
,
Tricoci
P
,
White
HD
,
Zeiher
AM
,
ODYSSEY OUTCOMES Committees and Investigators
.
Alirocumab and cardiovascular outcomes after acute coronary syndrome
.
N Engl J Med
2018
;
379
:
2097
2107
.

57

Tardy
C
,
Goffinet
M
,
Boubekeur
N
,
Ackermann
R
,
Sy
G
,
Bluteau
A
,
Cholez
G
,
Keyserling
C
,
Lalwani
N
,
Paolini
JF
,
Dasseux
JL
,
Barbaras
R
,
Baron
R
.
CER-001, a HDL-mimetic, stimulates the reverse lipid transport and atherosclerosis regression in high cholesterol diet-fed LDL-receptor deficient mice
.
Atherosclerosis
2014
;
232
:
110
118
.

58

Badimon
JJ
,
Badimon
L
,
Galvez
A
,
Dische
R
,
Fuster
V
.
High density lipoprotein plasma fractions inhibit aortic fatty streaks in cholesterol-fed rabbits
.
Lab Invest
1989
;
60
:
455
461
.

59

Nicholls
SJ
,
Cutri
B
,
Worthley
SG
,
Kee
P
,
Rye
KA
,
Bao
S
,
Barter
PJ
.
Impact of short-term administration of high-density lipoproteins and atorvastatin on atherosclerosis in rabbits
.
Arterioscler Thromb Vasc Biol
2005
;
25
:
2416
2421
.

60

Nissen
SE
,
Tsunoda
T
,
Tuzcu
EM
,
Schoenhagen
P
,
Cooper
CJ
,
Yasin
M
,
Eaton
GM
,
Lauer
MA
,
Sheldon
WS
,
Grines
CL
,
Halpern
S
,
Crowe
T
,
Blankenship
JC
,
Kerensky
R
.
Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial
.
JAMA
2003
;
290
:
2292
2300
.

61

Nicholls
SJ
,
Andrews
J
,
Kastelein
JJP
,
Merkely
B
,
Nissen
SE
,
Ray
KK
,
Schwartz
GG
,
Worthley
SG
,
Keyserling
C
,
Dasseux
JL
,
Griffith
L
,
Kim
SW
,
Janssan
A
,
Di Giovanni
G
,
Pisaniello
AD
,
Scherer
DJ
,
Psaltis
PJ
,
Butters
J
.
Effect of serial infusions of CER-001, a pre-beta high-density lipoprotein mimetic, on coronary atherosclerosis in patients following acute coronary syndromes in the CER-001 atherosclerosis regression acute coronary syndrome trial: a randomized clinical trial
.
JAMA Cardiol
2018
;
3
:
815
822
.

62

Nicholls
SJ
,
Puri
R
,
Ballantyne
CM
,
Jukema
JW
,
Kastelein
JJP
,
Koenig
W
,
Wright
RS
,
Kallend
D
,
Wijngaard
P
,
Borgman
M
,
Wolski
K
,
Nissen
SE
.
Effect of infusion of high-density lipoprotein mimetic containing recombinant apolipoprotein A-I milano on coronary disease in patients with an acute coronary syndrome in the MILANO-PILOT trial: a randomized clinical trial
.
JAMA Cardiol
2018
;
3
:
806
814
.

63

Tardif
JC
,
Ballantyne
CM
,
Barter
P
,
Dasseux
JL
,
Fayad
ZA
,
Guertin
MC
,
Kastelein
JJ
,
Keyserling
C
,
Klepp
H
,
Koenig
W
,
L'Allier
PL
,
Lesperance
J
,
Luscher
TF
,
Paolini
JF
,
Tawakol
A
,
Waters
DD
,
Can HDL Infusions Significantly QUicken Atherosclerosis REgression (CHI-SQUARE) Investigators
.
Effects of the high-density lipoprotein mimetic agent CER-001 on coronary atherosclerosis in patients with acute coronary syndromes: a randomized trial
.
Eur Heart J
2014
;
35
:
3277
3286
.

64

Tardif
JC
,
Gregoire
J
,
L'Allier
PL
,
Ibrahim
R
,
Lesperance
J
,
Heinonen
TM
,
Kouz
S
,
Berry
C
,
Basser
R
,
Lavoie
MA
,
Guertin
MC
,
Rodes-Cabau
J
,
Effect of rHDL on Atherosclerosis-Safety and Efficacy (ERASE) Investigators
.
Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial
.
JAMA
2007
;
297
:
1675
1682
.

65

Gibson
CM
,
Kastelein
JJP
,
Phillips
AT
,
Aylward
PE
,
Yee
MK
,
Tendera
M
,
Nicholls
SJ
,
Pocock
S
,
Goodman
SG
,
Alexander
JH
,
Lincoff
AM
,
Bode
C
,
Duffy
D
,
Heise
M
,
Berman
G
,
Mears
SJ
,
Tricoci
P
,
Deckelbaum
LI
,
Steg
PG
,
Ridker
P
,
Mehran
R
.
Rationale and design of ApoA-I Event Reducing in Ischemic Syndromes II (AEGIS-II): a phase 3, multicenter, double-blind, randomized, placebo-controlled, parallel-group study to investigate the efficacy and safety of CSL112 in subjects after acute myocardial infarction
.
Am Heart J
2021
;
231
:
121
127
.

66

Armitage
J
,
Holmes
MV
,
Preiss
D
.
Cholesteryl ester transfer protein inhibition for preventing cardiovascular events: JACC review topic of the week
.
J Am Coll Cardiol
2019
;
73
:
477
487
.

67

Nissen
SE
,
Tardif
JC
,
Nicholls
SJ
,
Revkin
JH
,
Shear
CL
,
Duggan
WT
,
Ruzyllo
W
,
Bachinsky
WB
,
Lasala
GP
,
Tuzcu
EM
,
ILLUSTRATE Investigators
.
Effect of torcetrapib on the progression of coronary atherosclerosis
.
N Engl J Med
2007
;
356
:
1304
1316
.

68

Barter
PJ
,
Caulfield
M
,
Eriksson
M
,
Grundy
SM
,
Kastelein
JJ
,
Komajda
M
,
Lopez-Sendon
J
,
Mosca
L
,
Tardif
JC
,
Waters
DD
,
Shear
CL
,
Revkin
JH
,
Buhr
KA
,
Fisher
MR
,
Tall
AR
,
Brewer
B
,
ILLUMINATE Investigators
.
Effects of torcetrapib in patients at high risk for coronary events
.
N Engl J Med
2007
;
357
:
2109
2122
.

69

Group
HTRC
,
Bowman
L
,
Hopewell
JC
,
Chen
F
,
Wallendszus
K
,
Stevens
W
,
Collins
R
,
Wiviott
SD
,
Cannon
CP
,
Braunwald
E
,
Sammons
E
,
Landray
MJ
.
Effects of anacetrapib in patients with atherosclerotic vascular disease
.
N Engl J Med
2017
;
377
:
1217
1227
.

70

Lincoff
AM
,
Nicholls
SJ
,
Riesmeyer
JS
,
Barter
PJ
,
Brewer
HB
,
Fox
KAA
,
Gibson
CM
,
Granger
C
,
Menon
V
,
Montalescot
G
,
Rader
D
,
Tall
AR
,
McErlean
E
,
Wolski
K
,
Ruotolo
G
,
Vangerow
B
,
Weerakkody
G
,
Goodman
SG
,
Conde
D
,
McGuire
DK
,
Nicolau
JC
,
Leiva-Pons
JL
,
Pesant
Y
,
Li
W
,
Kandath
D
,
Kouz
S
,
Tahirkheli
N
,
Mason
D
,
Nissen
SE
,
ACCELERATE Investigators
.
Evacetrapib and cardiovascular outcomes in high-risk vascular disease
.
N Engl J Med
2017
;
376
:
1933
1942
.

71

Furtado
JD
,
Ruotolo
G
,
Nicholls
SJ
,
Dullea
R
,
Carvajal-Gonzalez
S
,
Sacks
FM
.
Pharmacological inhibition of CETP (Cholesteryl Ester Transfer Protein) increases HDL (High-Density Lipoprotein) that contains ApoC3 and other HDL subspecies associated with higher risk of coronary heart disease
.
Arterioscler Thromb Vasc Biol
2021
;
2
:
227
237
.

72

Lawler
PR
,
Bhatt
DL
,
Godoy
LC
,
Luscher
TF
,
Bonow
RO
,
Verma
S
,
Ridker
PM
.
Targeting cardiovascular inflammation: next steps in clinical translation
.
Eur Heart J
2021
;
42
:
113
131
.

73

Mohler
ER
3rd,
Ballantyne
CM
,
Davidson
MH
,
Hanefeld
M
,
Ruilope
LM
,
Johnson
JL
,
Zalewski
A
,
Darapladib
I
.
The effect of darapladib on plasma lipoprotein-associated phospholipase A2 activity and cardiovascular biomarkers in patients with stable coronary heart disease or coronary heart disease risk equivalent: the results of a multicenter, randomized, double-blind, placebo-controlled study
.
J Am Coll Cardiol
2008
;
51
:
1632
1641
.

74

Mallat
Z
,
Lambeau
G
,
Tedgui
A
.
Lipoprotein-associated and secreted phospholipases A(2) in cardiovascular disease: roles as biological effectors and biomarkers
.
Circulation
2010
;
122
:
2183
2200
.

75

Rosenson
RS
,
Stafforini
DM
.
Modulation of oxidative stress, inflammation, and atherosclerosis by lipoprotein-associated phospholipase A2
.
J Lipid Res
2012
;
53
:
1767
1782
.

76

Kolodgie
FD
,
Burke
AP
,
Skorija
KS
,
Ladich
E
,
Kutys
R
,
Makuria
AT
,
Virmani
R
.
Lipoprotein-associated phospholipase A2 protein expression in the natural progression of human coronary atherosclerosis
.
Arterioscler Thromb Vasc Biol
2006
;
26
:
2523
2529
.

77

Lp
PLASC
,
Thompson
A
,
Gao
P
,
Orfei
L
,
Watson
S
,
Di Angelantonio
E
,
Kaptoge
S
,
Ballantyne
C
,
Cannon
CP
,
Criqui
M
,
Cushman
M
,
Hofman
A
,
Packard
C
,
Thompson
SG
,
Collins
R
,
Danesh
J
.
Lipoprotein-associated phospholipase A(2) and risk of coronary disease, stroke, and mortality: collaborative analysis of 32 prospective studies
.
Lancet
2010
;
375
:
1536
1544
.

78

Serruys
PW
,
Garcia-Garcia
HM
,
Buszman
P
,
Erne
P
,
Verheye
S
,
Aschermann
M
,
Duckers
H
,
Bleie
O
,
Dudek
D
,
Botker
HE
,
von Birgelen
C
,
D'Amico
D
,
Hutchinson
T
,
Zambanini
A
,
Mastik
F
,
van Es
GA
,
van der Steen
AF
,
Vince
DG
,
Ganz
P
,
Hamm
CW
,
Wijns
W
,
Zalewski
A
,
Integrated Biomarker and Imaging Study-2 Investigators
.
Effects of the direct lipoprotein-associated phospholipase A(2) inhibitor darapladib on human coronary atherosclerotic plaque
.
Circulation
2008
;
118
:
1172
1182
.

79

Brugaletta
S
,
Garcia-Garcia
HM
,
Serruys
PW
,
Maehara
A
,
Farooq
V
,
Mintz
GS
,
de Bruyne
B
,
Marso
SP
,
Verheye
S
,
Dudek
D
,
Hamm
CW
,
Farhat
N
,
Schiele
F
,
McPherson
J
,
Lerman
A
,
Moreno
PR
,
Wennerblom
B
,
Fahy
M
,
Templin
B
,
Morel
MA
,
van Es
GA
,
Stone
GW
.
Relationship between palpography and virtual histology in patients with acute coronary syndromes
.
JACC Cardiovasc Imaging
2012
;
5
:
S19
27
.

80

Investigators
S
,
White
HD
,
Held
C
,
Stewart
R
,
Tarka
E
,
Brown
R
,
Davies
RY
,
Budaj
A
,
Harrington
RA
,
Steg
PG
,
Ardissino
D
,
Armstrong
PW
,
Avezum
A
,
Aylward
PE
,
Bryce
A
,
Chen
H
,
Chen
MF
,
Corbalan
R
,
Dalby
AJ
,
Danchin
N
,
De Winter
RJ
,
Denchev
S
,
Diaz
R
,
Elisaf
M
,
Flather
MD
,
Goudev
AR
,
Granger
CB
,
Grinfeld
L
,
Hochman
JS
,
Husted
S
,
Kim
HS
,
Koenig
W
,
Linhart
A
,
Lonn
E
,
Lopez-Sendon
J
,
Manolis
AJ
,
Mohler
ER
III
,
Nicolau
JC
,
Pais
P
,
Parkhomenko
A
,
Pedersen
TR
,
Pella
D
,
Ramos-Corrales
MA
,
Ruda
M
,
Sereg
M
,
Siddique
S
,
Sinnaeve
P
,
Smith
P
,
Sritara
P
,
Swart
HP
,
Sy
RG
,
Teramoto
T
,
Tse
HF
,
Watson
D
,
Weaver
WD
,
Weiss
R
,
Viigimaa
M
,
Vinereanu
D
,
Zhu
J
,
Cannon
CP
,
Wallentin
L
.
Darapladib for preventing ischemic events in stable coronary heart disease
.
N Engl J Med
2014
;
370
:
1702
1711
.

81

O'Donoghue
ML
,
Braunwald
E
,
White
HD
,
Lukas
MA
,
Tarka
E
,
Steg
PG
,
Hochman
JS
,
Bode
C
,
Maggioni
AP
,
Im
K
,
Shannon
JB
,
Davies
RY
,
Murphy
SA
,
Crugnale
SE
,
Wiviott
SD
,
Bonaca
MP
,
Watson
DF
,
Weaver
WD
,
Serruys
PW
,
Cannon
CP
,
SOLID-TIMI 52 Investigators
,
Steen
DL
.
Effect of darapladib on major coronary events after an acute coronary syndrome: the SOLID-TIMI 52 randomized clinical trial
.
JAMA
2014
;
312
:
1006
1015
.

82

Westlake
SL
,
Colebatch
AN
,
Baird
J
,
Kiely
P
,
Quinn
M
,
Choy
E
,
Ostor
AJ
,
Edwards
CJ
.
The effect of methotrexate on cardiovascular disease in patients with rheumatoid arthritis: a systematic literature review
.
Rheumatology (Oxford)
2010
;
49
:
295
307
.

83

Micha
R
,
Imamura
F
,
Wyler von Ballmoos
M
,
Solomon
DH
,
Hernan
MA
,
Ridker
PM
,
Mozaffarian
D
.
Systematic review and meta-analysis of methotrexate use and risk of cardiovascular disease
.
Am J Cardiol
2011
;
108
:
1362
1370
.

84

Liu
D
,
Lv
H
,
Liu
Q
,
Sun
Y
,
Hou
S
,
Zhang
L
,
Yang
M
,
Han
B
,
Wang
G
,
Wang
X
,
Du
W
,
Nie
H
,
Zhang
R
,
Huang
X
,
Hou
J
,
Yu
B
.
Atheroprotective effects of methotrexate via the inhibition of YAP/TAZ under disturbed flow
.
J Transl Med
2019
;
17
:
378
.

85

Ridker
PM
,
Everett
BM
,
Pradhan
A
,
MacFadyen
JG
,
Solomon
DH
,
Zaharris
E
,
Mam
V
,
Hasan
A
,
Rosenberg
Y
,
Iturriaga
E
,
Gupta
M
,
Tsigoulis
M
,
Verma
S
,
Clearfield
M
,
Libby
P
,
Goldhaber
SZ
,
Seagle
R
,
Ofori
C
,
Saklayen
M
,
Butman
S
,
Singh
N
,
Le May
M
,
Bertrand
O
,
Johnston
J
,
Paynter
NP
,
Glynn
RJ
,
Investigators CIRT
.
Low-dose methotrexate for the prevention of atherosclerotic events
.
N Engl J Med
2019
;
380
:
752
762
.

86

Swanson
KV
,
Deng
M
,
Ting
JP
.
The NLRP3 inflammasome: molecular activation and regulation to therapeutics
.
Nat Rev Immunol
2019
;
19
:
477
489
.

87

Libby
P
.
Interleukin-1 beta as a target for atherosclerosis therapy: biological basis of CANTOS and beyond
.
J Am Coll Cardiol
2017
;
70
:
2278
2289
.

88

Ridker
PM
,
Rane
M
.
Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease
.
Circ Res
2021
;
128
:
1728
1746
.

89

Ridker
PM
,
Everett
BM
,
Thuren
T
,
MacFadyen
JG
,
Chang
WH
,
Ballantyne
C
,
Fonseca
F
,
Nicolau
J
,
Koenig
W
,
Anker
SD
,
Kastelein
JJP
,
Cornel
JH
,
Pais
P
,
Pella
D
,
Genest
J
,
Cifkova
R
,
Lorenzatti
A
,
Forster
T
,
Kobalava
Z
,
Vida-Simiti
L
,
Flather
M
,
Shimokawa
H
,
Ogawa
H
,
Dellborg
M
,
Rossi
PRF
,
Troquay
RPT
,
Libby
P
,
Glynn
RJ
,
CANTOS Trial Group
.
Antiinflammatory therapy with canakinumab for atherosclerotic disease
.
N Engl J Med
2017
;
377
:
1119
1131
.

90

Everett
BM
,
MacFadyen
JG
,
Thuren
T
,
Libby
P
,
Glynn
RJ
,
Ridker
PM
.
Inhibition of interleukin-1beta and reduction in atherothrombotic cardiovascular events in the CANTOS trial
.
J Am Coll Cardiol
2020
;
76
:
1660
1670
.

91

Imazio
M
,
Nidorf
M
.
Colchicine and the heart
.
Eur Heart J
2021
;
42
:
2745
2760
.

92

Deftereos
S
,
Giannopoulos
G
,
Raisakis
K
,
Kossyvakis
C
,
Kaoukis
A
,
Panagopoulou
V
,
Driva
M
,
Hahalis
G
,
Pyrgakis
V
,
Alexopoulos
D
,
Manolis
AS
,
Stefanadis
C
,
Cleman
MW
.
Colchicine treatment for the prevention of bare-metal stent restenosis in diabetic patients
.
J Am Coll Cardiol
2013
;
61
:
1679
1685
.

93

Nidorf
SM
,
Eikelboom
JW
,
Budgeon
CA
,
Thompson
PL
.
Low-dose colchicine for secondary prevention of cardiovascular disease
.
J Am Coll Cardiol
2013
;
61
:
404
410
.

94

Nidorf
SM
,
Fiolet
ATL
,
Mosterd
A
,
Eikelboom
JW
,
Schut
A
,
Opstal
TSJ
,
The
SHK
,
Xu
XF
,
Ireland
MA
,
Lenderink
T
,
Latchem
D
,
Hoogslag
P
,
Jerzewski
A
,
Nierop
P
,
Whelan
A
,
Hendriks
R
,
Swart
H
,
Schaap
J
,
Kuijper
AFM
,
van Hessen
MWJ
,
Saklani
P
,
Tan
I
,
Thompson
AG
,
Morton
A
,
Judkins
C
,
Bax
WA
,
Dirksen
M
,
Alings
M
,
Hankey
GJ
,
Budgeon
CA
,
Tijssen
JGP
,
Cornel
JH
,
Thompson
PL
,
LoDoCo2 Trial Investigators
.
Colchicine in patients with chronic coronary disease
.
N Engl J Med
2020
;
383
:
1838
1847
.

95

Tardif
JC
,
Kouz
S
,
Waters
DD
,
Bertrand
OF
,
Diaz
R
,
Maggioni
AP
,
Pinto
FJ
,
Ibrahim
R
,
Gamra
H
,
Kiwan
GS
,
Berry
C
,
Lopez-Sendon
J
,
Ostadal
P
,
Koenig
W
,
Angoulvant
D
,
Gregoire
JC
,
Lavoie
MA
,
Dube
MP
,
Rhainds
D
,
Provencher
M
,
Blondeau
L
,
Orfanos
A
,
L'Allier
PL
,
Guertin
MC
,
Roubille
F
.
Efficacy and safety of low-dose colchicine after myocardial infarction
.
N Engl J Med
2019
;
381
:
2497
2505
.

96

Tong
DC
,
Quinn
S
,
Nasis
A
,
Hiew
C
,
Roberts-Thomson
P
,
Adams
H
,
Sriamareswaran
R
,
Htun
NM
,
Wilson
W
,
Stub
D
,
van Gaal
W
,
Howes
L
,
Collins
N
,
Yong
A
,
Bhindi
R
,
Whitbourn
R
,
Lee
A
,
Hengel
C
,
Asrress
K
,
Freeman
M
,
Amerena
J
,
Wilson
A
,
Layland
J
.
Colchicine in patients with acute coronary syndrome: the Australian COPS randomized clinical trial
.
Circulation
2020
;
142
:
1890
1900
.

97

Fiolet
ATL
,
Opstal
TSJ
,
Mosterd
A
,
Eikelboom
JW
,
Jolly
SS
,
Keech
AC
,
Kelly
P
,
Tong
DC
,
Layland
J
,
Nidorf
SM
,
Thompson
PL
,
Budgeon
C
,
Tijssen
JGP
,
Cornel
JH
.
Efficacy and safety of low-dose colchicine in patients with coronary disease: a systematic review and meta-analysis of randomized trials
.
Eur Heart J
2021
;
42
:
2765
2775
.

98

Silva
GM
,
Franca-Falcao
MS
,
Calzerra
NTM
,
Luz
MS
,
Gadelha
DDA
,
Balarini
CM
,
Queiroz
TM
.
Role of renin-angiotensin system components in atherosclerosis: focus on Ang-II, ACE2, and Ang-1-7
.
Front Physiol
2020
;
11
:
1067
.

99

Mason
RP
.
Mechanisms of plaque stabilization for the dihydropyridine calcium channel blocker amlodipine: review of the evidence
.
Atherosclerosis
2002
;
165
:
191
199
.

100

Nissen
SE
,
Tuzcu
EM
,
Libby
P
,
Thompson
PD
,
Ghali
M
,
Garza
D
,
Berman
L
,
Shi
H
,
Buebendorf
E
,
Topol
EJ
,
CAMELOT Investigators
.
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
:
2217
2225
.

101

Rodriguez-Granillo
GA
,
Vos
J
,
Bruining
N
,
Garcia-Garcia
HM
,
de Winter
S
,
Ligthart
JM
,
Deckers
JW
,
Bertrand
M
,
Simoons
ML
,
Ferrari
R
,
Fox
KM
,
Remme
W
,
De Feyter
PJ
,
Investigators of the EUROPA Study
.
Long-term effect of perindopril on coronary atherosclerosis progression (from the perindopril's prospective effect on coronary atherosclerosis by angiography and intravascular ultrasound evaluation [PERSPECTIVE] study)
.
Am J Cardiol
2007
;
100
:
159
163
.

102

Luscher
TF
,
Pieper
M
,
Tendera
M
,
Vrolix
M
,
Rutsch
W
,
van den Branden
F
,
Gil
R
,
Bischoff
KO
,
Haude
M
,
Fischer
D
,
Meinertz
T
,
Munzel
T
.
A randomized placebo-controlled study on the effect of nifedipine on coronary endothelial function and plaque formation in patients with coronary artery disease: the ENCORE II study
.
Eur Heart J
2009
;
30
:
1590
1597
.

103

Pitt
B
,
Byington
RP
,
Furberg
CD
,
Hunninghake
DB
,
Mancini
GB
,
Miller
ME
,
Riley
W
,
PREVENT Investigators
,
Effect of amlodipine on the progression of atherosclerosis and the occurrence of clinical events. PREVENT Investigators
.
Circulation
2000
;
102
:
1503
1510
.

104

Poole-Wilson
PA
,
Lubsen
J
,
Kirwan
BA
,
van Dalen
FJ
,
Wagener
G
,
Danchin
N
,
Just
H
,
Fox
KA
,
Pocock
SJ
,
Clayton
TC
,
Motro
M
,
Parker
JD
,
Bourassa
MG
,
Dart
AM
,
Hildebrandt
P
,
Hjalmarson
A
,
Kragten
JA
,
Molhoek
GP
,
Otterstad
JE
,
Seabra-Gomes
R
,
Soler-Soler
J
,
Weber
S
,
Coronary disease Trial Investigating Outcome with Nifedipine gastrointestinal therapeutic system investigators
.
Effect of long-acting nifedipine on mortality and cardiovascular morbidity in patients with stable angina requiring treatment (ACTION trial): randomised controlled trial
.
Lancet
2004
;
364
:
849
857
.

105

Heart Outcomes Prevention Evaluation Study Investigators
,
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
.
N Engl J Med
2000
;
342
:
145
153
.

106

Fox
KM
,
Investigators EUtOrocewPiscAd
.
Efficacy of perindopril in reduction of cardiovascular events among patients with stable coronary artery disease: randomised, double-blind, placebo-controlled, multicentre trial (the EUROPA study)
.
Lancet
2003
;
362
:
782
788
.

107

Braunwald
E
,
Domanski
MJ
,
Fowler
SE
,
Geller
NL
,
Gersh
BJ
,
Hsia
J
,
Pfeffer
MA
,
Rice
MM
,
Rosenberg
YD
,
Rouleau
JL
,
PEACE Trial Investigators
.
Angiotensin-converting-enzyme inhibition in stable coronary artery disease
.
N Engl J Med
2004
;
351
:
2058
2068
.

108

Bots
ML
,
Remme
WJ
,
Luscher
TF
,
Fox
KM
,
Bertrand
M
,
Ferrari
R
,
Simoons
ML
,
Grobbee
DE
,
Investigators E-P
.
ACE inhibition and endothelial function: main findings of PERFECT, a sub-study of the EUROPA trial
.
Cardiovasc Drugs Ther
2007
;
21
:
269
279
.

109

Ceconi
C
,
Fox
KM
,
Remme
WJ
,
Simoons
ML
,
Bertrand
M
,
Parrinello
G
,
Kluft
C
,
Blann
A
,
Cokkinos
D
,
Ferrari
R
,
EUROPA Investigators
,
PERTINENT Investigators and the Statistical Committee
.
ACE inhibition with perindopril and endothelial function. Results of a substudy of the EUROPA study: PERTINENT
.
Cardiovasc Res
2007
;
73
:
237
246
.

110

Nissen
SE
,
Nicholls
SJ
,
Wolski
K
,
Nesto
R
,
Kupfer
S
,
Perez
A
,
Jure
H
,
De Larochelliere
R
,
Staniloae
CS
,
Mavromatis
K
,
Saw
J
,
Hu
B
,
Lincoff
AM
,
Tuzcu
EM
,
PERISCOPE Investigators
.
Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial
.
JAMA
2008
;
299
:
1561
1573
.

111

Gerstein
HC
,
Ratner
RE
,
Cannon
CP
,
Serruys
PW
,
Garcia-Garcia
HM
,
van Es
GA
,
Kolatkar
NS
,
Kravitz
BG
,
Miller
DM
,
Huang
C
,
Fitzgerald
PJ
,
Nesto
RW
,
Group
AS
.
Effect of rosiglitazone on progression of coronary atherosclerosis in patients with type 2 diabetes mellitus and coronary artery disease: the assessment on the prevention of progression by rosiglitazone on atherosclerosis in diabetes patients with cardiovascular history trial
.
Circulation
2010
;
121
:
1176
1187
.

112

de Jong
M
,
van der Worp
HB
,
van der Graaf
Y
,
Visseren
FLJ
,
Westerink
J
.
Pioglitazone and the secondary prevention of cardiovascular disease. A meta-analysis of randomized-controlled trials
.
Cardiovasc Diabetol
2017
;
16
:
134
.

113

Bach
RG
,
Brooks
MM
,
Lombardero
M
,
Genuth
S
,
Donner
TW
,
Garber
A
,
Kennedy
L
,
Monrad
ES
,
Pop-Busui
R
,
Kelsey
SF
,
Frye
RL
,
BARI 2D Investigators
.
Rosiglitazone and outcomes for patients with diabetes mellitus and coronary artery disease in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI 2D) trial
.
Circulation
2013
;
128
:
785
794
.

114

Green
JB
,
Bethel
MA
,
Armstrong
PW
,
Buse
JB
,
Engel
SS
,
Garg
J
,
Josse
R
,
Kaufman
KD
,
Koglin
J
,
Korn
S
,
Lachin
JM
,
McGuire
DK
,
Pencina
MJ
,
Standl
E
,
Stein
PP
,
Suryawanshi
S
,
Van de Werf
F
,
Peterson
ED
,
Holman
RR
,
Group
TS
.
Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2015
;
373
:
232
242
.

115

Bourantas
CV
,
Jaffer
FA
,
Gijsen
FJ
,
van Soest
G
,
Madden
SP
,
Courtney
BK
,
Fard
AM
,
Tenekecioglu
E
,
Zeng
Y
,
van der Steen
AFW
,
Emelianov
S
,
Muller
J
,
Stone
PH
,
Marcu
L
,
Tearney
GJ
,
Serruys
PW
.
Hybrid intravascular imaging: recent advances, technical considerations, and current applications in the study of plaque pathophysiology
.
Eur Heart J
2017
;
38
:
400
412
.

116

Garcia-Garcia
HM
,
Gomez-Lara
J
,
Gonzalo
N
,
Garg
S
,
Shin
ES
,
Goedhart
D
,
Serruys
PW
.
A comparison of the distribution of necrotic core in bifurcation and non-bifurcation coronary lesions: an in vivo assessment using intravascular ultrasound radiofrequency data analysis
.
EuroIntervention
2010
;
6
:
321
327
.

117

Gijsen
F
,
Katagiri
Y
,
Barlis
P
,
Bourantas
C
,
Collet
C
,
Coskun
U
,
Daemen
J
,
Dijkstra
J
,
Edelman
E
,
Evans
P
,
van der Heiden
K
,
Hose
R
,
Koo
BK
,
Krams
R
,
Marsden
A
,
Migliavacca
F
,
Onuma
Y
,
Ooi
A
,
Poon
E
,
Samady
H
,
Stone
P
,
Takahashi
K
,
Tang
D
,
Thondapu
V
,
Tenekecioglu
E
,
Timmins
L
,
Torii
R
,
Wentzel
J
,
Serruys
P
.
Expert recommendations on the assessment of wall shear stress in human coronary arteries: existing methodologies, technical considerations, and clinical applications
.
Eur Heart J
2019
;
40
:
3421
3433
.

118

Thondapu
V
,
Bourantas
CV
,
Foin
N
,
Jang
IK
,
Serruys
PW
,
Barlis
P
.
Biomechanical stress in coronary atherosclerosis: emerging insights from computational modelling
.
Eur Heart J
2017
;
38
:
81
92
.

119

Stone
PH
,
Maehara
A
,
Coskun
AU
,
Maynard
CC
,
Zaromytidou
M
,
Siasos
G
,
Andreou
I
,
Fotiadis
D
,
Stefanou
K
,
Papafaklis
M
,
Michalis
L
,
Lansky
AJ
,
Mintz
GS
,
Serruys
PW
,
Feldman
CL
,
Stone
GW
.
Role of low endothelial shear stress and plaque characteristics in the prediction of nonculprit major adverse cardiac events: the PROSPECT study
.
JACC Cardiovasc Imaging
2018
;
11
:
462
471
.

120

Costopoulos
C
,
Maehara
A
,
Huang
Y
,
Brown
AJ
,
Gillard
JH
,
Teng
Z
,
Stone
GW
,
Bennett
MR
.
Heterogeneity of plaque structural stress is increased in plaques leading to MACE
.
JACC Cardiovasc Imaging
2020
;
13
:
1206
1218
.

121

Brown
AJ
,
Teng
Z
,
Calvert
PA
,
Rajani
NK
,
Hennessy
O
,
Nerlekar
N
,
Obaid
DR
,
Costopoulos
C
,
Huang
Y
,
Hoole
SP
,
Goddard
M
,
West
NE
,
Gillard
JH
,
Bennett
MR
.
Plaque structural stress estimations improve prediction of future major adverse cardiovascular events after intracoronary imaging
.
Circ Cardiovasc Imaging
2016
;
9
:
e004172
.

122

Doradla
P
,
Otsuka
K
,
Nadkarni
A
,
Villiger
M
,
Karanasos
A
,
Zandvoort
L
,
Dijkstra
J
,
Zijlstra
F
,
Soest
GV
,
Daemen
J
,
Regar
E
,
Bouma
BE
,
Nadkarni
SK
.
Biomechanical stress profiling of coronary atherosclerosis: identifying a multifactorial metric to evaluate plaque rupture risk
.
JACC Cardiovasc Imaging
2020
;
13
:
804
816
.

123

Costopoulos
C
,
Huang
Y
,
Brown
AJ
,
Calvert
PA
,
Hoole
SP
,
West
NEJ
,
Gillard
JH
,
Teng
Z
,
Bennett
MR
.
Plaque rupture in coronary atherosclerosis is associated with increased plaque structural stress
.
JACC Cardiovasc Imaging
2017
;
10
:
1472
1483
.

124

Okafor
ON
,
Gorog
DA
.
Endogenous fibrinolysis: an important mediator of thrombus formation and cardiovascular risk
.
J Am Coll Cardiol
2015
;
65
:
1683
1699
.

125

Lordan
R
,
Tsoupras
A
,
Zabetakis
I
.
Platelet activation and prothrombotic mediators at the nexus of inflammation and atherosclerosis: potential role of antiplatelet agents
.
Blood Rev
2020
;
45
:
100694
.

126

Aikawa
M
,
Rabkin
E
,
Sugiyama
S
,
Voglic
SJ
,
Fukumoto
Y
,
Furukawa
Y
,
Shiomi
M
,
Schoen
FJ
,
Libby
P
.
An HMG-CoA reductase inhibitor, cerivastatin, suppresses growth of macrophages expressing matrix metalloproteinases and tissue factor in vivo and in vitro
.
Circulation
2001
;
103
:
276
283
.

127

Fukumoto
Y
,
Libby
P
,
Rabkin
E
,
Hill
CC
,
Enomoto
M
,
Hirouchi
Y
,
Shiomi
M
,
Aikawa
M
.
Statins alter smooth muscle cell accumulation and collagen content in established atheroma of watanabe heritable hyperlipidemic rabbits
.
Circulation
2001
;
103
:
993
999
.

128

Schuster
S
,
Rubil
S
,
Endres
M
,
Princen
HMG
,
Boeckel
JN
,
Winter
K
,
Werner
C
,
Laufs
U
.
Anti-PCSK9 antibodies inhibit pro-atherogenic mechanisms in APOE*3Leiden.CETP mice
.
Sci Rep
2019
;
9
:
11079
.

129

Vergallo
R
,
Crea
F
.
Atherosclerotic plaque healing
.
N Engl J Med
2020
;
383
:
846
857
.

130

Hougaard
M
,
Hansen
HS
,
Thayssen
P
,
Maehara
A
,
Antonsen
L
,
Junker
A
,
Mintz
GS
,
Jensen
LO
.
Influence of ezetimibe on plaque morphology in patients with ST elevation myocardial infarction assessed by optical coherence tomography: an OCTIVUS sub-study
.
Cardiovasc Revasc Med
2020
;
21
:
1417
1424
.

131

Yano
H
,
Horinaka
S
,
Ishimitsu
T
.
Effect of evolocumab therapy on coronary fibrous cap thickness assessed by optical coherence tomography in patients with acute coronary syndrome
.
J Cardiol
2020
;
75
:
289
295
.

Author notes

Conflict of interest: L.R. received research grants from Abbott, Boston Scientific, Infraredx and consulting fees from Medtronic. A.B. received payment for lectures and educational events from Medtronic. The remaining authors do not have any conflicts of interest to disclose.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)

Supplementary data