You are on page 1of 10

International Journal of Cardiology 192 (2015) 72–81

Contents lists available at ScienceDirect

International Journal of Cardiology

journal homepage: www.elsevier.com/locate/ijcard

Review

Atherosclerosis: Recent trials, new targets and future directions


Ricardo Ladeiras-Lopes a,b, Stefan Agewall c, Ahmed Tawakol d,e, Bart Staels f, Evan Stein g, Robert J. Mentz h,
Adelino Leite-Moreira a, Faiez Zannad i, Wolfgang Koenig j,k,l,⁎
a
Department of Physiology and Cardiothoracic Surgery, Cardiovascular Research and Development Centre, Faculty of Medicine of the University of Porto, Porto, Portugal
b
Department of Cardiology, Gaia/Espinho Hospital Centre, Vila Nova de Gaia, Portugal
c
Department of Cardiology, Oslo University Hospital Ullevål and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
d
Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
e
Cardiology Division, Harvard Medical School, Boston, MA, USA
f
INSERM UMR1011, Univ. Lille 2, Institut Pasteur de Lille, EGID, Lille, France
g
Metabolic and Atherosclerosis Research Centre, Cincinatti, OH, USA
h
Duke Clinical Research Institute and the Division of Cardiology, Duke University Hospital, Durham, NC, USA
i
INSERM, Centre d'Investigation Clinique-9501, Université de Lorraine, Nancy, France
j
Department of Internal Medicine II — Cardiology, University of Ulm Medical Center, Ulm, Germany
k
Deutsches Herzzentrum München, Technische Universität München, Munich Germany
l
DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Mortality from cardiovascular diseases (CVD) represents the primary cause of death worldwide. Prevention or
Received 30 December 2014 treatment of atherosclerosis and its clinical sequelae is a central goal in the management of patients with
Received in revised form 9 April 2015 established vascular disease or those at high-risk for vascular events. This paper provides a review of the contem-
Accepted 6 May 2015
porary pharmacological armamentarium targeting atherosclerosis and also highlights strategies to support fu-
Available online 8 May 2015
ture clinical trial design. Powering future trials targeting LDL-cholesterol to its absolute reduction and
Keywords:
including patients with a higher LDL-C despite optimal medical therapy (or unable to tolerate statins) will in-
Atherosclerosis crease the odds of meaningful results. Mendelian randomization studies may identify new causal risk factors
Pharmacology for CVD that would help in the selection of the patients most likely to benefit from a specific new compound. Fur-
Clinical research thermore, imaging techniques integrating a morphological and functional assessment such as IVUS, OCT, PET/CT
and PET/MRI may represent in a near future robust “soft” endpoints to support successful translation of early re-
search into meaningful phase III clinical outcome trials.
© 2015 Elsevier Ireland Ltd. All rights reserved.

1. Introduction LDL-C after an acute coronary syndrome (ACS) suffers a subsequent


adverse event within a 2-year period [4]. In this way, there is significant
Clinical complications of atherosclerosis like myocardial infarction residual cardiovascular risk that persists despite low LDL-C, highlighting
(MI), stroke and peripheral arterial disease represent the leading the role of “atherogenic dyslipidemia” that includes low HDL-C and high
cause of mortality and morbidity in the world [1]. Therefore, the devel- triglycerides [5].It is likely that longer duration of statin therapy combined
opment of drugs targeting the atherosclerotic process has been an im- with early introduction of LDL-C lowering will reduce residual risk, and
portant area of contemporary clinical research. with inexpensive and effective statins now available, recent guidelines
While statins are the cornerstone of prevention they are not the pan- in the USA has expanded such treatment to lower risk patients [6].
acea for eliminating atherosclerotic cardiovascular disease (ASCVD) [2]. A The utilization of complex imaging techniques including anatomical
meta-analysis published in 2004 including patients on standard statin and functional evaluations and a better selection of high-risk patients
therapy showed a 21.7% 5-year event rate for major adverse cardiovascu- may foster the successful design of trials. Indeed, a large number of
lar event (MACE) in patients with prior ASCVD [3]. Furthermore, almost 1 lipid-associated new targets or targets reflecting other pathways of the
in 10 high-risk patients treated with intensive statin therapy with low complex atherosclerotic process are being evaluated in mechanistic im-
aging studies as well as in large outcome trials in the endeavor of residual
vascular risk reduction (see Tables 1, 2 and 3). The latter will definitely in-
⁎ Corresponding author at: Klinik für Herz-& Kreislauferkrankungen, Deutsches clude patient-tailored approaches in order to modulate the complex ath-
Herzzentrum München, Technische Universität München, Lazarettstr. 3680636 Munich,
Germany. Tel.: +49 89 1218 3235; fax: +49 89 1218 4083.
erosclerotic process, emphasizing a strategy of personalized medicine [7].
E-mail addresses: ricardoladeiraslopes@gmail.com (R. Ladeiras-Lopes), This paper results from a discussion at the 2013 Cardiovascular
koenig@dhm.mhn.de (W. Koenig). Clinical Trialists' Forum held in Paris, France and aims to provide an

http://dx.doi.org/10.1016/j.ijcard.2015.05.013
0167-5273/© 2015 Elsevier Ireland Ltd. All rights reserved.
R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81 73

overview of the most recent clinical data on antiatherosclerotic com- with a significant change in carotid intima-media thickness (cIMT)
pounds, emphasizing new targets and strategies for robust clinical [11]. These results challenged not only the role of ezetimibe in CVD
trial design. prevention but also pointed to the limitations of biomarkers and sur-
rogate non-invasive markers to assess the efficacy of a lipid-lowering
drug.
2. Lipoprotein metabolism However, the results of the Improved Reduction of Outcomes:
Vytorin Efficacy International (IMPROVE-IT) Trial comparing simvastat-
2.1. Cholesterol absorption inhibition in plus ezetimibe with simvastatin alone in 18,144 patients included
within 5 days post-ACS showed a modest 6.4% relative risk reduction
Ezetimibe is a selective inhibitor of cholesterol absorption at the (RRR) of the primary endpoint and thus demonstrated a beneficial
brush border of the small intestine mediated by the Niemman–Pick role of ezetimibe in vascular protection [12]. Baseline LDL-C was
C1-like 1 protein, thus impairing the uptake of intestinal lumen micelles 95 mg/dl and was reduced to 69.5 mg/dL in the simvastatin group and
into the enterocyte [8,9] (Fig. 1). Lower cholesterol absorption depletes to 53.7 mg/dL in the combination group during 7 years of follow-up
the hepatic pool of cholesterol and upregulates hepatic LDL receptors, with a good safety profile.
therefore boosting LDL-C clearance from the circulation. Ezetimibe is
able to induce a significant LDL-C reduction of 15–20% alone or by ap- 2.2. PCSK9 inhibition
proximately 50% combined with simvastatin [10].
Although the results of ezetimibe and simvastatin in Hypercholes- The proprotein convertase subtilisin/kexin (PCSK) type 9 is a serine
terolemia Enhances Atherosclerotic Regression (ENHANCE) Trial protease which modulates lipoprotein metabolism by its circulating
including patients with familial hypercholesterolemia showed a reduc- component binding with LDL to the hepatic LDL receptor promoting he-
tion in LDL-C and hsCRP when a combination therapy of simvastatin and patic internalization and intracellular degradation of the LDL receptor,
ezetimibe was compared to simvastatin alone, yet it was not associated preventing its recycling (Fig. 1) [9,13]. The latter is a fundamental player
in LDL clearance from the circulation. In addition, pre-clinical research
has also identified roles for PCSK9 in peripheral tissues. For instance,
Table 1 PCSK9 is induced by inflammation [14] and small interfering RNA
Drug targets for atherosclerosis and current research development phase. (siRNA) knockdown of PCSK9 in human macrophages attenuates the in-
flammatory response elicited by oxidized LDL (oxLDL) [15]. Lastly, stat-
Target Clinical
research in therapy increases the expression of PCSK9 [16]. The integration of
phase these data, together with overwhelming evidence that patients with
LDL lowering
loss-of-function mutations of PCSK9 have 15–30% reductions in LDL-C
PCSK9 antibodies and 47–88% reduction in the risk of coronary heart disease (CHD)
Alirocumab (SAR236553/REGN727) 3 [19], supports a role for PCSK9 as an interesting target for anti-
Evolocumab (AMG145) 3 atherosclerosis drug therapy.
Bococizimab (PF-04950615/RN316) 3
There are several ways to interfere with PCSK9: mimetic peptides and
MTTP (microsomal triglyceride transfer protein) inhibitors 3
Antisense apoB (mipomersen) 3 adnectins (with similarities to small monoclonal antibodies), small-
ATP-citrate lyase inhibitor (ETC-1002) 2 molecule inhibitors, gene silencing (e.g., antisense oligonucleotides and
TR-β agonist (MGL-3196) 1 siRNA) and monoclonal antibodies [13]. To date, the most promising
Lp(a) strategy to impair the action of PCSK9 appears to be the use of monoclo-
Antisense apo(a) 1
Triglycerides
nal antibodies. Indeed, two monoclonal antibodies targeting PCSK9,
Antisense apo C-III (ISIS 304801) 2 AMG145 (evolocumab, Amgen), SAR236553/REGN727 (alirocumab,
PPAR agonists (α:K877, α/δ:GFT505, δ:CER-002) 2 Sanofi/Regeneron) and bococizumab (Pfizer) reported encouraging re-
Omega-3 free fatty acids (eicosapentaenoic acid and 3 sults in phase 1 and 2 trials. The body of evidence accumulated since
docosahexaenoic acid)
2009 has shown that these drugs elicit a significant and sustained
Acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1) inhibitors 2
(LCQ908) 40–70% reduction of LDL-C in patients with familial hypercholesterol-
Angiopoietin-like proteins (ANGPTLs) 3 and 4 inhibitors Pre-clinical emia (FH) [18–22] and non-FH [21,23–26]. LDL-C lowering was observed
HDL in patients with PCSK9 inhibitors, alone or in combination with a statin,
Reconstituted HDL (CSL-112) 2 and also in statin-intolerant patients [27]. Phase 3 trials are currently in
Recombinant HDL (CER-001) 2
Apo A-I inducer (RVX-208) 2
progress to assess the role of the following drugs on cardiovascular
Apo A-I milano (ETC-216/MDCO-216) 1 events: evolocumab (FOURIER; 28,000 patients with prior MI or stroke)
Apo A-I mimetics (D6F APP018, ATI-5261, …) 2 [28], alirocumab (ODYSSEY OUTCOMES; 18,000 patients with recent
ABCA1 inducer Pre-clinical ACS) [29] and bococizumab (SPIRE 1 and 2; 17,000 and 9,000 patients,
Antagomir miR-33 Pre-clinical
respectively, with high CV risk as defined by LDL-C levels) [30,31].
CETPi (evacetrapib, anaceptrapib) 3
rLCAT (APC-501) Pre-clinical The potential disadvantages of these agents include their subcutane-
LXR-623 Pre-clinical ous administration (although it may be performed monthly or twice
SR-B1 inhibitor (ITX5061) 1 monthly, which may be an advantage compared to a daily drug use)
Inflammation/atherosclerosis and injection site reactions (in about 6% of patients both with
Inflammation
sPLA2 (varespladib) 3
evolocumab [32] and alirocumab [32]). Furthermore, long-term im-
LpPLA2 (darapladib) 3 mune reactions even to fully human antibodies may appear, although
ox-LDL antibodies 2 available evidence from trials suggests that they are rare. The larger
5-lipoxygenase inhibitors (VIA-2291) 2 phase 3 trials underway may help to clarify the long-term tolerability
Methotrexate (CIRT) 3
of these drugs. The recently published results of a 52-week treatment
CCR-2 inhibition 2
CRP antisense 2 with evolocumab (DESCARTES; every 4 weeks) showed sustained effi-
IL1b/IL1RA/Inflammasome inhibition (canakinumab) 3 cacy in reducing LDL-C with acceptable safety and tolerability [33]. Fur-
Immunization strategies thermore, recently announced findings showed that alirocumab on top
Antibodies against modified apoB epitopes (MLDL1278A) 2 of statin therapy (administered every 2 weeks) in very-high risk pa-
ApoB vaccination strategies Pre-clinical
tients with hypercholesterolemia elicited a 48% reduction in MACE
74 R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81

Fig. 1. Schematic representation of promising targets for atherosclerosis drug therapy. CETP — Cholesteryl ester transfer protein; EL — endothelial lipase; LDLr — LDL receptor; Lp-PLA2 —
lipoprotein-associated phospholipase A2; MTTP — Microsomal triglyceride transfer protein; NPC1L1 — Niemann–Pick C1-Like 1; oxLDL — oxidized LDL particles; sPLA2 — secretory
phospholipase A2.

over 78 weeks of treatment [34]. Similar results have been reported for possibly overcoming the problem of hepatic liver accumulation with
evolocumab from the OSLER program with evolocumab [35]. The results previous inhibitors. However, enterocyte-specific MTTP inhibitors may
of the larger, outcome-driven trials may help to clarify the role of these be associated with gastrointestinal adverse effects and deficiency of
promising agents in the treatment of atherosclerosis. fat-soluble vitamins [45], therefore further research is needed to clarify
the efficacy and tolerability of these agents.

2.3. Microsomal triglyceride transfer protein (MTTP) inhibition


2.4. Antisense oligonucleotides targeting apolipoproteins
MTTP is an intracellular lipid-transfer protein with a central role in
the assembly of chylomicrons and VLDL in enterocytes and hepatocytes, Antisense oligonucleotides (ASO) are short (about 20 nucleotides)
respectively [9,36]. Patients with abetalipoproteinemia have extremely single-stranded nucleic acid analogs that bind to complementary se-
low levels of apoB-containing lipoproteins (chylomicrons, VLDL, IDL quences in target mRNAs, preventing their translation into the protein
and LDL) in circulation due to a defect in MTTP activity [37], suggesting product [46].
a potential therapeutic role for MTTP inhibition. Mipomersen is an ASO that blocks hepatic synthesis of apo B-100, a
Lomitapide is a MTTP inhibitor approved in the US and in the EU for central component of all proatherogenic lipoproteins. It has been shown
the treatment of homozygous FH. Approval was based on the recently to decrease VLDL-C and LDL-C levels [47–49]. Its consistent efficacy led
published results of an open-label phase III trial including 29 patients to the FDA approval for the treatment of homozygous FH in January
with homozygous FH which demonstrated significant reductions 2013. Mipomersen requires weekly subcutaneous administration and,
of LDL-C (30–50%), triglycerides (31–45%), apoB (43–49%) and as lomitapide, is associated with hepatic fat accumulation [47].
Lp(a) (15–19%) [38]. Lomitapide had been demonstrated in a phase II Other ASOs targeting different apolipoproteins have also appeared
trial to elicit additional LDL-C reduction as add-on therapy to ezetimibe in the last several years, aiming at decreasing triglycerides and in-
in patients with non-FH [39]. However, the potential for an increase in creasing HDL-C and also decreasing Lp(a). ISIS-APOCIII is an ASO
hepatic fat [40] and liver enzyme elevations in these trials raised con- targeting apo C-III that reduces triglycerides and increases HDL-C with
cerns about potential hepatotoxicity of lomitapide. an acceptable safety profile [50]. Furthermore, an ASO targeting
Future outcome trials with lomitapide as well as additional apolipoprotein(a) is also under development, eliciting a specific, robust
studies with experimental MTTP inhibitors (implitapide [41,42] and and sustained dose-dependent lowering of Lp(a), an independent risk
CP-346086 [43]) are needed. A small-molecule (SLx-4090) designed factor for CVD [51]. Additional clinical studies assessing the clinical ben-
to selectively inhibit enterocyte MTTP is also under investigation [44], efit of this new generation of therapies are encouraged.
Table 2
Recently published outcome-driven phase 3 trials.

Trial, year Drug therapy Population Number Mean baseline Length of Effect on lipid Primary outcome Results (primary Comments
published of lipids (mg/dL) follow-up concentration outcome)
(reference) patients (months)
enrolled

ILLUMINATE, Torcetrapib 60 mg daily + 45–75 years, 15,067 LDL 80; HDL 49; 12 ↓ LDL 22 mg/dL; ↑HDL Death from CAD, non-fatal 6.2% event-rate in the Torcetrapib increased cardiovascular
2007 [78] atorvastatin versus atorvastatin history of CVD apoA1 128; (prematurely 6 mg/dL; ↓ TG 10 mg/dL; MI, stroke, hospitalization for intervention group; 5.0% and non-cardiovascular death; 5.4

R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81


only or type 2 apoB 73 terminated) ↑ apoA1 25% (at 3 months); unstable angina in placebo; HR = 1.25 mm Hg increase in SAP
diabetes ↓ apoB 13% (at 3 months) (1.09–1.44)
AIM-HIGH, ER Niacin (1500–2000 mg) + ≥45 years, 3,414 LDL 74; HDL 35; 36 ↓ LDL 14%; ↑HDL 25%; Death from CAD, non-fatal 16.4% event-rate in the No incremental benefit of niacin in
2011 [72] simvastatin 40–80 mg+/− history of CVD TG 168; apoA-1 ↓ TG 31% MI, ischemic stroke, intervention group; reducing CV events despite
ezetimibe 10 mg daily versus 122; apoB 83 hospitalization for ACS or 16.2% in placebo; HR = significant increase in HDL-C and
simvastatin 40–80 mg+/− symptom-driven coronary or 1.02 (0.87–1.21) decrease in TG
ezetimibe 10 mg daily cerebral revascularization
Dal-OUTCOMES, Dalcetrapib 600 mg daily + ≥45 years; 15,871 LDL 76; HDL 42; 31 Minimal effect on LDL; Major coronary event 8.3% event rate with Substantial increase in HDL with
2012 [60] OMT versus OMT recent ACS apoA1 137; ↑HDL 31–40%; ↑ TG 4–10%; (including death from CAD) intervention; 8.0% with no effect on CV outcomes; 0.6 mm
apoB 81 ↑ apoA1 9%; minimal effect and ischemic stroke place; HR 1.04 g increase in SAP; ↑18% CRP levels
on apoB (0.93–1.16)
HPS-2 THRIVE, ER niacin (2 g) + laropiprant 50–80 years, 25,673 LDL 63; HDL 44; 48 ↓ LDL 10 mg/dL; ↑HDL Major coronary event 13.2% event-rate at 4 Significant excess of new-onset
2013 [73] (40 mg) daily versus simvastatin history of CVD TG 125 34 mg/dL; ↓ TG 33 mg/dL (including death from CAD), years in the intervention diabetes, diabetic complications,
40 mg (+/− ezetimibe) stroke (including death or group; 13.7% in placebo; bleeding, infection in the
hemorrhagic), HR = 0.96 (0.90–1.03) intervention group
revascularization
VISTA-16, 2014 Varespladib 500 mg daily + ≥40 years; 5,145 LDL 105; HDL 4 ↓ LDL 29%; ↑HDL 5.1%; Death from CAD, non fatal 6.1% event rate with Significant increased risk of MI and
[102] OMT versus OMT recent ACS 43; TG 154 (prematurely ↓ TG 22% MI, stroke, hospitalization for intervention; 5.1% with combined cardiovascular mortality,
terminated) unstable angina place; HR 1.25 MI and stroke in varespladib-treated
(0.97–1.61) patients; 6-month survival data
collected in only 31% of patients;
↓85% CRP levels
STABILITY, Darapladib 160 mg daily + ≥18 years; 15,828 LDL 80 (median) 44 N/A Cardiovascular death, MI or 9.7% event rate in No benefit of darapladib in stable
2014 [97] OMT versus OMT established CVD stroke intervention group; CAD
or T2DM with 10.4% in placebo; HR
documented 0.94 (0.85–1.03)
CAD
SOLID-TIMI 52, Darapladib 160 mg daily + ≥18 years; 13,026 LDL 75; HDL 42; 30 N/A Death from CAD, non-fatal 16.3% event rate in the Darapladib was associated with
2014 [98] OMT versus OMT recent ACS TG 135 MI, or urgent coronary intervention group; increased odor-related concerns
( 30 days) revascularization 15.6% with placebo; HR and diarrhea (10.6% vs 5.6%)
1.00 (0.91–1.09)

75
76 R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81

Table 3
Ongoing outcome-driven phase 3 trials.

Trial Drug therapy Population Estimated Time frame Start Primary outcome Estimated
(reference) enrollment (years) date completion
date

IMPROVE-IT [12] Ezetimibe 10 mg + ≥18 years; recent ACS; LDL-C 18,141 Minimum October All-cause death, MI, stroke September 2014
simvastatin 40 mg 125 mg/dL (not on statin) or (completed) follow-up of 2005
(single tablet) versus LDL-C 100 mg/dL (on statin 2.5 years for
simvastatin 40 mg therapy) each patient
REVEAL [79] Anacetrapib 100 mg daily ≥50 years; established ASCVD 30,000 4 June 2011 Major coronary event January 2017
(MI, ischemic stroke, carotid (coronary death, myocardial
revascularization, PAD) or infarction or coronary
T2DM and symptomatic CAD revascularization)
ACCELERATE [80] Evacetrapib 130 mg daily ≥18 years; established ASCVD 12,000 4 October CV death, MI, stroke, January 2016
(ACS, cerebrovascular, PAD) or 2012 coronary revascularization or
T2DM with documented CAD hospitalization for unstable
angina
FOURIER [28] Evolocumab (AMG145) 40–85 years; established ASCVD 22,500 5 January Cardiovascular death, MI, February 2018
every 2 weeks or monthly (ACS, cerebrovascular, PAD); 2013 coronary revascularization,
LDL-C ≥ 70 mg/dL OR non- hospitalization for unstable
HDL-C ≥ 100 mg/dL; fasting angina, stroke
triglycerides ≤ 400 mg/dL
ODYSSEY Alirocumab (SAR236553) ≥40 years; recent ACS 18,000 Up to 6 years October Cardiovascular death, MI or January 2018
OUTCOMES [29] 75 mg every 2 weeks (b52 weeks) 2012 stroke, hospitalization for
unstable angina
SPIRE 1 [30] Bococizumab ≥18 years; on background 12,000 5 October Cardiovascular death, MI or August 2017
(PF-04950615) 150 mg lipid lowering treatment; 2013 stroke, hospitalization for
every 2 weeks LDL-C ≥ 70 and 100 mg/dL unstable angina needing
OR non-HDL-C ≥ 100 urgent revascularization
and 130 mg/dL
SPIRE 2 [31] Bococizumab ≥18 years; on background 6300 5 October Cardiovascular death, MI or August 2017
(PF-04950615) 150 mg lipid lowering treatment; 2013 stroke, hospitalization for
every 2 weeks LDL-C ≥ 100 mg/dL OR unstable angina needing
non-HDL-C ≥ 130 mg/dL urgent revascularization
CANTOS [104] Canakinumab 50/150/300 ≥18 years; MI in the last 17,200 3 April 2011 Cardiovascular death, MI or April 2017
mg every 3 months 30 days; hs-CRP N 2 mg/L stroke
CIRT [105] Methotrexate 15–20 mg ≥18 years; MI in the last 7000 Up to 6 years April 2013 Cardiovascular death, MI or December 2018
weekly plus 1 mg folic acid 5 years; T2DM or metabolic stroke
6 days/week syndrome

2.5. Targeting HDL from macrophages, an index of HDL function [65], is impaired in
humans with ASCVD and dysfunctional oxidized HDL particles [66].
The epidemiological association between low HDL-C and increased Myeloperoxidase (MPO) has a definite role on the immune phago-
risk of CVD in the general population is strong and consistent [52]. How- cytic response but this enzyme also represents a potential link between
ever, Mendelian randomization studies using variants in genes control- inflammation and atherosclerosis. Its circulating levels are increased in
ling HDL-C levels, such as the adenosine triphosphate binding cassette stable coronary artery disease and ACS [67,68] and are associated with
A1 (ABCA1) [53] and lecithin-cholesterol acyl transferase (LCAT) [54] cardiovascular mortality [69]. Oxidative modification of apoA-I leading
genes, do not support a causal role for HDL-C in determining CV risk. to impaired HDL function may partially account for the deleterious car-
Furthermore, gene polymorphisms at the CETP and endothelial lipase diovascular effect of MPO [70]. Again, fully functional HDL particles
loci associated with increased HDL-C do not confer cardioprotection might be more important than merely high HDL-C levels.
[55]. Therefore, raising HDL may not uniformly lower CV risk and cur- The clinical impact of pharmacological interventions to increase
rent studies focus on identifying determinants of HDL functionality, plasma HDL-C at present is questionable. Indeed, the available evidence
rather than on merely HDL-C raising. suggests that this therapeutic strategy is either neutral or harmful to our
Furthermore, there is conflicting data from studies assessing the as- patients. Therefore, we would like to see a better characterization of
sociation between low HDL-C and residual CV risk of patients with HDL function in future studies, in order to translate such new evidence
established atherosclerotic disease on intensive statin therapy. A recent to improved therapeutic approaches.
post-hoc analysis of the COURAGE trial showed an increased risk of
MACE in patients with low HDL-C, despite optimal medical therapy in- 2.5.1. Niacin
cluding intensive statin treatment [56]. On the other hand, several stud- Niacin decreases plasma triglycerides, LDL-C, Lp(a) and increases
ies found no association between low HDL-C and MACEs in patients HDL-C by several mechanisms, including the inhibition of adipose tissue
using intensive lipid lowering medication [57–60]. lipolysis and hepatic triglyceride synthesis [71]. While the neutral re-
At present, strategies shift from approaches solely aiming to increase sults of the AIM-HIGH trial are still in our mind (no decrease in cardio-
HDL-C levels to ones that involve a better understanding of HDL func- vascular death, non-fatal MI, hospitalization for ACS or coronary
tion [61,62], through analysis of its functionality and composition in- revascularization, despite significant improvements in HDL-C and tri-
cluding several lipids and proteins [63]. There is a need for robust glyceride levels) [72], the role of niacin in the treatment of hypercholes-
biomarkers of HDL function that could be addressed in future clinical tri- terolemia and CVD prevention suffered another major drawback with
als. For example, in patients receiving high-dose statin therapy, the the results of the Heart Protection Study 2-Treatment of HDL to Reduce
number of HDL particles may be a better marker of residual risk than the Incidence of Vascular Events (HPS2-THRIVE) trial. This large phase
HDL-C or apoA-I levels [64]. Furthermore, cholesterol efflux capacity III trial demonstrated no benefit of adding niacin and laropiprant to
R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81 77

simvastatin (with or without ezetimibe) in reducing MACEs [73], with a 2.6. MicroRNAs
significant excess of serious adverse events (hemorrhagic stroke, infec-
tions, diabetes and myopathy) in the niacin/laropiprant group [74]. MicroRNAs (miRNAs) are short non-coding RNAs that modulate
Interestingly, despite a 20% reduction in LDL-C and 17% increase messenger RNA stability, therefore exerting a post-transcriptional regu-
in HDL-C, no impact on hard outcomes was demonstrated in 25,673 pa- lator role that may represent a novel and promising way of targeting the
tients with a baseline LDL-C of 64 mg/dL. This low baseline LDL-C atherosclerotic process [89]. Indeed, miRNAs seem to play an important
might have potentially hampered the trial (see below “Pathway for suc- but still vaguely characterized role on early atherosclerosis plaque for-
cessful trial design — Power to absolute reduction”). The evidence ob- mation, as well as on plaque rupture and erosion [90]. Current evidence
tained from this trial questions results from previous trials which suggests that a miRNA profile including the combination of several cir-
suggested a potential beneficial role of niacin in CVD prevention [75]. culating miRNAs will probably represent a better approach to identify
vulnerable patients and disease states. For instance, a specific miRNA
2.5.2. Cholesteryl ester transfer protein (CETP) inhibitors profile in patients with ACS has already been described [91].
CETP is a glycoprotein secreted by the liver that mediates lipid trans- Targeting miRNAs may represent a new pharmacological tool to
fer between lipoproteins in plasma [9,76]. CETP participates in the modulate the atherosclerotic process. For example, miRNA-33 a/b re-
indirect pathway of reverse cholesterol transport (RCT) by transferring presses the expression of the cholesterol transporter ABCA1, which is
cholesteryl esters from HDL to VLDL, IDL and LDL, in this way a pivotal regulator of HDL genesis. Inhibition of this mi-RNA in non-
reintroducing cholesterol in the endogenous lipid pathway. Therefore, human primates raised plasma HDL and lowered VLDL triglycerides, a
blocking CETP has the biological rationale to become a promising ther- promising finding for this new type of pharmacological intervention
apeutic target. [92]. At the moment we still have more questions than answers:
Preliminary studies demonstrated that CETP inhibitors substantially which miRNAs should be targeted? Should we target site-specific
increase HDL-C and decrease LDL-C [77], depending on their potency. miRNAs or will it be possible to administer a systemic inhibitor? Clearly,
However, the initial large trials did not demonstrate outcome benefits further studies are strongly encouraged to help us understand the role
with these agents (ILLUMINATE trial with torcetrapib [78] and dal- of miRNAs in atherosclerosis treatment.
OUTCOMES trial with dalcetrapib [60]). The results of several phase III
outcome-driven trials with two new, more potent agents are pending: 3. Inflammation and the atherosclerotic process
the Randomized Evaluation of the Effects of Anacetrapib through
Lipid-modification (REVEAL) [79] and A Study of Evacetrapib in High- Atherosclerosis is a chronic local inflammatory disease characterized
risk Vascular Disease (ACCELERATE) [80]. by a systemic low-grade inflammatory response that is associated with
MACE including MI and stroke [93,94]. The opportunity to modulate the
inflammatory pathway that plays a central role in the transition from a
2.5.3. Apolipoprotein A1 inducers stable to a vulnerable plaque, platelet and coagulation activation and
The upregulation of endogenous synthesis of apolipoprotein A-I thus increasing risk for clinical events is both attractive and feasible.
(apoA-I), the major protein carried on HDL particles, represents another The phospholipase A2 (PLA2) family includes several enzymes in-
potential target to increase HDL-C by generating nascent HDL particles volved in the generation of precursors of proinflammatory mediators
[9,81]. RVX-208 is an orally administered small molecule that (e.g., leukotrienes, eicosanoids, platelet-activating factors) and is also
upregulates apoA-I synthesis through an epigenetic mechanism (RVX- involved in lipoprotein remodeling yielding highly atherogenic oxidized
208 is an inhibitor of BET transcriptional regulators [82]). Patients LDL particles [95].
with coronary artery disease treated for 12 weeks with RVX-208 on Darapladib is an orally active inhibitor of lipoprotein-associated
top of statin therapy showed a dose-dependent increase in apoA-I and PLA2 (Lp-PLA2) that is currently in phase III testing despite an earlier
HDL-C levels [83]. Furthermore, RVX-208 is also associated with an in- neutral study assessing plaque deformability and CRP levels in the Inte-
crease in the number and size of HDL particles [81]. Yet, results of the re- grated Biomarkers and Imaging Study (IBIS)-2 trial (darapladib
cently reported ApoA-I Synthesis Stimulation and Intravascular prevented necrotic core expansion, though) [96]. The recently pub-
Ultrasound for Coronary Atheroma Regression Evaluation (ASSURE) lished results of the Stabilization of Atherosclerotic Plaque by Initiation
trial (323 patients followed for 24 weeks) failed to demonstrate plaque of Darapladib Therapy (STABILITY) trial, including patients with stable
regression as assessed by intravascular ultrasound imaging (IVUS) [84]; CAD, did not reduce time to first CV event [97], but reduced several sec-
however, increases in ApoA1 by RVX-208 were very modest. Since this ondary endpoints. Darapladib also did not significantly reduce CV
is not a validated surrogate endpoint in atherosclerosis trials, future events in patients with recent ACS in the SOLID-TIMI 52 (Stabilization
studies are needed to identify whether there is a role for this class of of Plaques using Darapladib — Thrombolysis in Myocardial Infarction)
agents. Consequently, there is still room for potent apoA-I inducers to trial [98]. Still, analyses focusing on treatment response depending on
be tested for efficacy against ASCVD. baseline levels of Lp-PLA2 activity are pending.
Varespladib targets isoforms IIa, V and X of secretory PLA2 (sPLA2).
2.5.4. HDL mimetics In phase II programs it showed efficacy reducing sPLA2 activity, LDL-C
An intravenous recombinant form of apoA-I Milano (a naturally oc- and CRP [99–101]. However, this drug not only failed to reduce the
curring variant of apoA-I associated with protection from vascular dis- risk of MACE in the Vascular Inflammation Suppression to Treat Acute
ease despite very low levels of HDL-C [85]), ETC-216, showed a Coronary Syndrome for 16 Weeks (VISTA-16) phase III trial, but actually
significant reduction in coronary atheroma volume using IVUS [86]. increased the risk of MI [102]. In line, a previous Mendelian randomiza-
Event-driven trials are needed to clarify the role of this therapy. tion study showed that that sPLA2 is not causally related to ASCVD
The recently published CHI-SQUARE (Can HDL Infusions Significant- [103].
ly Quicken Atherosclerosis Regression) trial was the largest randomized The CANTOS trial is assessing whether inhibition of inflammation by
clinical trial to date testing the efficacy of serial HDL infusions in patients blocking the inflammasome product and inflammatory cytokine IL-1β
with recent ACS [87]. The HDL mimetic consisted of a recombinant with canakinumab (subcutaneously administered fully human mono-
apoA-I combined with two phospholipids. This study missed its primary clonal antibody that selectively neutralizes IL-1β) will reduce cardiovas-
efficacy endpoint of a reduction in total coronary atheroma volume by cular events in 10,000 patients with prior MI [104]. Furthermore, the
IVUS. Therefore, despite the mechanistic potential of HDL mimetics CIRT trial will assess the role of low-dose methotrexate in the secondary
[88], further research is needed to clarify the role of these agents in ath- cardiovascular prevention of patients with prior MI and either metabol-
erosclerosis treatment. ic syndrome or type 2 diabetes mellitus [105]. These eagerly awaited
78 R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81

results may shed light on the role of targeting inflammation to prevent of “hard” endpoints for surrogate markers to demonstrate potential
cardiovascular events. clinical benefit has emerged as a promising trial perspective.
The intriguing idea of developing immunity to atherosclerosis As reviewed in this paper, the most recent drugs in development for
derives from the growing body of evidence targeting several atherosclerosis target functional (e.g., inflammation) and cellular
atherosclerosis-related antigens using active immunization or antibody (e.g., cholesterol efflux) processes (Fig. 1). In this way, imaging modali-
infusion [106]. Epitopes of apoB, the only protein permanently associat- ties that allow the direct visualization of atherosclerotic plaque
ed with LDL particles, are targeted in the development of pharmacolog- progression, remodeling and inflammation are an attractive way to
ical tools to induce atheroprotective immune responses. Indeed, mechanistically strengthen the clinical development process of a novel
elevated circulating levels of anti-apoB IgG were found to be inversely therapeutic strategy [116].
related to the severity of coronary atherosclerosis and associated with The most common invasive imaging techniques to evaluate coro-
lower risk of MI [107]. Further research is needed to refine basic re- nary atherosclerotic lesions are IVUS and optical coherence tomography
search data and pave the way for future studies. (OCT), both permitting post-procedural plaque characterization [117,
118]. The comprehensive assessment of plaque characteristics fostered
the development of plaque discrimination scores to identify patients
4. New targets for selected populations
with high-risk plaques, in this way being more prone to rupture and
to suffer from a clinical event [119]. Yet the concept of vulnerable plaque
4.1. Genetics and Mendelian randomization studies
is still discussed controversially. Furthermore, during recent years a
growing body of evidence demonstrated the huge potential of non-
Genome-wide association studies (GWAS) assess the association be-
invasive imaging techniques to characterize the complex atherosclerot-
tween genetic variants distributed throughout the genome and a phe-
ic lesion both anatomically and functionally [120]. The most promising
notypic trait [108]. In recent years, these techniques have identified
methods combine an integrated assessment of morphological and func-
gene variants associated with altered lipoprotein metabolism and the
tional plaque characteristics and include PET/CT and PET/MRI. These hy-
risk of CAD [109]. The aim of integrating the large body of genetic data
brid techniques may allow the non-invasive detection of high-risk
to better understand the causes of CVD and find new targets for drug
plaques and assist in the selection of patients at increased risk of MI.
therapy has resulted in novel study designs including Mendelian ran-
We believe they are the most promising techniques to successfully
domization studies aimed at demonstrating causality between the
serve as surrogate endpoints, yet their value has not been firmly
trait and ASCVD [110].
established.
To establish causality in CV disease, the viewpoint from Mendelian
There is a potential role for these imaging methods to facilitate the
randomization begins with epidemiological association and suggests
selection of high-risk patients and also serve as prospectively validated
that if a biomarker is causally associated with CVD risk, then a genetic
surrogate markers. However, additional research is needed to strength-
variant that affects its plasma concentration or activity should itself be
en the available evidence in the complex task translating successful re-
associated with CV risk [110]. A growing body of evidence exploring
sults with imaging endpoints to net clinical benefit. As an example, the
this concept confirmed LDL-C (using PCSK9 nonsense mutations) [17]
validity of cIMT as a surrogate in clinical trials was recently questioned
and Lp(a) [111] as causal risk factors for CVD but refuted the role of
by several meta-analyses showing that cIMT progression was not asso-
CRP [112], HDL-C (using the endothelial-lipase gene LIPG, as well as
ciated with MACEs in the general population [121]. On the other hand,
other single nucleotide polymorphisms) [55] and homocysteine [113]
coronary atheroma volume as assessed by IVUS predicts MACEs in pa-
as causal factors. Furthermore, Mendelian randomization studies em-
tients receiving high-intensity statin therapy [122]. Moreover, robust
phasize the importance of some (e.g., IL-6), but not all (e.g., CRP) inflam-
data comparing OCT information on plaque characteristics to “hard”
matory markers as causal factors for CVD [114,115] (Fig. 2). Clearly, the
outcomes are lacking [123]. Although there is a great potential for
availability of huge cohorts necessary for the complex Mendelian ran-
plaque characterization, no clear and validated criteria have been de-
domization studies will help identify markers of increased CV risk and
fined yet [124].
foster the development of new drug therapies to reduce CVD burden.
5.2. Power to absolute LDL-C reduction
5. Pathways for successful clinical trial design
The absolute event rate in cardiovascular outcomes trials for
5.1. Surrogate endpoints LDL-reduction is related to baseline LDL-C, i.e., the higher the baseline
LDL-C the greater the event rate in the “control” group [125]. Further-
In the era of dual antiplatelet therapy and intensive statin therapy in more, the relative risk reduction (RRR) is related to absolute LDL-C re-
patients with recent ACS, phase III clinical trials with “hard” endpoints duction and not to the percentage of decrease [126], suggesting that
(i.e., death or CV events) require large numbers of patients followed including patients with a higher baseline LDL-C will allow a greater
for several years to be adequately powered, being prohibitively expen- reduction in CV risk. Therefore, trials enrolling patients with a low base-
sive to perform. Numerous hurdles exist considering enrolment and line LDL-C are stalled by (i) lower absolute event rates, demanding
funding of these trials. To overcome these limitations, the substitution more patients and/or longer treatment duration, (ii) lower absolute
LDL-C reduction between groups translating into less risk reduction,
(iii) down-titration or cessation of drug therapy due to “too low”
LDL-C. In this way it is our opinion that future outcome studies
should include patients with a higher LDL-C than 1.8 mmol/L
(70 mg/dL) despite optimal medical therapy (or unable to tolerate
statins) and be powered considering the absolute reduction of LDL-C
in order to avoid missing the primary endpoint.

6. Conclusion

The interest in reducing the residual cardiovascular risk of patients


Fig. 2. Results from Mendelian randomization studies addressing the role of several mol- on currently considered optimal medical therapy is driving the search
ecules as risk factors or biomarkers of cardiovascular disease. for new targets and drugs to modulate the atherosclerotic process. The
R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81 79

latest evidence available confirms that atherosclerosis is dependent on [17] J.C. Cohen, E. Boerwinkle, T.H. Mosley Jr., H.H. Hobbs, Sequence variations in PCSK9,
low LDL, and protection against coronary heart disease, N Engl J Med 354 (2006)
the crucial interplay between lipoprotein metabolism and inflammation 1264–1272.
and represents a complex multifactorial system. Despite the successful [18] F. Raal, R. Scott, R. Somaratne, I. Bridges, G. Li, S.M. Wasserman, et al., Low-density
completion of many phase I and II programs in atherosclerosis, several lipoprotein cholesterol-lowering effects of AMG 145, a monoclonal antibody to
proprotein convertase subtilisin/kexin type 9 serine protease in patients with het-
drugs have recently failed to reduce cardiovascular events in a baseline erozygous familial hypercholesterolemia: the Reduction of LDL-C with PCSK9 Inhi-
population with low LDL-C and a robust background treatment. More bition in Heterozygous Familial Hypercholesterolemia Disorder (RUTHERFORD)
evidence is needed to bring forward solid biomarkers and imaging tech- randomized trial, Circulation 126 (2012) 2408–2417.
[19] E.A. Stein, D. Gipe, J. Bergeron, D. Gaudet, R. Weiss, R. Dufour, et al., Effect of a
niques to smooth the path for successful clinical translation. monoclonal antibody to PCSK9, REGN727/SAR236553, to reduce low-density lipo-
protein cholesterol in patients with heterozygous familial hypercholesterolaemia
on stable statin dose with or without ezetimibe therapy: a phase 2 randomised
controlled trial, Lancet 380 (2012) 29–36.
Conflict of interest
[20] J.M. McKenney, M.J. Koren, D.J. Kereiakes, C. Hanotin, A.C. Ferrand, E.A. Stein, Safety
and efficacy of a monoclonal antibody to proprotein convertase subtilisin/kexin
R.L.L., B.S. and A.L.M. have no conflicts of interest to declare. S.A., type 9 serine protease, SAR236553/REGN727, in patients with primary hypercho-
honoraria and consultancy, advisory board fees, from Astra Zeneca and lesterolemia receiving ongoing stable atorvastatin therapy, J Am Coll Cardiol 59
(2012) 2344–2353.
Thermo Fisher. A.T., grant support from Takeda and Genentech; consult- [21] E.A. Stein, S. Mellis, G.D. Yancopoulos, N. Stahl, D. Logan, W.B. Smith, et al., Effect of
ing Takeda, Actelion, Amgen. E.S., consultant fees in the last 3 years from a monoclonal antibody to PCSK9 on LDL cholesterol, N Engl J Med 366 (2012)
Amgen, BMS, Regeneron/Sanofi, Roche/Genetech, AstraZeneca, 1108–1118.
[22] J.J.P. Kastelein, H.N. Ginsberg, G.G.K.H. Langslet, R. Ceska, R. Dufour, et al., Efficacy
Gemphire, Catabasis and Cymabay. R.J.M., research support from AZ. and safety of alirocumab in patients with heterozygous familial hypercholesterol-
F.Z., grants to institution: Roche Diagnostics; steering committee fees: aemia (heFH) not adequately controlled with current lipid-lowering therapy: Re-
Bayer, Boston Scientific, Janssen, Novartis, Pfizer, Resmed and Takeda; sults of ODYSSEY FH I and FH II studies, 2014.
[23] R.P. Giugliano, N.R. Desai, P. Kohli, W.J. Rogers, R. Somaratne, F. Huang, et al., Effi-
consultant/scientific advisory board: Air Liquide, Amgen, CVRx, Servier, cacy, safety, and tolerability of a monoclonal antibody to proprotein convertase
St Jude, Stealth peptide. W.K., consulting fees from Roche, diaDexus, subtilisin/kexin type 9 in combination with a statin in patients with hypercholes-
Cerenis, Novartis, Amgen, The Medicines Company, Genzyme, terolaemia (LAPLACE-TIMI 57): a randomised, placebo-controlled, dose-ranging,
phase 2 study, Lancet 380 (2012) 2007–2017.
AstraZeneca and Pfizer, honoraria for lectures from AstraZeneca,
[24] M.J. Koren, R. Scott, J.B. Kim, B. Knusel, T. Liu, L. Lei, et al., Efficacy, safety, and tol-
Amgen, Novartis and MSD, and research support from Roche Diagnos- erability of a monoclonal antibody to proprotein convertase subtilisin/kexin type
tics, Abbott, Beckmann and Singulex. 9 as monotherapy in patients with hypercholesterolaemia (MENDEL): a
randomised, double-blind, placebo-controlled, phase 2 study, Lancet 380 (2012)
1995–2006.
[25] E.M. Roth, J.M. McKenney, C. Hanotin, G. Asset, E.A. Stein, Atorvastatin with or
References without an antibody to PCSK9 in primary hypercholesterolemia, N Engl J Med
367 (2012) 1891–1900.
[1] A.D. Lopez, C.D. Mathers, M. Ezzati, D.T. Jamison, C.J. Murray, Global and regional [26] C.P. Cannon, B. Cariou, D. Blom, J. McKenney, C. Lorenzato, R. Pordy, et al., Efficacy
burden of disease and risk factors, 2001: systematic analysis of population health and safety of alirocumab in high cardiovascular risk patients with inadequately
data, Lancet 367 (2006) 1747–1757. controlled hypercholesterolaemia on maximally tolerated daily statin: results
[2] U. Laufs, W.S. Weintraub, C.J. Packard, Beyond statins: what to expect from add-on from the ODYSSEY COMBO II study, 2014.
lipid regulating therapy? Eur Heart J 34 (2013) 2660–2665. [27] D. Sullivan, A.G. Olsson, R. Scott, J.B. Kim, A. Xue, V. Gebski, et al., Effect of a mono-
[3] C. Baigent, A. Keech, P.M. Kearney, L. Blackwell, G. Buck, C. Pollicino, et al., Efficacy clonal antibody to PCSK9 on low-density lipoprotein cholesterol levels in statin-in-
and safety of cholesterol-lowering treatment: prospective meta-analysis of data tolerant patients: the GAUSS randomized trial, JAMA 308 (2012) 2497–2506.
from 90,056 participants in 14 randomised trials of statins, Lancet 366 (2005) [28] U.S. National Institutes of Health, Further Cardiovascular Outcomes Research With
1267–1278. PCSK9 Inhibition in Subjects With Elevated Risk (FOURIER), 2013.
[4] C.P. Cannon, E. Braunwald, C.H. McCabe, D.J. Rader, J.L. Rouleau, R. Belder, et al., In- [29] U.S. National Institutes of Health, ODYSSEY Outcomes: Evaluation of Cardiovascu-
tensive versus moderate lipid lowering with statins after acute coronary syn- lar Outcomes After an Acute Coronary Syndrome During Treatment With
dromes, N Engl J Med 350 (2004) 1495–1504. Alirocumab, 2012.
[5] J.C. Fruchart, F. Sacks, M.P. Hermans, G. Assmann, W.V. Brown, R. Ceska, et al., The [30] U.S. National Institutes of Health, The Evaluation Of PF-04950615 (RN316), In Re-
Residual Risk Reduction Initiative: a call to action to reduce residual vascular risk in ducing The Occurrence Of Major Cardiovascular Events In High Risk Subjects
patients with dyslipidemia, Am J Cardiol 102 (2008) 1K–34K. (SPIRE-1), 2013.
[6] N.J. Stone, J. Robinson, A.H. Lichtenstein, C.N. Merz, C.B. Blum, R.H. Eckel, et al., 2013 [31] U.S. National Institutes of Health, The Evaluation Of PF-04950615 (RN316) In Re-
ACC/AHA Guideline on the Treatment of Blood Cholesterol to Reduce Atheroscle- ducing The Occurrence Of Major Cardiovascular Events In High Risk Subjects
rotic Cardiovascular Risk in Adults: A Report of the American College of Cardiolo- (SPIRE-2), 2013.
gy/American Heart Association Task Force on Practice Guidelines, Circulation 129 [32] J.G. Robinson, M. Farnier, M. Krempf, J. Bergeron, G. Luc, M.R. Averna, et al., Long-
(25 Suppl. 2) (2013) S1–S45. term safety, tolerability and efficacy of alirocumab versus placebo in high cardio-
[7] P. Kirchhof, K.R. Sipido, M.R. Cowie, T. Eschenhagen, K.A. Fox, H. Katus, et al., The vascular risk patients: first results from the ODYSSEY LONG TERM study in 2,341
continuum of personalized cardiovascular medicine: a position paper of the Euro- patients, 2014.
pean Society of Cardiology, Eur Heart J 35 (2014) 3250–3257. [33] D.J. Blom, T. Hala, M. Bolognese, M.J. Lillestol, P.D. Toth, L. Burgess, et al., A 52-week
[8] L.J. Wang, B.L. Song, Niemann-Pick C1-Like 1 and cholesterol uptake, Biochim placebo-controlled trial of evolocumab in hyperlipidemia, N Engl J Med 370 (2014)
Biophys Acta 1821 (2012) 964–972. 1809–1819.
[9] I. Ramasamy, Recent advances in physiological lipoprotein metabolism, Clin Chem [34] J.G. Robinson, M. Farnier, M. Krempf, J. Bergeron, G. Luc, M. Averna, et al., Efficacy
Lab Med (2013) 1–33. CCLM / FESCC. and Safety of Alirocumab in Reducing Lipids and Cardiovascular Events, N Engl J
[10] E. Dembowski, M.H. Davidson, Statin and ezetimibe combination therapy in car- Med 372 (2015) 1489–1499.
diovascular disease, Curr Opin Endocrinol Diabetes Obes 16 (2009) 183–188. [35] M.S. Sabatine, R.P. Giugliano, S.D. Wiviott, F.J. Raal, D.J. Blom, J. Robinson, et al., Ef-
[11] J.J. Kastelein, F. Akdim, E.S. Stroes, A.H. Zwinderman, M.L. Bots, A.F. Stalenhoef, ficacy and Safety of Evolocumab in Reducing Lipids and Cardiovascular Events, N
et al., Simvastatin with or without ezetimibe in familial hypercholesterolemia, N Engl J Med 372 (2015) 1500–1509.
Engl J Med 358 (2008) 1431–1443. [36] M.M. Hussain, J. Shi, P. Dreizen, Microsomal triglyceride transfer protein and its
[12] C.P. Cannon, IMPROVE-IT Trial: A Comparison of Ezetimibe/Simvastatin versus role in apoB-lipoprotein assembly, J Lipid Res 44 (2003) 22–32.
Simvastatin Monotherapy on Cardiovascular Outcomes After Acute Coronary Syn- [37] J.R. Wetterau, L.P. Aggerbeck, M.E. Bouma, C. Eisenberg, A. Munck, M. Hermier,
dromes, 2014. et al., Absence of microsomal triglyceride transfer protein in individuals with
[13] D. Urban, J. Poss, M. Bohm, U. Laufs, Targeting the proprotein convertase subtilisin/ abetalipoproteinemia, Science 258 (1992) 999–1001.
kexin type 9 for the treatment of dyslipidemia and atherosclerosis, J Am Coll [38] M. Cuchel, E.A. Meagher, H. du Toit Theron, D.J. Blom, A.D. Marais, R.A. Hegele,
Cardiol 62 (2013) 1401–1408. et al., Efficacy and safety of a microsomal triglyceride transfer protein inhibitor
[14] K.R. Feingold, A.H. Moser, J.K. Shigenaga, S.M. Patzek, C. Grunfeld, Inflammation in patients with homozygous familial hypercholesterolaemia: a single-arm,
stimulates the expression of PCSK9, Biochem Biophys Res Commun 374 (2008) open-label, phase 3 study, Lancet 381 (2013) 40–46.
341–344. [39] F.F. Samaha, J. McKenney, L.T. Bloedon, W.J. Sasiela, D.J. Rader, Inhibition of
[15] Z. Tang, L. Jiang, J. Peng, Z. Ren, D. Wei, C. Wu, et al., PCSK9 siRNA suppresses the microsomal triglyceride transfer protein alone or with ezetimibe in patients
inflammatory response induced by oxLDL through inhibition of NF-kappaB activa- with moderate hypercholesterolemia, Nat Clin Pract Cardiovasc Med 5 (2008)
tion in THP-1-derived macrophages, Int J Mol Med 30 (2012) 931–938. 497–505.
[16] Z. Awan, N.G. Seidah, J.G. MacFadyen, S. Benjannet, D.I. Chasman, P.M. Ridker, et al., [40] M. Cuchel, L.T. Bloedon, P.O. Szapary, D.M. Kolansky, M.L. Wolfe, A. Sarkis, et al., In-
Rosuvastatin, proprotein convertase subtilisin/kexin type 9 concentrations, and hibition of microsomal triglyceride transfer protein in familial hypercholesterol-
LDL cholesterol response: the JUPITER trial, Clin Chem 58 (2012) 183–189. emia, N Engl J Med 356 (2007) 148–156.
80 R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81

[41] M. Shiomi, T. Ito, MTP inhibitor decreases plasma cholesterol levels in LDL recep- [66] B. Shao, C. Tang, A. Sinha, P.S. Mayer, G.D. Davenport, N. Brot, et al., Humans with
tor-deficient WHHL rabbits by lowering the VLDL secretion, Eur J Pharmacol 431 atherosclerosis have impaired ABCA1 cholesterol efflux and enhanced high-densi-
(2001) 127–131. ty lipoprotein oxidation by myeloperoxidase, Circ Res 114 (2014) 1733–1742.
[42] K. Ueshima, H. Akihisa-Umeno, A. Nagayoshi, S. Takakura, M. Matsuo, S. Mutoh, [67] E. Cavusoglu, C. Ruwende, C. Eng, V. Chopra, S. Yanamadala, L.T. Clark, et al., Useful-
Implitapide, a microsomal triglyceride transfer protein inhibitor, reduces progres- ness of baseline plasma myeloperoxidase levels as an independent predictor of
sion of atherosclerosis in apolipoprotein E knockout mice fed a Western-type diet: myocardial infarction at two years in patients presenting with acute coronary syn-
involvement of the inhibition of postprandial triglyceride elevation, Biol Pharm drome, Am J Cardiol 99 (2007) 1364–1368.
Bull 28 (2005) 247–252. [68] G. Ndrepepa, S. Braun, J. Mehilli, N. von Beckerath, A. Schomig, A. Kastrati,
[43] C.E. Chandler, D.E. Wilder, J.L. Pettini, Y.E. Savoy, S.F. Petras, G. Chang, et al., Myeloperoxidase level in patients with stable coronary artery disease and acute
CP-346086: an MTP inhibitor that lowers plasma cholesterol and triglycer- coronary syndromes, Eur J Clin Invest 38 (2008) 90–96.
ides in experimental animals and in humans, J Lipid Res 44 (2003) [69] H. Scharnagl, M.E. Kleber, B. Genser, S. Kickmaier, W. Renner, G. Weihrauch, et al.,
1887–1901. Association of myeloperoxidase with total and cardiovascular mortality in individ-
[44] E. Kim, S. Campbell, O. Schueller, E. Wong, B. Cole, J. Kuo, et al., A small-molecule uals undergoing coronary angiography–the LURIC study, Int J Cardiol 174 (2014)
inhibitor of enterocytic microsomal triglyceride transfer protein, SLx-4090: bio- 96–105.
chemical, pharmacodynamic, pharmacokinetic, and safety profile, J Pharmacol [70] N. Lu, S. Xie, J. Li, R. Tian, Y.Y. Peng, Myeloperoxidase-mediated oxidation targets
Exp Ther 337 (2011) 775–785. serum apolipoprotein A-I in diabetic patients and represents a potential mecha-
[45] M.M. Hussain, P. Rava, M. Walsh, M. Rana, J. Iqbal, Multiple functions of microsom- nism leading to impaired anti-apoptotic activity of high density lipoprotein, Clin
al triglyceride transfer protein, Nutr Metab 9 (2012) 14. Chim Acta 441 (2015) 163–170.
[46] C.F. Bennett, E.E. Swayze, RNA targeting therapeutics: molecular mechanisms of [71] M.J. Chapman, W. Le Goff, M. Guerin, A. Kontush, Cholesteryl ester transfer protein:
antisense oligonucleotides as a therapeutic platform, Annu Rev Pharmacol Toxicol at the heart of the action of lipid-modulating therapy with statins, fibrates, niacin,
50 (2010) 259–293. and cholesteryl ester transfer protein inhibitors, Eur Heart J 31 (2010) 149–164.
[47] F.J. Raal, R.D. Santos, D.J. Blom, A.D. Marais, M.J. Charng, W.C. Cromwell, et al., [72] W.E. Boden, J.L. Probstfield, T. Anderson, B.R. Chaitman, P. Desvignes-Nickens, K.
Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL choles- Koprowicz, et al., Niacin in patients with low HDL cholesterol levels receiving in-
terol concentrations in patients with homozygous familial hypercholesterolaemia: tensive statin therapy, N Engl J Med 365 (2011) 2255–2267.
a randomised, double-blind, placebo-controlled trial, Lancet 375 (2010) [73] HPS2-THRIVE Collaborative Group, M.J. Landray, R. Haynes, J.C. Hopewell, S. Parish,
998–1006. T. Aung, et al., Effects of extended-release niacin with laropiprant in high-risk pa-
[48] E.A. Stein, R. Dufour, C. Gagne, D. Gaudet, C. East, J.M. Donovan, et al., Apolipopro- tients, N Engl J Med 371 (2014) 203–212.
tein B synthesis inhibition with mipomersen in heterozygous familial hypercholes- [74] HPS2-THRIVE Collaborative Group, HPS2-THRIVE randomized placebo-controlled
terolemia: results of a randomized, double-blind, placebo-controlled trial to assess trial in 25 673 high-risk patients of ER niacin/laropiprant: trial design, pre-speci-
efficacy and safety as add-on therapy in patients with coronary artery disease, Cir- fied muscle and liver outcomes, and reasons for stopping study treatment, Eur
culation 126 (2012) 2283–2292. Heart J 34 (2013) 1279–1291.
[49] M.P. McGowan, J.C. Tardif, R. Ceska, L.J. Burgess, H. Soran, I. Gouni-Berthold, et al., [75] E. Bruckert, J. Labreuche, P. Amarenco, Meta-analysis of the effect of nicotinic acid
Randomized, placebo-controlled trial of mipomersen in patients with severe hy- alone or in combination on cardiovascular events and atherosclerosis, Atheroscle-
percholesterolemia receiving maximally tolerated lipid-lowering therapy, PLoS rosis 210 (2010) 353–361.
One 7 (2012) e49006. [76] P.J. Barter, H.B. Brewer Jr., M.J. Chapman, C.H. Hennekens, D.J. Rader, A.R. Tall,
[50] I.S.I.S. Pharmaceuticals, ISIS-APOCIII Rx, 2014. Cholesteryl ester transfer protein: a novel target for raising HDL and inhibiting ath-
[51] I.S.I.S. Pharmaceuticals, ISIS-APO(a)Rx, 2013. erosclerosis, Arterioscler Thromb Vasc Biol 23 (2003) 160–167.
[52] E. Di Angelantonio, N. Sarwar, P. Perry, S. Kaptoge, K.K. Ray, A. Thompson, et al., [77] P.J. Barter, J.J. Kastelein, Targeting cholesteryl ester transfer protein for the preven-
Major lipids, apolipoproteins, and risk of vascular disease, JAMA 302 (2009) tion and management of cardiovascular disease, J Am Coll Cardiol 47 (2006)
1993–2000. 492–499.
[53] R. Frikke-Schmidt, B.G. Nordestgaard, M.C. Stene, A.A. Sethi, A.T. Remaley, P. [78] P.J. Barter, M. Caulfield, M. Eriksson, S.M. Grundy, J.J. Kastelein, M. Komajda, et al.,
Schnohr, et al., Association of loss-of-function mutations in the ABCA1 gene with Effects of torcetrapib in patients at high risk for coronary events, N Engl J Med
high-density lipoprotein cholesterol levels and risk of ischemic heart disease, 357 (2007) 2109–2122.
JAMA 299 (2008) 2524–2532. [79] U.S. National Institutes of Health, REVEAL: Randomized EValuation of the Effects of
[54] C.L. Haase, A. Tybjaerg-Hansen, A.A. Qayyum, J. Schou, B.G. Nordestgaard, R. Frikke- Anacetrapib Through Lipid-modification, 2010.
Schmidt, LCAT, HDL cholesterol and ischemic cardiovascular disease: a Mendelian [80] U.S. National Institutes of Health, A Study of Evacetrapib in High-Risk Vascular Dis-
randomization study of HDL cholesterol in 54,500 individuals, J Clin Endocrinol ease (ACCELERATE), 2012..
Metab 97 (2012) E248–E256. [81] D. Bailey, R. Jahagirdar, A. Gordon, A. Hafiane, S. Campbell, S. Chatur, et al.,
[55] B.F. Voight, G.M. Peloso, M. Orho-Melander, R. Frikke-Schmidt, M. Barbalic, M.K. RVX-208: a small molecule that increases apolipoprotein A-I and high-den-
Jensen, et al., Plasma HDL cholesterol and risk of myocardial infarction: a mende- sity lipoprotein cholesterol in vitro and in vivo, J Am Coll Cardiol 55 (2010)
lian randomisation study, Lancet 380 (2012) 572–580. 2580–2589.
[56] S. Acharjee, W.E. Boden, P.M. Hartigan, K.K. Teo, D.J. Maron, S.P. Sedlis, et al., Low [82] S. Picaud, C. Wells, I. Felletar, D. Brotherton, S. Martin, P. Savitsky, et al., RVX-208,
levels of high-density lipoprotein cholesterol and increased risk of cardiovascular an inhibitor of BET transcriptional regulators with selectivity for the second
events in stable ischemic heart disease patients: A post-hoc analysis from the bromodomain, Proc Natl Acad Sci U S A 110 (2013) 19754–19759.
COURAGE Trial (Clinical Outcomes Utilizing Revascularization and Aggressive [83] S.J. Nicholls, A. Gordon, J. Johansson, K. Wolski, C.M. Ballantyne, J.J. Kastelein, et al.,
Drug Evaluation), J Am Coll Cardiol 62 (2013) 1826–1833. Efficacy and safety of a novel oral inducer of apolipoprotein a-I synthesis in statin-
[57] A.P. van de Woestijne, Y. van der Graaf, A.H. Liem, M.J. Cramer, J. Westerink, F.L. treated patients with stable coronary artery disease a randomized controlled trial, J
Visseren, Low high-density lipoprotein cholesterol is not a risk factor for recurrent Am Coll Cardiol 57 (2011) 1111–1119.
vascular events in patients with vascular disease on intensive lipid-lowering med- [84] S.J. Nicholls, C.M. Ballantyne, P.J. Barter, H.B. Brewer, J.J.P. Kastelein, A. Gordon,
ication, J Am Coll Cardiol 62 (2013) 1834–1841. et al., Effect of on Oral Agent Inducing ApoA-I Synthesis on Progression of Coronary
[58] P.M. Ridker, J. Genest, S.M. Boekholdt, P. Libby, A.M. Gotto, B.G. Nordestgaard, et al., Atherosclerosis: Results of the ASSURE Study, 2013.
HDL cholesterol and residual risk of first cardiovascular events after treatment [85] C.R. Sirtori, L. Calabresi, G. Franceschini, D. Baldassarre, M. Amato, J. Johansson,
with potent statin therapy: an analysis from the JUPITER trial, Lancet 376 (2010) et al., Cardiovascular status of carriers of the apolipoprotein A-I(Milano) mutant:
333–339. the Limone sul Garda study, Circulation 103 (2001) 1949–1954.
[59] K.K. Ray, C.P. Cannon, R. Cairns, D.A. Morrow, P.M. Ridker, E. Braunwald, Prognostic [86] S.E. Nissen, T. Tsunoda, E.M. Tuzcu, P. Schoenhagen, C.J. Cooper, M. Yasin, et al., Ef-
utility of apoB/AI, total cholesterol/HDL, non-HDL cholesterol, or hs-CRP as predic- fect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with
tors of clinical risk in patients receiving statin therapy after acute coronary syn- acute coronary syndromes: a randomized controlled trial, JAMA 290 (2003)
dromes: results from PROVE IT-TIMI 22, Arterioscler Thromb Vasc Biol 29 (2009) 2292–2300.
424–430. [87] J.C. Tardif, C.M. Ballantyne, P. Barter, J.L. Dasseux, Z.A. Fayad, M.C. Guertin, et al., Ef-
[60] G.G. Schwartz, A.G. Olsson, M. Abt, C.M. Ballantyne, P.J. Barter, J. Brumm, et al., Ef- fects of the high-density lipoprotein mimetic agent CER-001 on coronary athero-
fects of dalcetrapib in patients with a recent acute coronary syndrome, N Engl J sclerosis in patients with acute coronary syndromes: a randomized trial, Eur
Med 367 (2012) 2089–2099. Heart J 35 (2014) 3277–3286.
[61] C. Besler, T.F. Luscher, U. Landmesser, Molecular mechanisms of vascular effects of [88] B.R. Krause, A.T. Remaley, Reconstituted HDL for the acute treatment of acute cor-
High-density lipoprotein: alterations in cardiovascular disease, EMBO Mol Med 4 onary syndrome, Curr Opin Lipidol 24 (2013) 480–486.
(2012) 251–268. [89] K.J. Rayner, C. Fernandez-Hernando, K.J. Moore, MicroRNAs regulating lipid metab-
[62] R.S. Rosenson, H.B. Brewer Jr., B. Ansell, P. Barter, M.J. Chapman, J.W. Heinecke, olism in atherogenesis, Thromb Haemost 107 (2012) 642–647.
et al., Translation of high-density lipoprotein function into clinical practice: current [90] I. Andreou, X. Sun, P.H. Stone, E.R. Edelman, M.W. Feinberg, miRNAs in atheroscle-
prospects and future challenges, Circulation 128 (2013) 1256–1267. rotic plaque initiation, progression, and rupture, Trends Mol Med 21 (2015)
[63] O. Quehenberger, E.A. Dennis, The human plasma lipidome, N Engl J Med 365 307–318.
(2011) 1812–1823. [91] J. Ren, J. Zhang, N. Xu, G. Han, Q. Geng, J. Song, et al., Signature of circulating
[64] S. Mora, R.J. Glynn, P.M. Ridker, High-density lipoprotein cholesterol, size, particle microRNAs as potential biomarkers in vulnerable coronary artery disease, PLoS
number, and residual vascular risk after potent statin therapy, Circulation 128 One 8 (2013) e80738.
(2013) 1189–1197. [92] K.J. Rayner, C.C. Esau, F.N. Hussain, A.L. McDaniel, S.M. Marshall, J.M. van Gils, et al.,
[65] A.V. Khera, M. Cuchel, M. de la Llera-Moya, A. Rodrigues, M.F. Burke, K. Jafri, et al., Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers
Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis, VLDL triglycerides, Nature 478 (2011) 404–407.
N Engl J Med 364 (2011) 127–135. [93] P. Libby, Inflammation in atherosclerosis, Nature 420 (2002) 868–874.
R. Ladeiras-Lopes et al. / International Journal of Cardiology 192 (2015) 72–81 81

[94] C. Weber, H. Noels, Atherosclerosis: current pathogenesis and therapeutic options, [111] P.R. Kamstrup, A. Tybjaerg-Hansen, R. Steffensen, B.G. Nordestgaard, Genetically el-
Nat Med 17 (2011) 1410–1422. evated lipoprotein(a) and increased risk of myocardial infarction, JAMA 301
[95] E. Hurt-Camejo, G. Camejo, H. Peilot, K. Oorni, P. Kovanen, Phospholipase A(2) in (2009) 2331–2339.
vascular disease, Circ Res 89 (2001) 298–304. [112] J. Zacho, A. Tybjaerg-Hansen, J.S. Jensen, P. Grande, H. Sillesen, B.G. Nordestgaard,
[96] P.W. Serruys, H.M. Garcia-Garcia, P. Buszman, P. Erne, S. Verheye, M. Aschermann, Genetically elevated C-reactive protein and ischemic vascular disease, N Engl J
et al., Effects of the direct lipoprotein-associated phospholipase A(2) inhibitor Med 359 (2008) 1897–1908.
darapladib on human coronary atherosclerotic plaque, Circulation 118 (2008) [113] R. Clarke, D.A. Bennett, S. Parish, P. Verhoef, M. Dotsch-Klerk, M. Lathrop, et al., Ho-
1172–1182. mocysteine and coronary heart disease: meta-analysis of MTHFR case-control
[97] H.D. White, C. Held, R. Stewart, E. Tarka, R. Brown, R.Y. Davies, et al., Darapladib for studies, avoiding publication bias, PLoS Med 9 (2012) e1001177.
preventing ischemic events in stable coronary heart disease, N Engl J Med 370 [114] A.D. Hingorani, J.P. Casas, The interleukin-6 receptor as a target for prevention of
(2014) 1702–1711. coronary heart disease: a mendelian randomisation analysis, Lancet 379 (2012)
[98] M.L. O'Donoghue, E. Braunwald, H.D. White, D.P. Steen, M.A. Lukas, E. Tarka, et al., 1214–1224.
Effect of darapladib on major coronary events after an acute coronary syndrome: [115] N. Sarwar, A.S. Butterworth, D.F. Freitag, J. Gregson, P. Willeit, D.N. Gorman, et al.,
the SOLID-TIMI 52 randomized clinical trial, JAMA 312 (2014) 1006–1015. Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-
[99] R.S. Rosenson, C. Hislop, D. McConnell, M. Elliott, Y. Stasiv, N. Wang, et al., Effects of analysis of 82 studies, Lancet 379 (2012) 1205–1213.
1-H-indole-3-glyoxamide (A-002) on concentration of secretory phospholipase A2 [116] R. Duivenvoorden, E. de Groot, E.S. Stroes, J.J. Kastelein, Surrogate markers in clin-
(PLASMA study): a phase II double-blind, randomised, placebo-controlled trial, ical trials–challenges and opportunities, Atherosclerosis 206 (2009) 8–16.
Lancet 373 (2009) 649–658. [117] R. Puri, E.M. Tuzcu, S.E. Nissen, S.J. Nicholls, Exploring coronary atherosclerosis
[100] R.S. Rosenson, M. Elliott, Y. Stasiv, C. Hislop, Randomized trial of an inhibitor of se- with intravascular imaging, Int J Cardiol 168 (2013) 670–679.
cretory phospholipase A2 on atherogenic lipoprotein subclasses in statin-treated [118] S. Agewall, E. DeGroot, P. Marcos-Alberca, J.L. Zamorano, A.A. Barrero, L.P. Badano,
patients with coronary heart disease, Eur Heart J 32 (2011) 999–1005. et al., Ultrasound and radiology surrogate endpoints in pharmacological studies,
[101] R.S. Rosenson, C. Hislop, M. Elliott, Y. Stasiv, M. Goulder, D. Waters, Effects of Atherosclerosis 224 (2012) 12–24.
varespladib methyl on biomarkers and major cardiovascular events in acute coro- [119] S.W. Murray, R.H. Stables, H.M. Garcia-Garcia, A.D. Grayson, M.A. Shaw, R.A. Perry,
nary syndrome patients, J Am Coll Cardiol 56 (2010) 1079–1088. et al., Construction and validation of a plaque discrimination score from the ana-
[102] S.J. Nicholls, J.J. Kastelein, G.G. Schwartz, D. Bash, R.S. Rosenson, M.A. Cavender, tomical and histological differences in coronary atherosclerosis: the Liverpool
et al., Varespladib and cardiovascular events in patients with an acute coronary IVUS-V-HEART (Intra Vascular UltraSound-Virtual-Histology Evaluation of Athero-
syndrome: the VISTA-16 randomized clinical trial, JAMA 311 (2014) 252–262. sclerosis Requiring Treatment) study, EuroIntervention 10 (2014) 815–823.
[103] M.V. Holmes, T. Simon, H.J. Exeter, L. Folkersen, F.W. Asselbergs, M. Guardiola, [120] P.G. Camici, O.E. Rimoldi, O. Gaemperli, P. Libby, Non-invasive anatomic and func-
et al., Secretory phospholipase A(2)-IIA and cardiovascular disease: a mendelian tional imaging of vascular inflammation and unstable plaque, Eur Heart J 33
randomization study, J Am Coll Cardiol 62 (2013) 1966–1976. (2012) 1309–1317.
[104] P.M. Ridker, T. Thuren, A. Zalewski, P. Libby, Interleukin-1beta inhibition and the [121] M.W. Lorenz, J.F. Polak, M. Kavousi, E.B. Mathiesen, H. Völzke, T.P. Tuomainen,
prevention of recurrent cardiovascular events: rationale and design of the et al., Carotid intima-media thickness progression to predict cardiovascular events
Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS), Am in the general population (the PROG-IMT collaborative project): a meta-analysis of
Heart J 162 (2011) 597–605. individual participant data, Lancet 379 (2012) 2053–2062.
[105] B.M. Everett, A.D. Pradhan, D.H. Solomon, N. Paynter, J. Macfadyen, E. Zaharris, [122] R. Puri, S.E. Nissen, M. Shao, C.M. Ballantyne, P.J. Barter, M.J. Chapman, et al., Coro-
et al., Rationale and design of the Cardiovascular Inflammation Reduction Trial: a nary atheroma volume and cardiovascular events during maximally intensive stat-
test of the inflammatory hypothesis of atherothrombosis, Am Heart J 166 (2013) in therapy, Eur Heart J 34 (2013) 3182–3190.
199–207. [123] S. Takarada, T. Imanishi, K. Ishibashi, T. Tanimoto, K. Komukai, Y. Ino, et al., The ef-
[106] G.K. Hansson, J. Nilsson, Vaccination against atherosclerosis? Induction of fect of lipid and inflammatory profiles on the morphological changes of lipid-rich
atheroprotective immunity, Semin Immunopathol 31 (2009) 95–101. plaques in patients with non-ST-segment elevated acute coronary syndrome: fol-
[107] P. Sjogren, G.N. Fredrikson, A. Samnegard, C.G. Ericsson, J. Ohrvik, R.M. Fisher, et al., low-up study by optical coherence tomography and intravascular ultrasound,
High plasma concentrations of autoantibodies against native peptide 210 of apoB- JACC Cardiovasc Interv 3 (2010) 766–772.
100 are related to less coronary atherosclerosis and lower risk of myocardial in- [124] G. van Soest, E. Regar, T.P. Goderie, N. Gonzalo, S. Koljenovic, G.J. van Leenders,
farction, Eur Heart J 29 (2008) 2218–2226. et al., Pitfalls in plaque characterization by OCT: image artifacts in native coronary
[108] S. Burgess, A. Butterworth, S.G. Thompson, Mendelian randomization analysis with arteries, JACC Cardiovasc Imaging 4 (2011) 810–813.
multiple genetic variants using summarized data, Genet Epidemiol 37 (2013) [125] J.G. Robinson, N.J. Stone, Identifying patients for aggressive cholesterol lowering:
658–665. the risk curve concept, Am J Cardiol 98 (2006) 1405–1408.
[109] C.J. Willer, S. Sanna, A.U. Jackson, A. Scuteri, L.L. Bonnycastle, R. Clarke, et al., Newly [126] B. Mihaylova, J. Emberson, L. Blackwell, A. Keech, J. Simes, E.H. Barnes, et al., The ef-
identified loci that influence lipid concentrations and risk of coronary artery dis- fects of lowering LDL cholesterol with statin therapy in people at low risk of vascu-
ease, Nat Genet 40 (2008) 161–169. lar disease: meta-analysis of individual data from 27 randomised trials, Lancet 380
[110] G. Thanassoulis, Mendelian randomization: how genetics is pushing the bound- (2012) 581–590.
aries of epidemiology to identify new causes of heart disease, Can J Cardiol 29
(2013) 30–36.

You might also like