Abstract

Epidemiological research over the last 50 years has discovered a plethora of biomarkers (including molecules, traits or other diseases) that associate with coronary artery disease (CAD) risk. Even the strongest association detected in such observational research precludes drawing conclusions about the causality underlying the relationship between biomarker and disease. Mendelian randomization (MR) studies can shed light on the causality of associations, i.e whether, on the one hand, the biomarker contributes to the development of disease or, on the other hand, the observed association is confounded by unrecognized exogenous factors or due to reverse causation, i.e. due to the fact that prevalent disease affects the level of the biomarker. However, conclusions from a MR study are based on a number of important assumptions. A prerequisite for such studies is that the genetic variant employed affects significantly the biomarker under investigation but has no effect on other phenotypes that might confound the association between the biomarker and disease. If this biomarker is a true causal risk factor for CAD, genotypes of the variant should associate with CAD risk in the direction predicted by the association of the biomarker with CAD. Given a random distribution of exogenous factors in individuals carrying respective genotypes, groups represented by the genotypes are highly similar except for the biomarker of interest. Thus, the genetic variant converts into an unconfounded surrogate of the respective biomarker. This scenario is nicely exemplified for LDL cholesterol. Almost every genotype found to increase LDL cholesterol level by a sufficient amount has also been found to increase CAD risk. Pending a number of conditions that needed to be fulfilled by the genetic variant under investigation (e.g. no pleiotropic effects) and the experimental set-up of the study, LDL cholesterol can be assumed to act as the functional component that links genotypes and CAD risk and, more importantly, it can be assumed that any modulation of LDL cholesterol—by whatever mechanism—would have similar effects on disease risk. Therefore, MR analysis has tremendous potential for identifying therapeutic targets that are likely to be causal for CAD. This review article discusses the opportunities and challenges of MR studies for CAD, highlighting several examples that involved multiple biomarkers, including various lipid and inflammation traits as well as hypertension, diabetes mellitus, and obesity.

Introduction

Identification and therapeutic targeting of causal cardiovascular risk factors such as hypertension and hypercholesterolaemia has translated into enormous improvements in prevention and therapy of coronary artery disease (CAD). Motivated by these fundamental achievements of modern medicine, epidemiological research has investigated hundreds of biomarkers (including circulating molecules, physiological traits, and other diseases) for association with CAD.1,2 Given the strength and reproducibility of many associations, and potential mechanisms linking the biomarker to CAD risk, several of the novel biomarkers were considered to be causal.3,4 Subsequently, programmes were initiated aiming at medical interventions to block the action of such ‘risk factors’, but in many cases the results did not confer the benefit as predicted from the epidemiological observations.5

The large number of biomarkers, the uncertainty about their causal role in the disease process, and the costs of drug development programmes highlight the need to develop methods that allow discrimination between ‘guilt by causation’ from ‘guilt by association’.6 In selected cases, Mendelian randomization (MR) studies may offer such discrimination.

Genomics of biomarkers and cardiovascular disease

Improved genotyping platforms analysing millions of single nucleotide polymorphisms (SNPs) and globally acting consortia involving tens of thousands of patients and controls have provided sufficient genetic information as well as statistical power to uncover even small genetic effects on various phenotypes. Specifically, genome-wide association (GWA) studies have uncovered multiple variants affecting, for example, LDL or HDL cholesterol, triglycerides, C-reactive protein, interleukin-6 (IL6), blood pressure, obesity, and diabetes mellitus, to name a few of the most intensively studied cardiovascular risk markers.7–12 In parallel, the genetic architecture of CAD has also been defined using a similar approach.13

Thereby, recent large-scale genetic research has laid the foundation for MR studies of CAD. Such studies merge genetic information on both biomarkers and disease (CAD) phenotypes. The goal is to unravel whether the risk marker is causally involved in the disease process (Figure 1A–D).14,15

Conceptual background for Mendelian randomization studies: (A) Biomarkers 1–4 are associated with coronary artery disease but causality is unclear. Genetic variants and environmental factors affect the levels of these biomarkers. (B) Here a genetic variant not only associates statistically significant with the biomarker (+), but also with the complex disease. As a DNA variant has no immediate effect on disease manifestation, it can be expected that its effect on the biomarker acts as an indispensible intermediate step. Thus, the biomarker is causally involved in the disease process. (C) Here the genetic variant shows a sizable effect on the biomarker (+) but no association with coronary artery disease. Thus, it can be assumed that an equivalent variability of the biomarker has likewise no effect on disease risk; the biomarker is not causally involved in disease manifestation. (D) In this case exogenous factors influence the biomarker as well as coronary artery disease risk. Even if the genetic variant associates with the biomarker, its causal involvement in coronary artery disease cannot be assumed, since the single nucleotide polymorphism does not associate with coronary artery disease risk.
Figure 1

Conceptual background for Mendelian randomization studies: (A) Biomarkers 1–4 are associated with coronary artery disease but causality is unclear. Genetic variants and environmental factors affect the levels of these biomarkers. (B) Here a genetic variant not only associates statistically significant with the biomarker (+), but also with the complex disease. As a DNA variant has no immediate effect on disease manifestation, it can be expected that its effect on the biomarker acts as an indispensible intermediate step. Thus, the biomarker is causally involved in the disease process. (C) Here the genetic variant shows a sizable effect on the biomarker (+) but no association with coronary artery disease. Thus, it can be assumed that an equivalent variability of the biomarker has likewise no effect on disease risk; the biomarker is not causally involved in disease manifestation. (D) In this case exogenous factors influence the biomarker as well as coronary artery disease risk. Even if the genetic variant associates with the biomarker, its causal involvement in coronary artery disease cannot be assumed, since the single nucleotide polymorphism does not associate with coronary artery disease risk.

Mendel's ‘randomization’ to risk alleles

Most SNPs displaying signals in GWA studies affect quantitatively the level of a phenotype (biomarker) depending on whether an individual had inherited 0, 1, or 2 copies of the variant. As an example, Figure 2 shows the average LDL cholesterol level in a European population, depending on the numbers of a genetic variant (rs2228671) in the LDL cholesterol receptor (LDLR) gene.16 It shows that LDL cholesterol level decreases with the copy number (0, 1, or 2) of the minor allele. The figure also shows that the allele associated with lower LDL cholesterol is indeed likewise associated with a lower risk of CAD.16 Pending a number of conditions that need to be fulfilled by the genetic variant under investigation (see below) and the experimental set-up of the study, such association strongly supports the conclusion that the risk factor (LDL cholesterol) is an intermediary and thus causal step in the observed relationship between the genetic variant and CAD risk.17

The effects of rs2228671 genotypes in the LDL receptor gene on LDL cholesterol (mg/dL) and coronary artery disease risk (% risk change) are shown as assessed by Linsel-Nitschke et al. across different cohorts compromising data from about 9000 individuals.16 The decrease of LDL serum concentration and the decrease in coronary artery disease risk go in parallel the number of T alleles. Since the gene has no other known functions it can be assumed that the LDL increase is causally involved in coronary artery disease.
Figure 2

The effects of rs2228671 genotypes in the LDL receptor gene on LDL cholesterol (mg/dL) and coronary artery disease risk (% risk change) are shown as assessed by Linsel-Nitschke et al. across different cohorts compromising data from about 9000 individuals.16 The decrease of LDL serum concentration and the decrease in coronary artery disease risk go in parallel the number of T alleles. Since the gene has no other known functions it can be assumed that the LDL increase is causally involved in coronary artery disease.

Reverse causation

In epidemiological settings, the direction of observed associations cannot be determined. In this case, MR studies might provide further information to elucidate the mechanisms behind this association. For example, similar to LDL cholesterol, C-reactive protein plasma level strongly associates with CAD risk18 and is affected by multiple genetic variants.19–21 However, in contrast to LDL cholesterol, variants affecting C-reactive protein neither individually nor jointly resulted in an increased risk of CAD.20,21 Thus, other explanations must be taken into account for interpreting the observed epidemiological correlation between increased C-reactive protein levels and CAD. The most likely cause for raised C-reactive protein levels in CAD patients might result from inflammatory processes in atherosclerotic plaques. Alternatively, unrecognized inflammatory diseases might influence C-reactive protein levels and CAD risk in parallel. Hence, in this case genetic variants helped to distinguish between ‘guilt’ and ‘innocence’.

Mendelian randomization studies vs. randomized clinical trials

Mendelian randomization studies and randomized clinical trials (RCTs) share many features.22 In a RCT, randomization should result in an equal distribution of clinical features in the study groups (e.g. age, sex, disease severity, social factors etc.) to minimize the chances of these affecting the outcome of a given intervention. This is best reflected by the baseline characteristics of study participants that ideally should be identical in the intervention and control groups. In other words, randomization should result in comparable patient groups except for the drug-mediated modulation of the biomarker. Likewise, Mendel's second law of independent assortment of alleles should result in an overall equal genetic background of individuals carrying the alleles under investigation. Perhaps more importantly, if cases and controls are drawn from the same population, social and environmental factors should be equally distributed in respective genotype groups. Thereby, groups represented by the respective genotypes should be rather comparable except for the biomarker that is modulated by the genetic variant.

These similarities between the two study designs allow concluding that a MR study can predict the outcome of a RCT that leads to a similar modulation of a biomarker as long as it strictly meets several criteria (see below). The advantage of the MR design is its much lower cost. Indeed, once a population has been genotyped on a genome-wide level, basically every biomarker which is at least in part genetically modulated can be studied in silico by exploration of the data set.

Caveats of Mendelian randomization studies

Pleiotropy

Perhaps the most important limitation to MR is pleiotropy whereby a genetic variant has other effects beyond its effect on the specific biomarker being studied. Such pleiotropy can affect the interpretation of MR studies in multiple ways. First, pleiotropic effects can counteract any effect of the variant on the disease acting via the biomarker, thus giving a null finding even when there is a true causal relationship between biomarker and disease. Alternately, a positive association between the genetic variant and disease may be due to pleiotropic effects and may be mistakenly interpreted as a causal association with the biomarker. As shown in Figure 3 the genetic variant rs964184 displayed genome-wide significant associations with LDL, triglycerides and HDL as well as CAD risk. All associations with the biomarkers could hypothetically explain the increase in CAD risk. Due to such pleiotropic effects it remains unclear which of the lipoproteins actually explains the association with CAD risk.

The genetic variant rs964184 gives an example of pleiotropic effects, which could hypothetically explain the increase in coronary artery disease risk. Due to such effects it remains unclear which of the lipoproteins actually explains the association with coronary artery disease risk.
Figure 3

The genetic variant rs964184 gives an example of pleiotropic effects, which could hypothetically explain the increase in coronary artery disease risk. Due to such effects it remains unclear which of the lipoproteins actually explains the association with coronary artery disease risk.

Such confounding by pleiotropy is least likely where the genetic variant being studied directly lies near the gene for the biomarker under study and affects its level (e.g. C reactive protein gene variants modulating C reactive protein level). However, pleiotropy becomes increasingly possible where the relationship between a variant and the biomarker is more complex (e.g. for a variant in a gene that codes for a protein that is only part of a complex lipoprotein or for a variant that affects a non-protein phenotype such as blood pressure).

Linkage disequilibrium

Genomic loci in close proximity on a given chromosome are usually inherited together. The closer the distance on a chromosome, the higher is the resulting linkage disequilibrium. As an example, a SNP affecting the expression of gene A may be in linkage disequilibrium with a SNP that affects expression of gene B. If the product of gene B is causally related to the disease outcome it would be wrong to conclude that gene A—or the dependent biomarker—is responsible for the phenotype, although such association could be found. Because of this inconsistency of Mendel's second law (independent inheritance of different traits), gene A and its product may be only indirectly associated with the disease phenotype in a MR approach. To avoid potential misinterpretations, it would be ideal to use only SNPs for MR studies that lie in genomic regions without any further proximity to loci that might circumvent the association of the SNP and the disease.

Quantitative effect of single nucleotide polymorphism on biomarker and statistical power

The effect of a SNP on a complex disease can be diluted at least on two levels. First, there are usually multiple genetic variants as well as environmental factors influencing the variability of a biomarker, such that the effect of an individual SNP may be small. Therefore, the variability of the biomarker based on the genetic variant under investigation should be sufficient to affect the disease phenotype, as any further assumptions made in MR studies are based on the quantitative effect of the genetic variant on the presumably intermediary phenotype. Studying single variants comes with the benefit that functional links between variant, affected gene and intermediary phenotype may be more evident limiting the chance for unexpected pleiotropic effects. If the effect of a single genetic variant is too small, additive effects can be studied by analysing several genetic variants in combination.23,24 However, this raises the possibility of potentially adding further pleiotropic effects. Second, any risk factor acts in concert with multiple others and explains only a fraction of the inherited or environmental component underlying a disease such as CAD. Thus, study populations have to be sufficiently large to detect small effects. Moreover, multiple analyses for the CAD association have been carried out on a single CAD GWAS meta-analysis.17,25 As this dataset contains most of current GWAS information on common SNPs and CAD risk, appropriate replications in independent samples are difficult to obtain.

Population stratification

If the population under investigation is not homogenous, but rather based on two or more substrata, any disease that runs at higher prevalence in one of these subpopulations may display association with all SNPs that are predominantly found in this group. This potential limitation merits specific attention if the genetic variant—biomarker—disease relationships are not studied in the same population, which is considered to be the ideal scenario for a MR study. Needing to combine different datasets to achieve enough power, e.g. findings from a GWAS meta-analysis on a biomarker with another GWAS meta-analysis on CAD, brings its own challenges.

Canalization

A potential difference between a genetic variant and clinical biomarker is that the first may affect the biomarker already during childhood (or even earlier) and the second may only be of relevance in the adult individual. Thus, counter-regulatory mechanisms that compensate for the effects related to the SNP in utero or during childhood may blur the association with the genetic variant and the disease. Vice versa, a lifetime exposure to a risk factor may amplify its effects as compared to an epidemiological assessment, as it has been shown for SNPs affecting LDL cholesterol or blood pressure.25,26

Biomarkers, traits and diseases studied for association with coronary artery disease in Mendelian randomization studies

The rapid speed with which MR studies are being applied to CAD is demonstrated by a literature search in PubMed, using the search terms MR and CAD (or myocardial infarction), which produced >110 hits. Considering studies, which involved analysis on the relationship between SNP—biomarker as well as the relationship between SNP—and CAD (with a minimum of 5000 cases), our search identified 20 biomarkers, traits, and diseases. These are summarized in Figure 4 and provide details in Supplementary material online, Table S1.

Brief overview about candidates tested in Mendelian randomization settings. While many biomarkers suggested a causal role in coronary artery disease in Mendelian randomization studies, others disappointed by negative results. The effect of diabetes mellitus single nucleotide polymorphisms was by far smaller than expected and barely significant. Numbers refer to the references in which the Mendelian randomization data have been reported.100–105
Figure 4

Brief overview about candidates tested in Mendelian randomization settings. While many biomarkers suggested a causal role in coronary artery disease in Mendelian randomization studies, others disappointed by negative results. The effect of diabetes mellitus single nucleotide polymorphisms was by far smaller than expected and barely significant. Numbers refer to the references in which the Mendelian randomization data have been reported.100–105

The value of MR studies is best illustrated by adequately powered analyses, in which the effects of a SNP on a biomarker predicted results from large-scale randomized controlled trials, in which a drug modulating the very same biomarker was tested. In this respect, SNPs decreasing serum type secretory phospholipase A2 (sPLA2-IIa) activity displayed no beneficial effects on coronary event rates, which is consistent to what has been shown for varespladip, a clinically tested sPLA2-IIa inhibitor.27 Vice versa, SNPs reducing 3-hydroxy-3-methylglutaryl-coenzyme-A (HMG-CoA) reductase activity—like statins—associated with lower coronary event rates.28 Below we discuss some of the main findings, and for important biomarkers, as well as their implications in terms of therapeutic targeting.

LDL cholesterol

The causal role for LDL in promoting CAD was well-established long before MR studies were conducted. In fact, the experience from families carrying LDL receptor mutations documented that markedly increased LDL cholesterol levels increase CAD risk.29 The same conclusion can be drawn from multiple epidemiological and interventional studies and—more recently—from MR studies.7,16,17

As clinical trials often follow their patients only for a few years,30,31 a particular advantage of the MR approach is that it can provide information on the impact of a lifetime modulation of a biomarker.7,16,17 In this respect, MR studies demonstrated that frequent variants in the LDL receptor gene, which increase LDL cholesterol already in childhood by 4 mg/dL, result in stronger effects on CAD risk than predicted by epidemiological or clinical studies for such degree of LDL variability.16 Vice versa, individuals carrying a rare PCSK9 allele, which lowers LDL way below population average (by 21–38 mg/dL), showed a marked 40–80% reduced incidence of myocardial infarction.26 Such MR studies clearly demonstrated the biological relevance of the biomarker LDL without performing a long-term interventional study. Moreover, the genetic studies encouraged the development of new drugs, including monoclonal antibodies against PCSK9, which displayed remarkable effects on LDL cholesterol levels and are now being tested in clinical trials for their potential to decrease coronary risk.32–34

HDL cholesterol

Another long-standing drug target has been HDL cholesterol, since a low HDL cholesterol level has been a widely replicated biomarker for CAD risk.35 However, despite consistent evidence from prospective cohort studies and extensive experimental research, a causal interference between HDL cholesterol and CAD has never been proven. Great efforts have been made to medically increase HDL cholesterol level, for example, by CETP-inhibitors, fibrates or nicotinic acid,36 but no consistent effects on CAD prevention have been shown for any of these drugs. Several genetic variants have been identified by GWA studies to influence HDL cholesterol concentrations in plasma.7 A MR project conducted in Copenhagen used a single SNP, rs4986970 in the LCAT gene, which was associated with a 13% decrease of HDL cholesterol but showed no association with MI risk.37 Comparable results were obtained for four SNPs encoding for apolipoprotein-A-I, a major component of HDL.38 Voight and colleagues recently reported that carriers of another SNP in the endothelial lipase gene, Asn396Ser, had higher HDL cholesterol (0.14 mmol/L or 3 mg/dL compared with non-carriers) but no reduction in incident CAD (OR: 0.99, 95% CI: 0.88–1.11, P = 0.85). In contrast, an increase of LDL cholesterol per 1 SD based on a respective change mediated by LDL SNPs in a genetic risk score, had profound effects on incident CAD (OR: 2.13, 95% CI: 1.69–2.69, P = 2 × 10−10).17 Given these consistent MR results with plasma HDL cholesterol, attempts to decrease CAD risk by measures solely aimed at raising plasma HDL cholesterol may prove futile. Indeed, recent studies using two CETP inhibitors that have markedly raised plasma HDL level have not shown any benefit in terms of CAD risk reduction.5

Cholesteryl ester transfer protein

Cholesteryl ester transfer protein facilitates the transfer of cholesteryl esters from HDL to LDL cholesterol and it is activity affects plasma concentrations of the two lipoproteins in an inverse manner.39,40 In Japanese families, a splicing defect of the CETP gene resulted in CETP deficiency and increased HDL cholesterol levels.41 Genetic studies on common SNPs revealed association between moderate reduction of CETP mass and activity (∼5–10%) and slightly reduced risk for CAD (5%).39,42 In a more recent large-scale MR analysis, Voight et al. found a reduced risk of myocardial infarction by 4% for a variant at the CETP locus, which increases HDL and decreases LDL cholesterol levels. Whether the observed effects on coronary events are due to the modulation of HDL or LDL cholesterol, or other effects mediated by CETP activity, remains obscure at this time.17

So far, four CETP inhibitors—Torcetrapib, Dalcetrapib, Anacetrapib, and Evacetrapib—have been tested in large RCTs.43 Torcetrapib and Dalcetrapib failed to reduce the incidence of CAD.5

In fact, Torcetrapib increased the rate of coronary events, potentially mediated by a pleiotropic drug effect, namely an increase in blood pressure.5 The remaining agents in this class being tested are Anacetrapib and Evacetrapib. In contrast to Torcetrapib, these drugs have more profound effects on LDL levels.44 Thus, these agents cannot address the effects of an isolated change of HDL on CAD risk. Moreover, at present time it remains open as to whether MR and CRT studies are congruent in showing that lower CETP activity goes along with a mild beneficial effect on coronary event rates.

Lipoprotein (a)

Lipoprotein (a) [LP(a)] has been shown to associate with CAD in both cross-sectional and longitudinal studies.45,46 With the ability of delivering cholesterol to atherosclerotic lesions and promoting thrombosis by inhibiting plasmin, LP(a) represents an interesting target for interventions. However, due to a lack of specific medication a RCT with the intention to lower LP(a) has not been conducted so far. Interestingly, the LP(a) gene locus was among the first for which genome-wide significant association with CAD was established47 and a SNP was found to increase both LP(a) levels and CAD risk.48 A subsequent well-conducted MR study further clearly revealed that LP(a)-associated genetic variants also affect myocardial infarction risk49 such that nowadays there is little doubt that LP(a) is a causal risk factor. Therefore drugs that target plasma LP(a) level, unlike plasma HDL cholesterol level, may prove to be very efficacious in preventing CAD.

Triglycerides

Remnant lipoproteins compromise a heterogeneous group of triglyceride-rich particles such as very low-density lipoptroteins and intermediate-density lipoproteins. These proteins share the ability to accumulate in atherosclerotic plaques and contribute their cholesterol-enriched content to the lesions. Fasting plasma triglyceride concentration was observed to associate with prevalent CAD many years ago.50,51 Non-fasting triglycerides—as a direct marker of elevated remnant cholesterol—also associated with the risk of CAD in prospective cohort studies.52 Bansal et al.53 directly compared fasting with non-fasting triglycerides for predicting CAD and revealed in ∼26 000 American women, after adjustments, that both were associated with risk of CAD.

To test whether elevated non-fasting triglycerides and calculated remnant cholesterol associates with CAD, Jørgensen et al.54 looked in a classical MR setting at a variant in the APOA5 gene which has been shown repeatedly to be one of the strongest genetic determinants of plasma triglyceride levels.7 The variant displayed a significant association with increased non-fasting triglycerides and remnant particles and also an increased odds ratio (1.87, P < 0.001) for the risk of a myocardial infarction. In addition, Sarwar et al.55 assessed the variant rs662799 within the same gene in relation to risk of CAD and found a significant association with coronary heart disease as well [OR: 1.18 (95% CI: 1.11–1.26; P = 2.6 × 10−07)]. Similar findings have been made when multiple SNPs affecting triglyceride levels were studied jointly.56,57 For example, Do et al. found a significant association (P = 1 × 10−9) between triglyceride SNPs and CAD even after adjusting for LDL- and HDL-related effects. Furthermore, they confirmed their finding while restricting the analysis to 44 SNPs with moderate-to-strong effects on triglyceride levels but minimal effect on LDL levels (P = 3 × 10−05 for association between triglycerides and CAD).56 Therefore, the totality of the MR analyses to date indicate that triglycerides are a causal risk factor for CAD although further work needs to be done on triglyceride subtypes regarding their specific risk contribution.

Lipoprotein-associated phospholipase A2

Lipoprotein-associated phospholipase A2 is an enzyme produced mainly by inflammatory cells, including macrophages and lymphocytes. It circulates bound to LDL particles and it produces pro-apoptotic and pro-inflammatory mediators (e.g. precursors of arachidonic acid), which may influence vascular function, growth of atherosclerotic plaques, and inflammation in plaques as well. Furthermore, LP-PLA2 has been described to accumulate in unstable and ruptured plaques.58 In epidemiological settings, LP-PLA2 mass and activity has been found to associate with CAD risk.59–62 However, a gain-of-function mutation in the PLASG7 gene encoding for LP-PLA2 was without effect on coronary atherosclerosis or CHD events in a recent meta-analysis including 26 000 Europeans.63 Likewise, a loss-of-function mutation (V279F) was without consistent effects in a meta-analysis on seven predominantly Asian case–control studies (albeit in south Koreans, this loss-of-function mutation led to a reduced risk of CAD).58 Thus, the majority of genetic data argued against a causal role of LP-PLA2.58,63 Indeed, darapladib, a medication which inhibits LP-PLA2 activity, failed to meet the endpoints in a trial enrolling 16 000 patients with a history of an acute coronary syndrome.64 Such negative effect might have predicted by the MR studies. Another study on darapladib (SOLID TIMI 52) investigates recurrent coronary events in 13 000 patients and is still ongoing.65

Serum type secretory phospholipase A2

Another enzyme, serum type secretory phospholipase A2 (sPLA2-IIa), has been associated with the risk cardiovascular events in prospective settings as well.66,67 Serum type secretory phospholipase A2 has various potential roles in the atherosclerotic process. For example, it hydrolyses phospholipids on lipoproteins, which leads to an increased binding of LDL to preotoglycans in the arterial wall and accelerates the formation of atherosclerotic plaques. Additionally, it enhances the amount of oxidative stress by generating arachidonic acid, lysophospholipids, and non-esterified fatty acids.58 Breitling et al. assessed in a single-cohort approach the association between variants encoding sPLA2-IIa and the serum concentration of sPLA2-IIa and secondary CAD events.68 Holmes et al. conducted a MR meta-analysis involving 19 population studies. Despite the fact that the allele of interest led to a remarkable reduction of sPLA2-IIa enzyme activity and sPLA2-IIa mass, they found no association with incident major vascular events, which raises doubt about a causal role of this enzyme.69 In fact, a trial testing a medication (varespladib in VISTA-16) designed to selectively block sPLA2-IIa was halted for lack of efficacy.27

C-reactive protein

Multiple studies have shown that plasma C-reactive protein level is robustly associated with prevalent and future CAD events.18,70 Moreover, studies employing statins not only demonstrated that elevated C-reactive protein is a good marker for identification of patients with elevated risk but also that a decrease of C-reactive protein levels by this medication goes along with a reduced incidence of cardiovascular events.71 If C-reactive protein acts as a causal factor for CAD, drugs that lower C-reactive protein levels should also reduce the risk of CAD. In fact, such drugs are currently under development.72 However, several large MR studies have now convincingly excluded a role of plasma C-reactive protein level in CAD.19–21,73 Thus, medical lowering of C-reactive protein levels is unlikely to be successful in preventing CAD.

Interleukin-6 receptor

Despite the doubts about C-reactive protein as a target for CAD prevention, inflammatory markers still remain an interesting field in the search for causative factors and novel drug targets. Interleukin 6 as a pro-inflammatory agent binds to the IL-6 receptor, which is located on hepatocytes, monocytes, and the endothelial wall and mediates inflammatory responses. In a prospective study, IL-6 was associated with adverse cardiovascular prognosis.74 In addition, patients with incident CAD had increased concentrations of circulating IL-6 prior to their event such that blockade of IL-6 binding might provide a way to reduce atherosclerosis and future CAD events.75,76 Tocilizumab, a monoclonal IL-6 antibody, has already proved its ability to sufficiently alter the course of rheumatoid arthritis by reducing articular inflammation.77 The question as to whether Tocilizumab can also prevent CAD has not yet been studied in RCTs. The genetics of the IL-6 receptor provides an adequate tool to investigate such association in a MR study. Patients carrying the allele leading to reduced IL-6 binding not only showed a significant lower inflammatory response but also a remarkable reduction in CAD events compared with individuals carrying the alternate allele.78 These findings raise hope that IL-6 receptor inhibition may be suitable for primary or secondary prevention of CAD. The IL-6 receptor variant also showed associations with reduced C-reactive protein and fibrinogen concentrations, most likely due to downstream effects of IL-6 signalling. Thus, although this SNP affects both C-reactive protein and fibrinogen level, it is not suitable to study these biomarkers—rather than IL-6—in a MR study design. This illustrates one of the caveats of MR studies whereby testing and interpretation are most robust when the variant directly affects the gene for the biomarker being evaluated.

Pentraxin 3

Pentraxin 3 (PTX3) has been shown to be involved in vascular inflammation.79 Dubin et al.80 revealed in a prospective study that PTX3 concentrations associate with increased risk for all-cause-mortality, cardiovascular events, and incident heart failure. Similar findings were made in case–control studies.81 Investigating three common PTX3 polymorphisms Barbati et al.82 observed association with PTX3 plasma levels but not with the risk of acute myocardial infarction, suggesting that PTX3 is likely not a causal risk factor.

Fibrinogen

Fibrinogen—with its ability to form fibrin—is the major component of blood clots and thus involved in the manifestation of atherothrombotic events.83 Moreover, fibrinogen affects multiple inflammatory conditions.84 Elevated levels of fibrinogen have been found in patients suffering from CAD and data from prospective studies established association between fibrinogen and risk of coronary heart disease.85 To study a possible causal relationship between the variability of fibrinogen serum levels and CAD, Sabater-Lleal et al.86 conducted a multi-ethnic meta-analysis of genome-wide association studies in over 100 000 subjects. The merged effect of the SNPs increasing fibrinogen showed no evidence for an association with CAD suggesting a lack of causality.

Blood pressure

Based on overwhelming evidence from epidemiological and interventional studies high blood pressure can be considered as a proven causal factor for CAD.87,88 Further evidence comes now from a genetic study.25 Specifically, 30 SNPs have been found to associate with systolic and diastolic blood pressure, each of them leading to an increase of 0.5–1.2 mmHg in systolic blood pressure.10 Analysing data from almost 22 500 CAD cases and 65 000 controls, the CARDIoGRAM investigators found an average increase of CAD risk by 3% per risk allele. Patients in the highest quintile in terms of numbers and effect sizes of blood pressure alleles had a 70% higher odds of having CAD, when compared with patients in the bottom quintile of a genetic risk score distribution.25 These findings were remarkable for the fact that—like LDL SNPs—blood pressure associated SNPs had stronger effects on CAD risk than expected by epidemiological studies. In observational settings, the true risk mediated by blood pressure might be underestimated due to a regression dilution bias that may result from inherent inaccuracies in the measurement of blood pressure. In contrast, a SNP may be a more precise denominator of a small variability in blood pressure as in the nowadays GWAS era this association (between a SNP and blood pressure) stems from ten thousands of measurements. Furthermore, the SNPs under investigation may affect mechanisms, i.e. endothelial function that—aside from blood pressure increase—may have a direct effect on vascular biology. These effects might be more closely related to the development of CAD than the variability in blood pressure itself.10

Body mass index and obesity

Obesity—widely assessed as elevated body mass index (BMI)—has consistently shown to associate with future risk of cardiovascular complications.89 Nevertheless, causality of the association has not yet been proved. Three loci (FTO, MC4R, and TMEM18) with the largest known effect on BMI were tested for their association with ischaemic heart disease in 75 627 individuals demonstrating that a genetically driven increase in BMI of 0.28 kg/m2 per individual allele translates into a significant 3% increase of CAD risk.90 This study adds to the emerging evidence that higher BMI or the condition leading to this play a causal role in the development of CAD. However, there are pleiotropic effects—namely type 2 diabetes—reported at least for FTO and MC4R risk alleles, which might affect the observed associations. Thus, the mechanism linking obesity-related genetic variants and CAD risk might include additional phenotypes. Further investigations are required to unmask such traits.

Diabetes mellitus

Although there is a two to three-fold higher risk of CAD in patients with diabetes, there is an ongoing debate about the exact nature of the relationship.91,92 Moreover, recent interventional studies have raised doubt about causality as aggressive blood glucose lowering failed to sufficiently reduce cardiovascular events.93 Genetic variants might provide an adequate tool to further answer the question of a causal interference. About 40 variants have been found to associate with type 2 diabetes in Western-Europeans. These variants were tested for their association with CAD in CARDIoGRAM. Interestingly, diabetes SNPs had only a mild impact on CAD (the average increase in CAD risk observed per individual type 2 diabetes risk allele was 1.0076, P = 0.02 for OR).94 Albeit this increase was statistically significant (P = 5,8 × 10−5), it contrasted also significantly from the effect that was expected (1.067, P = 7.1 × 10−10 for the difference between observed and expected effects), based on the effects of these alleles on diabetes risk and the effect of diabetes on CAD risk as observed in the Framingham Heart Study.95 This might be in part because of a potential overestimation of the respective SNP effects on diabetes in GWAS (winners curse). Another explanation refers to a hypothetical overestimation of diabetes effects on CAD in epidemiological settings. This is emphasized by recent findings from clinical trials which disappointed in that aggressive blood glucose lowering failed to further decrease coronary events.93 Moreover, when compared with other quantitative risk factors, e.g. genetically mediated high blood pressure or LDL cholesterol levels, type 2 diabetes often manifests in the late adulthood and only then starts to affect the risk of CAD. Such patients may be underrepresented in CARDIoGRAM.96

Telomere length

Several cross-sectional and longitudinal studies have shown an association between shorter mean leucocyte telomere length (LTL) and CAD.97–99 Whether this reflects a casual association or is a confounded association due to the known effect on telomere length of other putative CAD risk factors such as oxidative stress and inflammation has been a matter of some debate. Recently, through GWAS, Codd et al.24 identified seven variants associated with mean LTL, several in genes that are components of the telomerase complex. A genetic risk score analysis combining lead variants at all seven loci in the CARDIoGRAM CAD GWAS meta-analysis showed an association of the alleles associated with shorter LTL with an increased risk of CAD [21% (95% confidence interval, 5–35%) per standard deviation in LTL]. These findings suggest a causal association of shorter telomere length with risk of CAD, the mechanism of which merits further investigation.

Summary

Mendelian randomization has emerged as a valuable approach in investigating whether an association of a biomarker with CAD is casual or not. Already, the evidence points to several long-held candidates (plasma HDL cholesterol level, C-reactive protein) as not being causal. On the other hand the likely causal involvement of other biomarkers [LP(a), IL-6] has been enhanced providing greater confidence that efforts to target them therapeutically will prove rewarding. The instruments for carrying out MR studies are rapidly improving and will be of great benefit for future decision-making upon the development of novel drug targets. However, despite the convincing concept of MR analysis, several limitations and requirements have to be taken into consideration while designing and interpreting a MR study. In this regard, the MR study adds to established study designs (like RCT) without the ability to fully replace them.

Supplementary material

Supplementary material is available at European Heart Journal online.

Funding

This work was supported by the EU-funded Integrated Project CVgenes@target (HEALTH-F2-2013-601456), Transatlantic Networks of Excellence in Cardiovascular Research Program of the Fondation Leducq, and as well as the BMBF-funded German National Genome Network (NGFN-Plus) Project Atherogenomics (FKZ: 01GS0831) and e:AtheroSysMed (FKZ: 01ZX1313A). N.J.S. holds a chair funded by the British Heart Foundation and is a NIHR senior investigator.

References

1
Zakynthinos
E
Pappa
N
Inflammatory biomarkers in coronary artery disease
J Cardiol
2009
, vol. 
3
 (pg. 
317
-
333
)
2
Sun
X
Jia
Z
A brief review of biomarkers for preventing and treating cardiovascular diseases
J Cardiovasc Dis Res
2012
, vol. 
4
 (pg. 
251
-
254
)
3
Bisoendial
RJ
Kastelein
JJP
Levels
JHM
Zwaginga
JJ
van den Bogaard
B
Reitsma
PH
Meijers
JCM
Hartman
D
Levi
M
Stroes
ESG
Activation of inflammation and coagulation after infusion of C-reactive protein in humans
Circ Res
2005
, vol. 
7
 (pg. 
714
-
716
)
4
Tall
AR
An overview of reverse cholesterol transport
Eur Heart J
1998
, vol. 
19
 (pg. 
31
-
35
)
5
Barter
PJ
Caulfield
M
Eriksson
M
Grundy
SM
Kastelein
JJP
Komajda
M
Lopez-Sendon
J
Mosca
L
Tardif
J-C
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
, vol. 
21
 (pg. 
2109
-
2122
)
6
Kannel
WB
Castelli
WP
Gordon
T
Cholesterol in the prediction of atherosclerotic disease. New perspectives based on the Framingham study.
Ann Intern Med
1979
, vol. 
1
 (pg. 
85
-
91
)
7
Teslovich
TM
Musunuru
K
Smith
AV
Edmondson
AC
Stylianou
IM
Koseki
M
Pirruccello
JP
Ripatti
S
Chasman
DI
Willer
CJ
Johansen
CT
Fouchier
SW
Isaacs
A
Peloso
GM
Barbalic
M
Ricketts
SL
Bis
JC
Aulchenko
YS
Thorleifsson
G
Feitosa
MF
Chambers
J
Orho-Melander
M
Melander
O
Johnson
T
Li
X
Guo
X
Li
M
Shin Cho
Y
Jin Go
M
Jin Kim
Y
Lee
JY
Park
T
Kim
K
Sim
X
Twee-Hee Ong
R
Croteau-Chonka
DC
Lange
LA
Smith
JD
Song
K
Hua Zhao
J
Yuan
X
Luan
J
Lamina
C
Ziegler
A
Zhang
W
Zee
RY
Wright
AF
Witteman
JC
Wilson
JF
Willemsen
G
Wichmann
HE
Whitfield
JB
Waterworth
DM
Wareham
NJ
Waeber
G
Vollenweider
P
Voight
BF
Vitart
V
Uitterlinden
AG
Uda
M
Tuomilehto
J
Thompson
JR
Tanaka
T
Surakka
I
Stringham
HM
Spector
TD
Soranzo
N
Smit
JH
Sinisalo
J
Silander
K
Sijbrands
EJ
Scuteri
A
Scott
J
Schlessinger
D
Sanna
S
Salomaa
V
Saharinen
J
Sabatti
C
Ruokonen
A
Rudan
I
Rose
LM
Roberts
R
Rieder
M
Psaty
BM
Pramstaller
PP
Pichler
I
Perola
M
Penninx
BW
Pedersen
NL
Pattaro
C
Parker
AN
Pare
G
Oostra
BA
O'Donnell
CJ
Nieminen
MS
Nickerson
DA
Montgomery
GW
Meitinger
T
McPherson
R
McCarthy
MI
McArdle
W
Masson
D
Martin
NG
Marroni
F
Mangino
M
Magnusson
PK
Lucas
G
Luben
R
Loos
RJ
Lokki
ML
Lettre
G
Langenberg
C
Launer
LJ
Lakatta
EG
Laaksonen
R
Kyvik
KO
Kronenberg
F
König
IR
Khaw
KT
Kaprio
J
Kaplan
LM
Johansson
A
Jarvelin
MR
Janssens
AC
Ingelsson
E
Igl
W
Kees Hovingh
G
Hottenga
JJ
Hofman
A
Hicks
AA
Hengstenberg
C
Heid
IM
Hayward
C
Havulinna
AS
Hastie
ND
Harris
TB
Haritunians
T
Hall
AS
Gyllensten
U
Guiducci
C
Groop
LC
Gonzalez
E
Gieger
C
Freimer
NB
Ferrucci
L
Erdmann
J
Elliott
P
Ejebe
KG
Döring
A
Dominiczak
AF
Demissie
S
Deloukas
P
de Geus
EJ
de Faire
U
Crawford
G
Collins
FS
Chen
YD
Caulfield
MJ
Campbell
H
Burtt
NP
Bonnycastle
LL
Boomsma
DI
Boekholdt
SM
Bergman
RN
Barroso
I
Bandinelli
S
Ballantyne
CM
Assimes
TL
Quertermous
T
Altshuler
D
Seielstad
M
Wong
TY
Tai
ES
Feranil
AB
Kuzawa
CW
Adair
LS
Taylor
HA
Jr
Borecki
IB
Gabriel
SB
Wilson
JG
Holm
H
Thorsteinsdottir
U
Gudnason
V
Krauss
RM
Mohlke
KL
Ordovas
JM
Munroe
PB
Kooner
JS
Tall
AR
Hegele
RA
Kastelein
JJ
Schadt
EE
Rotter
JI
Boerwinkle
E
Strachan
DP
Mooser
V
Stefansson
K
Reilly
MP
Samani
NJ
Schunkert
H
Cupples
LA
Sandhu
MS
Ridker
PM
Rader
DJ
van Duijn
CM
Peltonen
L
Abecasis
GR
Boehnke
M
Kathiresan
S
Biological, clinical and population relevance of 95 loci for blood lipids
Nature
2010
, vol. 
7307
 (pg. 
707
-
713
)
8
Benjamin
EJ
Dupuis
J
Larson
MG
Lunetta
KL
Booth
SL
Govindaraju
DR
Kathiresan
S
Keaney
JF
Jr
Keyes
MJ
Lin
JP
Meigs
JB
Robins
SJ
Rong
J
Schnabel
R
Vita
JA
Wang
TJ
Wilson
PW
Wolf
PA
Vasan
RS
Genome-wide association with select biomarker traits in the Framingham Heart Study
BMC Med Genet
2007
, vol. 
8
 pg. 
S11
 
9
Shah
T
Zabaneh
D
Gaunt
T
Swerdlow
DI
Shah
S
Talmud
PJ
Day
IN
Whittaker
J
Holmes
MV
Sofat
R
Humphries
SE
Kivimaki
M
Kumari
M
Hingorani
AD
Casas
JP
Gene-centric analysis identifies variants associated with interleukin-6 levels and shared pathways with other inflammation markers
Circ Cardiovasc Genet
2013
, vol. 
2
 (pg. 
163
-
170
)
10
Ehret
GB
Munroe
PB
Rice
KM
Bochud
M
Johnson
AD
Chasman
DI
Smith
AV
Tobin
MD
Verwoert
GC
Hwang
SJ
Pihur
V
Vollenweider
P
O'Reilly
PF
Amin
N
Bragg-Gresham
JL
Teumer
A
Glazer
NL
Launer
L
Zhao
JH
Aulchenko
Y
Heath
S
Sõber
S
Parsa
A
Luan
J
Arora
P
Dehghan
A
Zhang
F
Lucas
G
Hicks
AA
Jackson
AU
Peden
JF
Tanaka
T
Wild
SH
Rudan
I
Igl
W
Milaneschi
Y
Parker
AN
Fava
C
Chambers
JC
Fox
ER
Kumari
M
Go
MJ
van der Harst
P
Kao
WH
Sjögren
M
Vinay
DG
Alexander
M
Tabara
Y
Shaw-Hawkins
S
Whincup
PH
Liu
Y
Shi
G
Kuusisto
J
Tayo
B
Seielstad
M
Sim
X
Nguyen
KD
Lehtimäki
T
Matullo
G
Wu
Y
Gaunt
TR
Onland-Moret
NC
Cooper
MN
Platou
CG
Org
E
Hardy
R
Dahgam
S
Palmen
J
Vitart
V
Braund
PS
Kuznetsova
T
Uiterwaal
CS
Adeyemo
A
Palmas
W
Campbell
H
Ludwig
B
Tomaszewski
M
Tzoulaki
I
Palmer
ND
Aspelund
T
Garcia
M
Chang
YP
O'Connell
JR
Steinle
NI
Grobbee
DE
Arking
DE
Kardia
SL
Morrison
AC
Hernandez
D
Najjar
S
McArdle
WL
Hadley
D
Brown
MJ
Connell
JM
Hingorani
AD
Day
IN
Lawlor
DA
Beilby
JP
Lawrence
RW
Clarke
R
Hopewell
JC
Ongen
H
Dreisbach
AW
Li
Y
Young
JH
Bis
JC
Kähönen
M
Viikari
J
Adair
LS
Lee
NR
Chen
MH
Olden
M
Pattaro
C
Bolton
JA
Köttgen
A
Bergmann
S
Mooser
V
Chaturvedi
N
Frayling
TM
Islam
M
Jafar
TH
Erdmann
J
Kulkarni
SR
Bornstein
SR
Grässler
J
Groop
L
Voight
BF
Kettunen
J
Howard
P
Taylor
A
Guarrera
S
Ricceri
F
Emilsson
V
Plump
A
Barroso
I
Khaw
KT
Weder
AB
Hunt
SC
Sun
YV
Bergman
RN
Collins
FS
Bonnycastle
LL
Scott
LJ
Stringham
HM
Peltonen
L
Perola
M
Vartiainen
E
Brand
SM
Staessen
JA
Wang
TJ
Burton
PR
Soler Artigas
M
Dong
Y
Snieder
H
Wang
X
Zhu
H
Lohman
KK
Rudock
ME
Heckbert
SR
Smith
NL
Wiggins
KL
Doumatey
A
Shriner
D
Veldre
G
Viigimaa
M
Kinra
S
Prabhakaran
D
Tripathy
V
Langefeld
CD
Rosengren
A
Thelle
DS
Corsi
AM
Singleton
A
Forrester
T
Hilton
G
McKenzie
CA
Salako
T
Iwai
N
Kita
Y
Ogihara
T
Ohkubo
T
Okamura
T
Ueshima
H
Umemura
S
Eyheramendy
S
Meitinger
T
Wichmann
HE
Cho
YS
Kim
HL
Lee
JY
Scott
J
Sehmi
JS
Zhang
W
Hedblad
B
Nilsson
P
Smith
GD
Wong
A
Narisu
N
Stančáková
A
Raffel
LJ
Yao
J
Kathiresan
S
O'Donnell
CJ
Schwartz
SM
Ikram
MA
Longstreth
WT
Jr
Mosley
TH
Seshadri
S
Shrine
NR
Wain
LV
Morken
MA
Swift
AJ
Laitinen
J
Prokopenko
I
Zitting
P
Cooper
JA
Humphries
SE
Danesh
J
Rasheed
A
Goel
A
Hamsten
A
Watkins
H
Bakker
SJ
van Gilst
WH
Janipalli
CS
Mani
KR
Yajnik
CS
Hofman
A
Mattace-Raso
FU
Oostra
BA
Demirkan
A
Isaacs
A
Rivadeneira
F
Lakatta
EG
Orru
M
Scuteri
A
Ala-Korpela
M
Kangas
AJ
Lyytikäinen
LP
Soininen
P
Tukiainen
T
Würtz
P
Ong
RT
Dörr
M
Kroemer
HK
Völker
U
Völzke
H
Galan
P
Hercberg
S
Lathrop
M
Zelenika
D
Deloukas
P
Mangino
M
Spector
TD
Zhai
G
Meschia
JF
Nalls
MA
Sharma
P
Terzic
J
Kumar
MV
Denniff
M
Zukowska-Szczechowska
E
Wagenknecht
LE
Fowkes
FG
Charchar
FJ
Schwarz
PE
Hayward
C
Guo
X
Rotimi
C
Bots
ML
Brand
E
Samani
NJ
Polasek
O
Talmud
PJ
Nyberg
F
Kuh
D
Laan
M
Hveem
K
Palmer
LJ
van der Schouw
YT
Casas
JP
Mohlke
KL
Vineis
P
Raitakari
O
Ganesh
SK
Wong
TY
Tai
ES
Cooper
RS
Laakso
M
Rao
DC
Harris
TB
Morris
RW
Dominiczak
AF
Kivimaki
M
Marmot
MG
Miki
T
Saleheen
D
Chandak
GR
Coresh
J
Navis
G
Salomaa
V
Han
BG
Zhu
X
Kooner
JS
Melander
O
Ridker
PM
Bandinelli
S
Gyllensten
UB
Wright
AF
Wilson
JF
Ferrucci
L
Farrall
M
Tuomilehto
J
Pramstaller
PP
Elosua
R
Soranzo
N
Sijbrands
EJ
Altshuler
D
Loos
RJ
Shuldiner
AR
Gieger
C
Meneton
P
Uitterlinden
AG
Wareham
NJ
Gudnason
V
Rotter
JI
Rettig
R
Uda
M
Strachan
DP
Witteman
JC
Hartikainen
AL
Beckmann
JS
Boerwinkle
E
Vasan
RS
Boehnke
M
Larson
MG
Järvelin
MR
Psaty
BM
Abecasis
GR
Chakravarti
A
Elliott
P
van Duijn
CM
Newton-Cheh
C
Levy
D
Caulfield
MJ
Johnson
T
International Consortium for Blood Pressure Genome-Wide Association Studies; CARDIoGRAM consortium; CKDGen Consortium; KidneyGen Consortium; EchoGen consortium; CHARGE-HF consortium
Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk
Nature
2011
, vol. 
7367
 (pg. 
103
-
109
)
11
Mägi
R
Manning
S
Yousseif
A
Pucci
A
Santini
F
Karra
E
Querci
G
Pelosini
C
McCarthy
MI
Lindgren
CM
Batterham
RL
Contribution of 32 GWAS-identified common variants to severe obesity in European adults referred for bariatric surgery
Plos One
2013
, vol. 
8
 pg. 
70735
 
12
Morris
AP
Voight
BF
Teslovich
TM
Ferreira
T
Segrè
AV
Steinthorsdottir
V
Strawbridge
RJ
Khan
H
Grallert
H
Mahajan
A
Prokopenko
I
Kang
HM
Dina
C
Esko
T
Fraser
RM
Kanoni
S
Kumar
A
Lagou
V
Langenberg
C
Luan
J
Lindgren
CM
Müller-Nurasyid
M
Pechlivanis
S
Rayner
NW
Scott
LJ
Wiltshire
S
Yengo
L
Kinnunen
L
Rossin
EJ
Raychaudhuri
S
Johnson
AD
Dimas
AS
Loos
RJ
Vedantam
S
Chen
H
Florez
JC
Fox
C
Liu
CT
Rybin
D
Couper
DJ
Kao
WH
Li
M
Cornelis
MC
Kraft
P
Sun
Q
van Dam
RM
Stringham
HM
Chines
PS
Fischer
K
Fontanillas
P
Holmen
OL
Hunt
SE
Jackson
AU
Kong
A
Lawrence
R
Meyer
J
Perry
JR
Platou
CG
Potter
S
Rehnberg
E
Robertson
N
Sivapalaratnam
S
Stančáková
A
Stirrups
K
Thorleifsson
G
Tikkanen
E
Wood
AR
Almgren
P
Atalay
M
Benediktsson
R
Bonnycastle
LL
Burtt
N
Carey
J
Charpentier
G
Crenshaw
AT
Doney
AS
Dorkhan
M
Edkins
S
Emilsson
V
Eury
E
Forsen
T
Gertow
K
Gigante
B
Grant
GB
Groves
CJ
Guiducci
C
Herder
C
Hreidarsson
AB
Hui
J
James
A
Jonsson
A
Rathmann
W
Klopp
N
Kravic
J
Krjutškov
K
Langford
C
Leander
K
Lindholm
E
Lobbens
S
Männistö
S
Mirza
G
Mühleisen
TW
Musk
B
Parkin
M
Rallidis
L
Saramies
J
Sennblad
B
Shah
S
Sigurðsson
G
Silveira
A
Steinbach
G
Thorand
B
Trakalo
J
Veglia
F
Wennauer
R
Winckler
W
Zabaneh
D
Campbell
H
van Duijn
C
Uitterlinden
AG
Hofman
A
Sijbrands
E
Abecasis
GR
Owen
KR
Zeggini
E
Trip
MD
Forouhi
NG
Syvänen
AC
Eriksson
JG
Peltonen
L
Nöthen
MM
Balkau
B
Palmer
CN
Lyssenko
V
Tuomi
T
Isomaa
B
Hunter
DJ
Qi
L
Shuldiner
AR
Roden
M
Barroso
I
Wilsgaard
T
Beilby
J
Hovingh
K
Price
JF
Wilson
JF
Rauramaa
R
Lakka
TA
Lind
L
Dedoussis
G
Njølstad
I
Pedersen
NL
Khaw
KT
Wareham
NJ
Keinanen-Kiukaanniemi
SM
Saaristo
TE
Korpi-Hyövälti
E
Saltevo
J
Laakso
M
Kuusisto
J
Metspalu
A
Collins
FS
Mohlke
KL
Bergman
RN
Tuomilehto
J
Boehm
BO
Gieger
C
Hveem
K
Cauchi
S
Froguel
P
Baldassarre
D
Tremoli
E
Humphries
SE
Saleheen
D
Danesh
J
Ingelsson
E
Ripatti
S
Salomaa
V
Erbel
R
Jöckel
KH
Moebus
S
Peters
A
Illig
T
de Faire
U
Hamsten
A
Morris
AD
Donnelly
PJ
Frayling
TM
Hattersley
AT
Boerwinkle
E
Melander
O
Kathiresan
S
Nilsson
PM
Deloukas
P
Thorsteinsdottir
U
Groop
LC
Stefansson
K
Hu
F
Pankow
JS
Dupuis
J
Meigs
JB
Altshuler
D
Boehnke
M
McCarthy
MI
Wellcome Trust Case Control Consortium; Meta-Analyses of Glucose and Insulin-related traits Consortium (MAGIC) Investigators; Genetic Investigation of ANthropometric Traits (GIANT) Consortium; Asian Genetic Epidemiology Network–Type 2 Diabetes (AGEN-T2D) Consortium; South Asian Type 2 Diabetes (SAT2D) Consortium; DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) Consortium
Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes
Nat Genet
2012
, vol. 
9
 (pg. 
981
-
990
)
13
Deloukas
P
Kanoni
S
Willenborg
C
Farrall
M
Assimes
TL
Thompson
JR
Ingelsson
E
Saleheen
D
Erdmann
J
Goldstein
BA
Stirrups
K
König
IR
Cazier
JB
Johansson
A
Hall
AS
Lee
JY
Willer
CJ
Chambers
JC
Esko
T
Folkersen
L
Goel
A
Grundberg
E
Havulinna
AS
Ho
WK
Hopewell
JC
Eriksson
N
Kleber
ME
Kristiansson
K
Lundmark
P
Lyytikäinen
LP
Rafelt
S
Shungin
D
Strawbridge
RJ
Thorleifsson
G
Tikkanen
E
Van Zuydam
N
Voight
BF
Waite
LL
Zhang
W
Ziegler
A
Absher
D
Altshuler
D
Balmforth
AJ
Barroso
I
Braund
PS
Burgdorf
C
Claudi-Boehm
S
Cox
D
Dimitriou
M
Do
R
Doney
AS
El Mokhtari
N
Eriksson
P
Fischer
K
Fontanillas
P
Franco-Cereceda
A
Gigante
B
Groop
L
Gustafsson
S
Hager
J
Hallmans
G
Han
BG
Hunt
SE
Kang
HM
Illig
T
Kessler
T
Knowles
JW
Kolovou
G
Kuusisto
J
Langenberg
C
Langford
C
Leander
K
Lokki
ML
Lundmark
A
McCarthy
MI
Meisinger
C
Melander
O
Mihailov
E
Maouche
S
Morris
AD
Müller-Nurasyid
M
Nikus
K
Peden
JF
Rayner
NW
Rasheed
A
Rosinger
S
Rubin
D
Rumpf
MP
Schäfer
A
Sivananthan
M
Song
C
Stewart
AF
Tan
ST
Thorgeirsson
G
van der Schoot
CE
Wagner
PJ
Wells
GA
Wild
PS
Yang
TP
Amouyel
P
Arveiler
D
Basart
H
Boehnke
M
Boerwinkle
E
Brambilla
P
Cambien
F
Cupples
AL
de Faire
U
Dehghan
A
Diemert
P
Epstein
SE
Evans
A
Ferrario
MM
Ferrières
J
Gauguier
D
Go
AS
Goodall
AH
Gudnason
V
Hazen
SL
Holm
H
Iribarren
C
Jang
Y
Kähönen
M
Kee
F
Kim
HS
Klopp
N
Koenig
W
Kratzer
W
Kuulasmaa
K
Laakso
M
Laaksonen
R
Lee
JY
Lind
L
Ouwehand
WH
Parish
S
Park
JE
Pedersen
NL
Peters
A
Quertermous
T
Rader
DJ
Salomaa
V
Schadt
E
Shah
SH
Sinisalo
J
Stark
K
Stefansson
K
Trégouët
DA
Virtamo
J
Wallentin
L
Wareham
N
Zimmermann
ME
Nieminen
MS
Hengstenberg
C
Sandhu
MS
Pastinen
T
Syvänen
AC
Hovingh
GK
Dedoussis
G
Franks
PW
Lehtimäki
T
Metspalu
A
Zalloua
PA
Siegbahn
A
Schreiber
S
Ripatti
S
Blankenberg
SS
Perola
M
Clarke
R
Boehm
BO
O'Donnell
C
Reilly
MP
März
W
Collins
R
Kathiresan
S
Hamsten
A
Kooner
JS
Thorsteinsdottir
U
Danesh
J
Palmer
CN
Roberts
R
Watkins
H
Schunkert
H
Samani
NJ
CARDIoGRAMplusC4D Consortium; DIAGRAM Consortium; CARDIOGENICS Consortium; MuTHER Consortium; Wellcome Trust Case Control Consortium
Large-scale association analysis identifies new risk loci for coronary artery disease
Nat Genet
2013
, vol. 
1
 (pg. 
25
-
33
)
14
Schunkert
H
Samani
NJ
Elevated C-reactive protein in atherosclerosis—chicken or egg?
N Engl J Med
2008
, vol. 
18
 (pg. 
1953
-
1955
)
15
Lawlor
DA
Harbord
RM
Sterne
JAC
Timpson
N
Davey Smith
G
Mendelian randomization: using genes as instruments for making causal inferences in epidemiology
Stat Med
2008
, vol. 
8
 (pg. 
1133
-
1163
)
16
Linsel-Nitschke
P
Götz
A
Erdmann
J
Braenne
I
Braund
P
Hengstenberg
C
Stark
K
Fischer
M
Schreiber
S
El Mokhtari
NE
Schaefer
A
Schrezenmeir
J
Rubin
D
Hinney
A
Reinehr
T
Roth
C
Ortlepp
J
Hanrath
P
Hall
AS
Mangino
M
Lieb
W
Lamina
C
Heid
IM
Doering
A
Gieger
C
Peters
A
Meitinger
T
Wichmann
HE
König
IR
Ziegler
A
Kronenberg
F
Samani
NJ
Schunkert
H
Wellcome Trust Case Control Consortium (WTCCC); Cardiogenics Consortium
Lifelong reduction of LDL-cholesterol related to a common variant in the LDL-receptor gene decreases the risk of coronary artery disease—a Mendelian randomisation study
PLos One
2008
, vol. 
8
 pg. 
2986
 
17
Voight
BF
Peloso
GM
Orho-Melander
M
Frikke-Schmidt
R
Barbalic
M
Jensen
MK
Hindy
G
Hólm
H
Ding
EL
Johnson
T
Schunkert
H
Samani
NJ
Clarke
R
Hopewell
JC
Thompson
JF
Li
M
Thorleifsson
G
Newton-Cheh
C
Musunuru
K
Pirruccello
JP
Saleheen
D
Chen
L
Stewart
A
Schillert
A
Thorsteinsdottir
U
Thorgeirsson
G
Anand
S
Engert
JC
Morgan
T
Spertus
J
Stoll
M
Berger
K
Martinelli
N
Girelli
D
McKeown
PP
Patterson
CC
Epstein
SE
Devaney
J
Burnett
MS
Mooser
V
Ripatti
S
Surakka
I
Nieminen
MS
Sinisalo
J
Lokki
ML
Perola
M
Havulinna
A
de Faire
U
Gigante
B
Ingelsson
E
Zeller
T
Wild
P
de Bakker
PI
Klungel
OH
Maitland-van der Zee
AH
Peters
BJ
de Boer
A
Grobbee
DE
Kamphuisen
PW
Deneer
VH
Elbers
CC
Onland-Moret
NC
Hofker
MH
Wijmenga
C
Verschuren
WM
Boer
JM
van der Schouw
YT
Rasheed
A
Frossard
P
Demissie
S
Willer
C
Do
R
Ordovas
JM
Abecasis
GR
Boehnke
M
Mohlke
KL
Daly
MJ
Guiducci
C
Burtt
NP
Surti
A
Gonzalez
E
Purcell
S
Gabriel
S
Marrugat
J
Peden
J
Erdmann
J
Diemert
P
Willenborg
C
König
IR
Fischer
M
Hengstenberg
C
Ziegler
A
Buysschaert
I
Lambrechts
D
Van de Werf
F
Fox
KA
El Mokhtari
NE
Rubin
D
Schrezenmeir
J
Schreiber
S
Schäfer
A
Danesh
J
Blankenberg
S
Roberts
R
McPherson
R
Watkins
H
Hall
AS
Overvad
K
Rimm
E
Boerwinkle
E
Tybjaerg-Hansen
A
Cupples
LA
Reilly
MP
Melander
O
Mannucci
PM
Ardissino
D
Siscovick
D
Elosua
R
Stefansson
K
O'Donnell
CJ
Salomaa
V
Rader
DJ
Peltonen
L
Schwartz
SM
Altshuler
D
Kathiresan
S
Plasma HDL cholesterol and risk of myocardial infarction: a Mendelian randomisation study
Lancet
2012
, vol. 
9841
 (pg. 
572
-
580
)
18
Kaptoge
S
Di Angelantonio
E
Lowe
G
Pepys
MB
Thompson
SG
Collins
R
Danesh
J
Emerging Risk Factors Collaboration
C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis
Lancet
2010
, vol. 
9709
 (pg. 
132
-
140
)
19
Wensley
F
Gao
P
Burgess
S
Kaptoge
S
Di Angelantonio
E
Shah
T
Engert
JC
Clarke
R
Davey-Smith
G
Nordestgaard
BG
Saleheen
D
Samani
NJ
Sandhu
M
Anand
S
Pepys
MB
Smeeth
L
Whittaker
J
Casas
JP
Thompson
SG
Hingorani
AD
Danesh
J
C Reactive Protein Coronary Heart Disease Genetics Collaboration (CCGC)
Association between C reactive protein and coronary heart disease: mendelian randomisation analysis based on individual participant data
BMJ
2011
, vol. 
342
 pg. 
548
 
20
Zacho
J
Tybjaerg-Hansen
A
Jensen
JS
Grande
P
Sillesen
H
Nordestgaard
BG
Genetically elevated C-reactive protein and ischemic vascular disease
N Engl J Med
2008
, vol. 
18
 (pg. 
1897
-
1908
)
21
Elliott
P
Chambers
JC
Zhang
W
Clarke
R
Hopewell
JC
Peden
JF
Erdmann
J
Braund
P
Engert
JC
Bennett
D
Coin
L
Ashby
D
Tzoulaki
I
Brown
IJ
Mt-Isa
S
McCarthy
MI
Peltonen
L
Freimer
NB
Farrall
M
Ruokonen
A
Hamsten
A
Lim
N
Froguel
P
Waterworth
DM
Vollenweider
P
Waeber
G
Jarvelin
MR
Mooser
V
Scott
J
Hall
AS
Schunkert
H
Anand
SS
Collins
R
Samani
NJ
Watkins
H
Kooner
JS
Genetic Loci associated with C-reactive protein levels and risk of coronary heart disease
JAMA
2009
, vol. 
1
 (pg. 
37
-
48
)
22
Hingorani
A
Humphries
S
Nature's randomised trials
Lancet
2005
, vol. 
9501
 (pg. 
1906
-
1908
)
23
Havulinna
AS
Kettunen
J
Ukkola
O
Osmond
C
Eriksson
JG
Kesäniemi
YA
Jula
A
Peltonen
L
Kontula
K
Salomaa
V
Newton-Cheh
C
A blood pressure genetic risk score is a significant predictor of incident cardiovascular events in 32,669 individuals
Hypertension
2013
, vol. 
5
 (pg. 
987
-
994
)
24
Codd
V
Nelson
CP
Albrecht
E
Mangino
M
Deelen
J
Buxton
JL
Hottenga
JJ
Fischer
K
Esko
T
Surakka
I
Broer
L
Nyholt
DR
Mateo Leach
I
Salo
P
Hägg
S
Matthews
MK
Palmen
J
Norata
GD
O'Reilly
PF
Saleheen
D
Amin
N
Balmforth
AJ
Beekman
M
de Boer
RA
Böhringer
S
Braund
PS
Burton
PR
de Craen
AJ
Denniff
M
Dong
Y
Douroudis
K
Dubinina
E
Eriksson
JG
Garlaschelli
K
Guo
D
Hartikainen
AL
Henders
AK
Houwing-Duistermaat
JJ
Kananen
L
Karssen
LC
Kettunen
J
Klopp
N
Lagou
V
van Leeuwen
EM
Madden
PA
Mägi
R
Magnusson
PK
Männistö
S
McCarthy
MI
Medland
SE
Mihailov
E
Montgomery
GW
Oostra
BA
Palotie
A
Peters
A
Pollard
H
Pouta
A
Prokopenko
I
Ripatti
S
Salomaa
V
Suchiman
HE
Valdes
AM
Verweij
N
Viñuela
A
Wang
X
Wichmann
HE
Widen
E
Willemsen
G
Wright
MJ
Xia
K
Xiao
X
van Veldhuisen
DJ
Catapano
AL
Tobin
MD
Hall
AS
Blakemore
AI
van Gilst
WH
Zhu
H
Consortium
C
Erdmann
J
Reilly
MP
Kathiresan
S
Schunkert
H
Talmud
PJ
Pedersen
NL
Perola
M
Ouwehand
W
Kaprio
J
Martin
NG
van Duijn
CM
Hovatta
I
Gieger
C
Metspalu
A
Boomsma
DI
Jarvelin
MR
Slagboom
PE
Thompson
JR
Spector
TD
van der Harst
P
Samani
NJ
Identification of seven loci affecting mean telomere length and their association with disease
Nat Genet
2013
, vol. 
4
 (pg. 
422
-
427
)
25
Lieb
W
Jansen
H
Loley
C
Pencina
MJ
Nelson
CP
Newton-Cheh
C
Kathiresan
S
Reilly
MP
Assimes
TL
Boerwinkle
E
Hall
AS
Hengstenberg
C
Laaksonen
R
McPherson
R
Thorsteinsdottir
U
Ziegler
A
Peters
A
Thompson
JR
König
IR
Erdmann
J
Samani
NJ
Vasan
RS
Schunkert
H
Assimes
TL
Deloukas
P
Erdmann
J
Holm
H
Kathiresan
S
König
IR
McPherson
R
Reilly
MP
Roberts
R
Samani
NJ
Schunkert
H
Stewart
AF
CARDIoGRAM
Genetic predisposition to higher blood pressure increases coronary artery disease risk
Hypertension
2013
, vol. 
5
 (pg. 
995
-
1001
)
26
Cohen
JC
Boerwinkle
E
Mosley
TH
Jr
Hobbs
HH
Sequence variations in PCSK9, low LDL, and protection against coronary heart disease
N Engl J Med
2006
, vol. 
12
 (pg. 
1264
-
1272
)
27
Nicholls
SJ
Cavender
MA
Kastelein
JJ
Schwartz
G
Waters
DD
Rosenson
RS
Bash
D
Hislop
C
Inhibition of secretory phospholipase A(2) in patients with acute coronary syndromes: rationale and design of the vascular inflammation suppression to treat acute coronary syndrome for 16 weeks (VISTA-16) trial
Cardiovasc Drugs Ther Spons Int Soc Cardiovasc Pharmacother
2012
, vol. 
1
 (pg. 
71
-
75
)
28
Kathiresan
S
Melander
O
Anevski
D
Guiducci
C
Burtt
NP
Roos
C
Hirschhorn
JN
Berglund
G
Hedblad
B
Groop
L
Altshuler
DM
Newton-Cheh
C
Orho-Melander
M
Polymorphisms associated with cholesterol and risk of cardiovascular events
N Engl J Med
2008
, vol. 
12
 (pg. 
1240
-
1249
)
29
Souverein
OW
Defesche
JC
Zwinderman
AH
Kastelein
JJ
Tanck
MW
Influence of LDL-receptor mutation type on age at first cardiovascular event in patients with familial hypercholesterolaemia
Eur Heart J
2007
, vol. 
3
 (pg. 
299
-
304
)
30
Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S)
Lancet
1994
, vol. 
8934
 (pg. 
1383
-
1389
)
31
Baigent
C
Keech
A
Kearney
PM
Blackwell
L
Buck
G
Pollicino
C
Kirby
A
Sourjina
T
Peto
R
Collins
R
Simes
R
Cholesterol Treatment Trialists’ (CTT) Collaborators
Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins
Lancet
2005
, vol. 
9493
 (pg. 
1267
-
1278
)
32
Kohli
P
Desai
NR
Giugliano
RP
Kim
JB
Somaratne
R
Huang
F
Knusel
B
McDonald
S
Abrahamsen
T
Wasserman
SM
Scott
R
Sabatine
MS
Design and rationale of the LAPLACE-TIMI 57 trial: a phase II, double-blind, placebo-controlled study of the efficacy and tolerability of a monoclonal antibody inhibitor of PCSK9 in subjects with hypercholesterolemia on background statin therapy
Clin Cardiol
2012
, vol. 
7
 (pg. 
385
-
391
)
33
Giugliano
RP
Desai
NR
Kohli
P
Rogers
WJ
Somaratne
R
Huang
F
Liu
T
Mohanavelu
S
Hoffman
EB
McDonald
ST
Abrahamsen
TE
Wasserman
SM
Scott
R
Sabatine
MS
LAPLACE-TIMI 57 Investigators
Efficacy, safety, and tolerability of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 in combination with a statin in patients with hypercholesterolaemia (LAPLACE-TIMI 57): a randomised, placebo-controlled, dose-ranging, phase 2 study
Lancet
2012
, vol. 
9858
 (pg. 
2007
-
2017
)
34
McKenney
JM
Koren
MJ
Kereiakes
DJ
Hanotin
C
Ferrand
AC
Stein
EA
Safety and efficacy of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 serine protease, SAR236553/REGN727, in patients with primary hypercholesterolemia receiving ongoing stable atorvastatin therapy
J Am Coll Cardiol
2012
, vol. 
25
 (pg. 
2344
-
2353
)
35
Gordon
T
Castelli
WP
Hjortland
MC
Kannel
WB
Dawber
TR
High density lipoprotein as a protective factor against coronary heart disease. The Framingham study
Am J Med
1977
, vol. 
5
 (pg. 
707
-
714
)
36
Katz
PM
Leiter
LA
Drugs targeting high-density lipoprotein cholesterol for coronary artery disease management
Can J Cardiol
2012
, vol. 
6
 (pg. 
667
-
677
)
37
Haase
CL
Tybjærg-Hansen
A
Qayyum
AA
Schou
J
Nordestgaard
BG
Frikke-Schmidt
R
LCAT, HDL cholesterol and ischemic cardiovascular disease: a Mendelian randomization study of HDL cholesterol in 54,500 individuals
J Clin Endocrinol Metab
2012
, vol. 
2
 (pg. 
248
-
256
)
38
Frikke-Schmidt
R
Nordestgaard
BG
Stene
MC
Sethi
AA
Remaley
AT
Schnohr
P
Grande
P
Tybjaerg-Hansen
A
Association of loss-of-function mutations in the ABCA1 gene with high-density lipoprotein cholesterol levels and risk of ischemic heart disease
JAMA
2008
, vol. 
21
 (pg. 
2524
-
2532
)
39
Thompson
A
Di Angelantonio
E
Sarwar
N
Erqou
S
Saleheen
D
Dullaart
RP
Keavney
B
Ye
Z
Danesh
J
Association of cholesteryl ester transfer protein genotypes with CETP mass and activity, lipid levels, and coronary risk
JAMA
2008
, vol. 
23
 (pg. 
2777
-
2788
)
40
Barter
PJ
Kastelein
JJ
Targeting cholesteryl ester transfer protein for the prevention and management of cardiovascular disease
J Am Coll Cardiol
2006
, vol. 
47
 (pg. 
492
-
499
)
41
Inazu
A
Brown
ML
Hesler
CB
Agellon
LB
Koizumi
J
Takata
K
Maruhama
Y
Mabuchi
H
Tall
AR
Increased high-density lipoprotein levels caused by a common cholesteryl-ester transfer protein gene mutation
N Engl J Med
1990
, vol. 
18
 (pg. 
1234
-
1238
)
42
Ridker
PM
Paré
G
Parker
AN
Zee
RY
Miletich
JP
Chasman
DI
Polymorphism in the CETP gene region, HDL cholesterol, and risk of future myocardial infarction: Genomewide analysis among 18 245 initially healthy women from the Women's Genome Health Study
Circ Cardiovasc Genet
2009
, vol. 
1
 (pg. 
26
-
33
)
43
Wright
RS
Recent clinical trials evaluating benefit of drug therapy for modification of HDL cholesterol
Curr Opin Cardiol
2013
, vol. 
4
 (pg. 
389
-
398
)
44
Cannon
CP
Shah
S
Dansky
HM
Davidson
M
Brinton
EA
Gotto
AM
Stepanavage
M
Liu
SX
Gibbons
P
Ashraf
TB
Zafarino
J
Mitchel
Y
Barter
P
Determining the Efficacy and Tolerability Investigators
Safety of anacetrapib in patients with or at high risk for coronary heart disease
N Engl J Med
2010
, vol. 
25
 (pg. 
2406
-
2415
)
45
Holmer
SR
Hengstenberg
C
Kraft
HG
Mayer
B
Pöll
M
Kürzinger
S
Fischer
M
Löwel
H
Klein
G
Riegger
GA
Schunkert
H
Association of polymorphisms of the apolipoprotein(a) gene with lipoprotein(a) levels and myocardial infarction
Circulation
2003
, vol. 
5
 (pg. 
696
-
701
)
46
Nordestgaard
BG
Chapman
MJ
Ray
K
Borén
J
Andreotti
F
Watts
GF
Ginsberg
H
Amarenco
P
Catapano
A
Descamps
OS
Fisher
E
Kovanen
PT
Kuivenhoven
JA
Lesnik
P
Masana
L
Reiner
Z
Taskinen
MR
Tokgözoglu
L
Tybjærg-Hansen
A
European Atherosclerosis Society Consensus Panel
Lipoprotein(a) as a cardiovascular risk factor: current status
Eur Heart J
2010
, vol. 
23
 (pg. 
2844
-
2853
)
47
Trégouët
DA
König
IR
Erdmann
J
Munteanu
A
Braund
PS
Hall
AS
Grosshennig
A
Linsel-Nitschke
P
Perret
C
DeSuremain
M
Meitinger
T
Wright
BJ
Preuss
M
Balmforth
AJ
Ball
SG
Meisinger
C
Germain
C
Evans
A
Arveiler
D
Luc
G
Ruidavets
JB
Morrison
C
van der Harst
P
Schreiber
S
Neureuther
K
Schäfer
A
Bugert
P
El Mokhtari
NE
Schrezenmeir
J
Stark
K
Rubin
D
Wichmann
HE
Hengstenberg
C
Ouwehand
W
Ziegler
A
Tiret
L
Thompson
JR
Cambien
F
Schunkert
H
Samani
NJ
Wellcome Trust Case Control Consortium; Cardiogenics Consortium
Genome-wide haplotype association study identifies the SLC22A3-LPAL2-LPA gene cluster as a risk locus for coronary artery disease
Nat Genet
2009
, vol. 
3
 (pg. 
283
-
285
)
48
Clarke
R
Peden
JF
Hopewell
JC
Kyriakou
T
Goel
A
Heath
SC
Parish
S
Barlera
S
Franzosi
MG
Rust
S
Bennett
D
Silveira
A
Malarstig
A
Green
FR
Lathrop
M
Gigante
B
Leander
K
de Faire
U
Seedorf
U
Hamsten
A
Collins
R
Watkins
H
Farrall
M
PROCARDIS Consortium
Genetic variants associated with Lp(a) lipoprotein level and coronary disease
N Engl J Med
2009
, vol. 
26
 (pg. 
2518
-
2528
)
49
Kamstrup
PR
Tybjaerg-Hansen
A
Steffensen
R
Nordestgaard
BG
Genetically elevated lipoprotein(a) and increased risk of myocardial infarction
JAMA
2009
, vol. 
22
 (pg. 
2331
-
2339
)
50
Castelli
WP
Doyle
JT
Gordon
T
Hames
CG
Hjortland
MC
Hulley
SB
Kagan
A
Zukel
WJ
HDL cholesterol and other lipids in coronary heart disease. The cooperative lipoprotein phenotyping study
Circulation
1977
, vol. 
5
 (pg. 
767
-
772
)
51
Kannel
WB
Vasan
RS
Triglycerides as vascular risk factors: new epidemiologic insights
Curr Opin Cardiol
2009
, vol. 
4
 (pg. 
345
-
350
)
52
Nordestgaard
BG
Benn
M
Schnohr
P
Tybjaerg-Hansen
A
Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women
JAMA
2007
, vol. 
3
 (pg. 
299
-
308
)
53
Bansal
S
Buring
JE
Rifai
N
Mora
S
Sacks
FM
Ridker
PM
Fasting compared with nonfasting triglycerides and risk of cardiovascular events in women
JAMA
2007
, vol. 
3
 (pg. 
309
-
316
)
54
Jørgensen
AB
Frikke-Schmidt
R
West
AS
Grande
P
Nordestgaard
BG
Tybjærg-Hansen
A
Genetically elevated non-fasting triglycerides and calculated remnant cholesterol as causal risk factors for myocardial infarction
Eur Heart J
2013
, vol. 
24
 (pg. 
1826
-
1833
)
55
Sarwar
N
Sandhu
MS
Ricketts
SL
Butterworth
AS
Di Angelantonio
E
Boekholdt
SM
Ouwehand
W
Watkins
H
Samani
NJ
Saleheen
D
Lawlor
D
Reilly
MP
Hingorani
AD
Talmud
PJ
Danesh
J
Triglyceride Coronary Disease Genetics Consortium and Emerging Risk Factors Collaboration
Triglyceride-mediated pathways and coronary disease: collaborative analysis of 101 studies
Lancet
2010
, vol. 
9726
 (pg. 
1634
-
1639
)
56
Do
R
Willer
CJ
Schmidt
EM
Sengupta
S
Gao
C
Peloso
GM
Gustafsson
S
Kanoni
S
Ganna
A
Chen
J
Buchkovich
ML
Mora
S
Beckmann
JS
Bragg-Gresham
JL
Chang
HY
Demirkan
A
Den Hertog
HM
Donnelly
LA
Ehret
GB
Esko
T
Feitosa
MF
Ferreira
T
Fischer
K
Fontanillas
P
Fraser
RM
Freitag
DF
Gurdasani
D
Heikkilä
K
Hyppönen
E
Isaacs
A
Jackson
AU
Johansson
A
Johnson
T
Kaakinen
M
Kettunen
J
Kleber
ME
Li
X
Luan
J
Lyytikäinen
LP
Magnusson
PK
Mangino
M
Mihailov
E
Montasser
ME
Müller-Nurasyid
M
Nolte
IM
O'Connell
JR
Palmer
CD
Perola
M
Petersen
AK
Sanna
S
Saxena
R
Service
SK
Shah
S
Shungin
D
Sidore
C
Song
C
Strawbridge
RJ
Surakka
I
Tanaka
T
Teslovich
TM
Thorleifsson
G
Van den Herik
EG
Voight
BF
Volcik
KA
Waite
LL
Wong
A
Wu
Y
Zhang
W
Absher
D
Asiki
G
Barroso
I
Been
LF
Bolton
JL
Bonnycastle
LL
Brambilla
P
Burnett
MS
Cesana
G
Dimitriou
M
Doney
AS
Döring
A
Elliott
P
Epstein
SE
Eyjolfsson
GI
Gigante
B
Goodarzi
MO
Grallert
H
Gravito
ML
Groves
CJ
Hallmans
G
Hartikainen
AL
Hayward
C
Hernandez
D
Hicks
AA
Holm
H
Hung
YJ
Illig
T
Jones
MR
Kaleebu
P
Kastelein
JJ
Khaw
KT
Kim
E
Klopp
N
Komulainen
P
Kumari
M
Langenberg
C
Lehtimäki
T
Lin
SY
Lindström
J
Loos
RJ
Mach
F
McArdle
WL
Meisinger
C
Mitchell
BD
Müller
G
Nagaraja
R
Narisu
N
Nieminen
TV
Nsubuga
RN
Olafsson
I
Ong
KK
Palotie
A
Papamarkou
T
Pomilla
C
Pouta
A
Rader
DJ
Reilly
MP
Ridker
PM
Rivadeneira
F
Rudan
I
Ruokonen
A
Samani
N
Scharnagl
H
Seeley
J
Silander
K
Stančáková
A
Stirrups
K
Swift
AJ
Tiret
L
Uitterlinden
AG
van Pelt
LJ
Vedantam
S
Wainwright
N
Wijmenga
C
Wild
SH
Willemsen
G
Wilsgaard
T
Wilson
JF
Young
EH
Zhao
JH
Adair
LS
Arveiler
D
Assimes
TL
Bandinelli
S
Bennett
F
Bochud
M
Boehm
BO
Boomsma
DI
Borecki
IB
Bornstein
SR
Bovet
P
Burnier
M
Campbell
H
Chakravarti
A
Chambers
JC
Chen
YD
Collins
FS
Cooper
RS
Danesh
J
Dedoussis
G
de Faire
U
Feranil
AB
Ferrières
J
Ferrucci
L
Freimer
NB
Gieger
C
Groop
LC
Gudnason
V
Gyllensten
U
Hamsten
A
Harris
TB
Hingorani
A
Hirschhorn
JN
Hofman
A
Hovingh
GK
Hsiung
CA
Humphries
SE
Hunt
SC
Hveem
K
Iribarren
C
Järvelin
MR
Jula
A
Kähönen
M
Kaprio
J
Kesäniemi
A
Kivimaki
M
Kooner
JS
Koudstaal
PJ
Krauss
RM
Kuh
D
Kuusisto
J
Kyvik
KO
Laakso
M
Lakka
TA
Lind
L
Lindgren
CM
Martin
NG
März
W
McCarthy
MI
McKenzie
CA
Meneton
P
Metspalu
A
Moilanen
L
Morris
AD
Munroe
PB
Njølstad
I
Pedersen
NL
Power
C
Pramstaller
PP
Price
JF
Psaty
BM
Quertermous
T
Rauramaa
R
Saleheen
D
Salomaa
V
Sanghera
DK
Saramies
J
Schwarz
PE
Sheu
WH
Shuldiner
AR
Siegbahn
A
Spector
TD
Stefansson
K
Strachan
DP
Tayo
BO
Tremoli
E
Tuomilehto
J
Uusitupa
M
van Duijn
CM
Vollenweider
P
Wallentin
L
Wareham
NJ
Whitfield
JB
Wolffenbuttel
BH
Altshuler
D
Ordovas
JM
Boerwinkle
E
Palmer
CN
Thorsteinsdottir
U
Chasman
DI
Rotter
JI
Franks
PW
Ripatti
S
Cupples
LA
Sandhu
MS
Rich
SS
Boehnke
M
Deloukas
P
Mohlke
KL
Ingelsson
E
Abecasis
GR
Daly
MJ
Neale
BM
Kathiresan
S
Common variants associated with plasma triglycerides and risk for coronary artery disease
Nat Genet
2013
, vol. 
11
 (pg. 
1345
-
1352
)
57
Willer
CJ
Schmidt
EM
Sengupta
S
Peloso
GM
Gustafsson
S
Kanoni
S
Ganna
A
Chen
J
Buchkovich
ML
Mora
S
Beckmann
JS
Bragg-Gresham
JL
Chang
HY
Demirkan
A
Den Hertog
HM
Do
R
Donnelly
LA
Ehret
GB
Esko
T
Feitosa
MF
Ferreira
T
Fischer
K
Fontanillas
P
Fraser
RM
Freitag
DF
Gurdasani
D
Heikkilä
K
Hyppönen
E
Isaacs
A
Jackson
AU
Johansson
A
Johnson
T
Kaakinen
M
Kettunen
J
Kleber
ME
Li
X
Luan
J
Lyytikäinen
LP
Magnusson
PK
Mangino
M
Mihailov
E
Montasser
ME
Müller-Nurasyid
M
Nolte
IM
O'Connell
JR
Palmer
CD
Perola
M
Petersen
AK
Sanna
S
Saxena
R
Service
SK
Shah
S
Shungin
D
Sidore
C
Song
C
Strawbridge
RJ
Surakka
I
Tanaka
T
Teslovich
TM
Thorleifsson
G
Van den Herik
EG
Voight
BF
Volcik
KA
Waite
LL
Wong
A
Wu
Y
Zhang
W
Absher
D
Asiki
G
Barroso
I
Been
LF
Bolton
JL
Bonnycastle
LL
Brambilla
P
Burnett
MS
Cesana
G
Dimitriou
M
Doney
AS
Döring
A
Elliott
P
Epstein
SE
Eyjolfsson
GI
Gigante
B
Goodarzi
MO
Grallert
H
Gravito
ML
Groves
CJ
Hallmans
G
Hartikainen
AL
Hayward
C
Hernandez
D
Hicks
AA
Holm
H
Hung
YJ
Illig
T
Jones
MR
Kaleebu
P
Kastelein
JJ
Khaw
KT
Kim
E
Klopp
N
Komulainen
P
Kumari
M
Langenberg
C
Lehtimäki
T
Lin
SY
Lindström
J
Loos
RJ
Mach
F
McArdle
WL
Meisinger
C
Mitchell
BD
Müller
G
Nagaraja
R
Narisu
N
Nieminen
TV
Nsubuga
RN
Olafsson
I
Ong
KK
Palotie
A
Papamarkou
T
Pomilla
C
Pouta
A
Rader
DJ
Reilly
MP
Ridker
PM
Rivadeneira
F
Rudan
I
Ruokonen
A
Samani
N
Scharnagl
H
Seeley
J
Silander
K
Stancáková
A
Stirrups
K
Swift
AJ
Tiret
L
Uitterlinden
AG
van Pelt
LJ
Vedantam
S
Wainwright
N
Wijmenga
C
Wild
SH
Willemsen
G
Wilsgaard
T
Wilson
JF
Young
EH
Zhao
JH
Adair
LS
Arveiler
D
Assimes
TL
Bandinelli
S
Bennett
F
Bochud
M
Boehm
BO
Boomsma
DI
Borecki
IB
Bornstein
SR
Bovet
P
Burnier
M
Campbell
H
Chakravarti
A
Chambers
JC
Chen
YD
Collins
FS
Cooper
RS
Danesh
J
Dedoussis
G
de Faire
U
Feranil
AB
Ferrières
J
Ferrucci
L
Freimer
NB
Gieger
C
Groop
LC
Gudnason
V
Gyllensten
U
Hamsten
A
Harris
TB
Hingorani
A
Hirschhorn
JN
Hofman
A
Hovingh
GK
Hsiung
CA
Humphries
SE
Hunt
SC
Hveem
K
Iribarren
C
Järvelin
MR
Jula
A
Kähönen
M
Kaprio
J
Kesäniemi
A
Kivimaki
M
Kooner
JS
Koudstaal
PJ
Krauss
RM
Kuh
D
Kuusisto
J
Kyvik
KO
Laakso
M
Lakka
TA
Lind
L
Lindgren
CM
Martin
NG
März
W
McCarthy
MI
McKenzie
CA
Meneton
P
Metspalu
A
Moilanen
L
Morris
AD
Munroe
PB
Njølstad
I
Pedersen
NL
Power
C
Pramstaller
PP
Price
JF
Psaty
BM
Quertermous
T
Rauramaa
R
Saleheen
D
Salomaa
V
Sanghera
DK
Saramies
J
Schwarz
PE
Sheu
WH
Shuldiner
AR
Siegbahn
A
Spector
TD
Stefansson
K
Strachan
DP
Tayo
BO
Tremoli
E
Tuomilehto
J
Uusitupa
M
van Duijn
CM
Vollenweider
P
Wallentin
L
Wareham
NJ
Whitfield
JB
Wolffenbuttel
BH
Ordovas
JM
Boerwinkle
E
Palmer
CN
Thorsteinsdottir
U
Chasman
DI
Rotter
JI
Franks
PW
Ripatti
S
Cupples
LA
Sandhu
MS
Rich
SS
Boehnke
M
Deloukas
P
Kathiresan
S
Mohlke
KL
Ingelsson
E
Abecasis
GR
Global Lipids Genetics Consortium
Discovery and refinement of loci associated with lipid levels
Nat Genet
2013
, vol. 
11
 (pg. 
1274
-
1283
)
58
Rosenson
RS
Hurt-Camejo
E
Phospholipase A2 enzymes and the risk of atherosclerosis
Eur Heart J
2012
, vol. 
23
 (pg. 
2899
-
2909
)
59
Brilakis
ES
McConnell
JP
Lennon
RJ
Elesber
AA
Meyer
JG
Berger
PB
Association of lipoprotein-associated phospholipase A2 levels with coronary artery disease risk factors, angiographic coronary artery disease, and major adverse events at follow-up
Eur Heart J
2005
, vol. 
2
 (pg. 
137
-
144
)
60
Cook
NR
Paynter
NP
Manson
JE
Martin
LW
Robinson
JG
Wassertheil-Smoller
S
Ridker
PM
Clinical utility of lipoprotein-associated phospholipase A₂ for cardiovascular disease prediction in a multiethnic cohort of women
Clin Chem
2012
, vol. 
9
 (pg. 
1352
-
1363
)
61
Maiolino
G
Pedon
L
Cesari
M
Frigo
AC
Wolfert
RL
Barisa
M
Pagliani
L
Rossitto
G
Seccia
TM
Zanchetta
M
Rossi
GP
Lipoprotein-associated phospholipase A2 activity predicts cardiovascular events in high risk coronary artery disease patients
PLos One
2012
, vol. 
10
 pg. 
48171
 
62
Ridker
PM
MacFadyen
JG
Wolfert
RL
Koenig
W
Relationship of lipoprotein-associated phospholipase A₂ mass and activity with incident vascular events among primary prevention patients allocated to placebo or to statin therapy: an analysis from the JUPITER trial
Clin Chem
2012
, vol. 
5
 (pg. 
877
-
886
)
63
Casas
JP
Ninio
E
Panayiotou
A
Palmen
J
Cooper
JA
Ricketts
SL
Sofat
R
Nicolaides
AN
Corsetti
JP
Fowkes
FG
Tzoulaki
I
Kumari
M
Brunner
EJ
Kivimaki
M
Marmot
MG
Hoffmann
MM
Winkler
K
März
W
Ye
S
Stirnadel
HA
Boekholdt
SM
Khaw
KT
Humphries
SE
Sandhu
MS
Hingorani
AD
Talmud
PJ
PLA2G7 genotype, lipoprotein-associated phospholipase A2 activity, and coronary heart disease risk in 10 494 cases and 15 624 controls of European Ancestry
Circulation
2010
, vol. 
21
 (pg. 
2284
-
2293
)
64
GlaxoSmithKline
GSK announces top-line results from pivotal Phase III study of darapladib in chronic coronary heart disease
 
65
O'Donoghue
ML
Braunwald
E
White
HD
Serruys
P
Steg
PG
Hochman
J
Maggioni
AP
Bode
C
Weaver
D
Johnson
JL
Cicconetti
G
Lukas
MA
Tarka
E
Cannon
CP
Study design and rationale for the Stabilization of pLaques usIng Darapladib-Thrombolysis in Myocardial Infarction (SOLID-TIMI 52) trial in patients after an acute coronary syndrome
Am Heart J
2011
, vol. 
4
 (pg. 
613
-
619
)
66
Mallat
Z
Steg
PG
Benessiano
J
Tanguy
ML
Fox
KA
Collet
JP
Dabbous
OH
Henry
P
Carruthers
KF
Dauphin
A
Arguelles
CS
Masliah
J
Hugel
B
Montalescot
G
Freyssinet
JM
Asselain
B
Tedgui
A
Circulating secretory phospholipase A2 activity predicts recurrent events in patients with severe acute coronary syndromes
J Am Coll Cardiol
2005
, vol. 
7
 (pg. 
1249
-
1257
)
67
Boekholdt
SM
Keller
TT
Wareham
NJ
Luben
R
Bingham
SA
Day
NE
Sandhu
MS
Jukema
JW
Kastelein
JJ
Hack
CE
Khaw
KT
Serum levels of type II secretory phospholipase A2 and the risk of future coronary artery disease in apparently healthy men and women: the EPIC-Norfolk Prospective Population Study
Arterioscler Thromb Vasc Biol
2005
, vol. 
4
 (pg. 
839
-
846
)
68
Breitling
LP
Koenig
W
Fischer
M
Mallat
Z
Hengstenberg
C
Rothenbacher
D
Brenner
H
Type II secretory phospholipase A2 and prognosis in patients with stable coronary heart disease: Mendelian randomization study
PLos One
2011
, vol. 
7
 pg. 
22318
 
69
Holmes
MV
Simon
T
Exeter
HJ
Folkersen
L
Asselbergs
FW
Guardiola
M
Cooper
JA
Palmen
J
Hubacek
JA
Carruthers
KF
Horne
BD
Brunisholz
KD
Mega
JL
van Iperen
EP
Li
M
Leusink
M
Trompet
S
Verschuren
JJ
Hovingh
GK
Dehghan
A
Nelson
CP
Kotti
S
Danchin
N
Scholz
M
Haase
CL
Rothenbacher
D
Swerdlow
DI
Kuchenbaecker
KB
Staines-Urias
E
Goel
A
van't Hooft
F
Gertow
K
de Faire
U
Panayiotou
AG
Tremoli
E
Baldassarre
D
Veglia
F
Holdt
LM
Beutner
F
Gansevoort
RT
Navis
GJ
Mateo Leach
I
Breitling
LP
Brenner
H
Thiery
J
Dallmeier
D
Franco-Cereceda
A
Boer
JM
Stephens
JW
Hofker
MH
Tedgui
A
Hofman
A
Uitterlinden
AG
Adamkova
V
Pitha
J
Onland-Moret
NC
Cramer
MJ
Nathoe
HM
Spiering
W
Klungel
OH
Kumari
M
Whincup
PH
Morrow
DA
Braund
PS
Hall
AS
Olsson
AG
Doevendans
PA
Trip
MD
Tobin
MD
Hamsten
A
Watkins
H
Koenig
W
Nicolaides
AN
Teupser
D
Day
IN
Carlquist
JF
Gaunt
TR
Ford
I
Sattar
N
Tsimikas
S
Schwartz
GG
Lawlor
DA
Morris
RW
Sandhu
MS
Poledne
R
Maitland-van der Zee
AH
Khaw
KT
Keating
BJ
van der Harst
P
Price
JF
Mehta
SR
Yusuf
S
Witteman
JC
Franco
OH
Jukema
JW
de Knijff
P
Tybjaerg-Hansen
A
Rader
DJ
Farrall
M
Samani
NJ
Kivimaki
M
Fox
KA
Humphries
SE
Anderson
JL
Boekholdt
SM
Palmer
TM
Eriksson
P
Paré
G
Hingorani
AD
Sabatine
MS
Mallat
Z
Casas
JP
Talmud
PJ
Secretory phospholipase A2-IIA and cardiovascular disease: a Mendelian randomization study
J Am Coll Cardiol
2013
, vol. 
13
 (pg. 
1966
-
1976
)
70
Koenig
W
Sund
M
Fröhlich
M
Fischer
HG
Löwel
H
Döring
A
Hutchinson
WL
Pepys
MB
C-Reactive protein, a sensitive marker of inflammation, predicts future risk of coronary heart disease in initially healthy middle-aged men: results from the MONICA (Monitoring Trends and Determinants in Cardiovascular Disease) Augsburg Cohort Study, 1984 to 1992
Circulation
1999
, vol. 
2
 (pg. 
237
-
242
)
71
Ridker
PM
Hennekens
CH
Buring
JE
Rifai
N
C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women
N Engl J Med
2000
, vol. 
12
 (pg. 
836
-
843
)
72
Jialal
I
Devaraj
S
Smith
G
Lam
KS
Kumaresan
PR
A novel peptide inhibitor attenuates C-reactive protein's pro-inflammatory effects in-vivo
Int J Cardiol
2013
, vol. 
4
 (pg. 
3909
-
3912
)
73
Casas
JP
Shah
T
Cooper
J
Hawe
E
McMahon
AD
Gaffney
D
Packard
CJ
O'Reilly
DS
Juhan-Vague
I
Yudkin
JS
Tremoli
E
Margaglione
M
Di Minno
G
Hamsten
A
Kooistra
T
Stephens
JW
Hurel
SJ
Livingstone
S
Colhoun
HM
Miller
GJ
Bautista
LE
Meade
T
Sattar
N
Humphries
SE
Hingorani
AD
Insight into the nature of the CRP-coronary event association using Mendelian randomization
Int J Epidemiol
2006
, vol. 
4
 (pg. 
922
-
931
)
74
Omicron Hartaigh
B
Thomas
GN
Bosch
JA
Hemming
K
Pilz
S
Loerbroks
A
Kleber
ME
Grammer
TB
Fischer
JE
Silbernagel
G
Tomaschitz
A
März
W
Evaluation of 9 biomarkers for predicting 10-year cardiovascular risk in patients undergoing coronary angiography: findings from the LUdwigshafen RIsk and Cardiovascular Health (LURIC) study
Int J Cardiol
2013
, vol. 
3
 (pg. 
2609
-
2615
)
75
Ridker
PM
Rifai
N
Stampfer
MJ
Hennekens
CH
Plasma concentration of interleukin-6 and the risk of future myocardial infarction among apparently healthy men
Circulation
2000
, vol. 
15
 (pg. 
1767
-
1177
)
76
von der Thüsen
JH
Kuiper
J
van Berkel
TJ
Biessen
EA
Interleukins in atherosclerosis: molecular pathways and therapeutic potential
Pharmacol Rev
2003
, vol. 
1
 (pg. 
133
-
166
)
77
Md Yusof
MY
Emery
P
Targeting interleukin-6 in rheumatoid arthritis
Drug
2013
, vol. 
4
 (pg. 
341
-
356
)
78
Sarwar
N
Butterworth
AS
Freitag
DF
Gregson
J
Willeit
P
Gorman
DN
Gao
P
Saleheen
D
Rendon
A
Nelson
CP
Braund
PS
Hall
AS
Chasman
DI
Tybjærg-Hansen
A
Chambers
JC
Benjamin
EJ
Franks
PW
Clarke
R
Wilde
AA
Trip
MD
Steri
M
Witteman
JC
Qi
L
van der Schoot
CE
de Faire
U
Erdmann
J
Stringham
HM
Koenig
W
Rader
DJ
Melzer
D
Reich
D
Psaty
BM
Kleber
ME
Panagiotakos
DB
Willeit
J
Wennberg
P
Woodward
M
Adamovic
S
Rimm
EB
Meade
TW
Gillum
RF
Shaffer
JA
Hofman
A
Onat
A
Sundström
J
Wassertheil-Smoller
S
Mellström
D
Gallacher
J
Cushman
M
Tracy
RP
Kauhanen
J
Karlsson
M
Salonen
JT
Wilhelmsen
L
Amouyel
P
Cantin
B
Best
LG
Ben-Shlomo
Y
Manson
JE
Davey-Smith
G
de Bakker
PI
O'Donnell
CJ
Wilson
JF
Wilson
AG
Assimes
TL
Jansson
JO
Ohlsson
C
Tivesten
Å
Ljunggren
Ö
Reilly
MP
Hamsten
A
Ingelsson
E
Cambien
F
Hung
J
Thomas
GN
Boehnke
M
Schunkert
H
Asselbergs
FW
Kastelein
JJ
Gudnason
V
Salomaa
V
Harris
TB
Kooner
JS
Allin
KH
Nordestgaard
BG
Hopewell
JC
Goodall
AH
Ridker
PM
Hólm
H
Watkins
H
Ouwehand
WH
Samani
NJ
Kaptoge
S
Di Angelantonio
E
Harari
O
Danesh
J
IL6R Genetics Consortium Emerging Risk Factors Collaboration
Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies
Lancet
2012
, vol. 
9822
 (pg. 
1205
-
1213
)
79
Bonacina
F
Baragetti
A
Catapano
AL
Norata
GD
Long pentraxin 3: experimental and clinical relevance in cardiovascular diseases
Mediators Inflamm
2013
, vol. 
2013
 pg. 
725102
 
80
Dubin
R
Li
Y
Ix
JH
Shlipak
MG
Whooley
M
Peralta
CA
Associations of pentraxin-3 with cardiovascular events, incident heart failure, and mortality among persons with coronary heart disease: data from the Heart and Soul Study
Am Heart J
2012
, vol. 
2
 (pg. 
274
-
279
)
81
Suzuki
S
Takeishi
Y
Niizeki
T
Koyama
Y
Kitahara
T
Sasaki
T
Sagara
M
Kubota
I
Pentraxin 3, a new marker for vascular inflammation, predicts adverse clinical outcomes in patients with heart failure
Am Heart J
2008
, vol. 
1
 (pg. 
75
-
81
)
82
Barbati
E
Specchia
C
Villella
M
Rossi
ML
Barlera
S
Bottazzi
B
Crociati
L
d'Arienzo
C
Fanelli
R
Garlanda
C
Gori
F
Mango
R
Mantovani
A
Merla
G
Nicolis
EB
Pietri
S
Presbitero
P
Sudo
Y
Villella
A
Franzosi
MG
Influence of pentraxin 3 (PTX3) genetic variants on myocardial infarction risk and PTX3 plasma levels
PLoS One
2012
, vol. 
7
 pg. 
53030
 
83
Herrick
S
Blanc-Brude
O
Gray
A
Laurent
G
Fibrinogen
Int J Biochem Cell Biol
1999
, vol. 
7
 (pg. 
741
-
746
)
84
Davalos
D
Akassoglou
K
Fibrinogen as a key regulator of inflammation in disease
Semin Immunopathol
2012
, vol. 
1
 (pg. 
43
-
62
)
85
Danesh
J
Lewington
S
Thompson
SG
Lowe
GD
Collins
R
Kostis
JB
Wilson
AC
Folsom
AR
Wu
K
Benderly
M
Goldbourt
U
Willeit
J
Kiechl
S
Yarnell
JW
Sweetnam
PM
Elwood
PC
Cushman
M
Psaty
BM
Tracy
RP
Tybjaerg-Hansen
A
Haverkate
F
de Maat
MP
Fowkes
FG
Lee
AJ
Smith
FB
Salomaa
V
Harald
K
Rasi
R
Vahtera
E
Jousilahti
P
Pekkanen
J
D'Agostino
R
Kannel
WB
Wilson
PW
Tofler
G
Arocha-Piñango
CL
Rodriguez-Larralde
A
Nagy
E
Mijares
M
Espinosa
R
Rodriquez-Roa
E
Ryder
E
Diez-Ewald
MP
Campos
G
Fernandez
V
Torres
E
Marchioli
R
Valagussa
F
Rosengren
A
Wilhelmsen
L
Lappas
G
Eriksson
H
Cremer
P
Nagel
D
Curb
JD
Rodriguez
B
Yano
K
Salonen
JT
Nyyssönen
K
Tuomainen
TP
Hedblad
B
Lind
P
Loewel
H
Koenig
W
Meade
TW
Cooper
JA
De Stavola
B
Knottenbelt
C
Miller
GJ
Cooper
JA
Bauer
KA
Rosenberg
RD
Sato
S
Kitamura
A
Naito
Y
Palosuo
T
Ducimetiere
P
Amouyel
P
Arveiler
D
Evans
AE
Ferrieres
J
Juhan-Vague
I
Bingham
A
Schulte
H
Assmann
G
Cantin
B
Lamarche
B
Després
JP
Dagenais
GR
Tunstall-Pedoe
H
Woodward
M
Ben-Shlomo
Y
Davey Smith
G
Palmieri
V
Yeh
JL
Rudnicka
A
Ridker
P
Rodeghiero
F
Tosetto
A
Shepherd
J
Ford
I
Robertson
M
Brunner
E
Shipley
M
Feskens
EJ
Kromhout
D
Dickinson
A
Ireland
B
Juzwishin
K
Kaptoge
S
Lewington
S
Memon
A
Sarwar
N
Walker
M
Wheeler
J
White
I
Wood
A
Fibrinogen Studies Collaboration
Plasma fibrinogen level and the risk of major cardiovascular diseases and nonvascular mortality: an individual participant meta-analysis
JAMA
2005
, vol. 
14
 (pg. 
1799
-
1809
)
86
Sabater-Lleal
M
Huang
J
Chasman
D
Naitza
S
Dehghan
A
Johnson
AD
Teumer
A
Reiner
AP
Folkersen
L
Basu
S
Rudnicka
AR
Trompet
S
Mälarstig
A
Baumert
J
Bis
JC
Guo
X
Hottenga
JJ
Shin
SY
Lopez
LM
Lahti
J
Tanaka
T
Yanek
LR
Oudot-Mellakh
T
Wilson
JF
Navarro
P
Huffman
JE
Zemunik
T
Redline
S
Mehra
R
Pulanic
D
Rudan
I
Wright
AF
Kolcic
I
Polasek
O
Wild
SH
Campbell
H
Curb
JD
Wallace
R
Liu
S
Eaton
CB
Becker
DM
Becker
LC
Bandinelli
S
Räikkönen
K
Widen
E
Palotie
A
Fornage
M
Green
D
Gross
M
Davies
G
Harris
SE
Liewald
DC
Starr
JM
Williams
FM
Grant
PJ
Spector
TD
Strawbridge
RJ
Silveira
A
Sennblad
B
Rivadeneira
F
Uitterlinden
AG
Franco
OH
Hofman
A
van Dongen
J
Willemsen
G
Boomsma
DI
Yao
J
Swords Jenny
N
Haritunians
T
McKnight
B
Lumley
T
Taylor
KD
Rotter
JI
Psaty
BM
Peters
A
Gieger
C
Illig
T
Grotevendt
A
Homuth
G
Völzke
H
Kocher
T
Goel
A
Franzosi
MG
Seedorf
U
Clarke
R
Steri
M
Tarasov
KV
Sanna
S
Schlessinger
D
Stott
DJ
Sattar
N
Buckley
BM
Rumley
A
Lowe
GD
McArdle
WL
Chen
MH
Tofler
GH
Song
J
Boerwinkle
E
Folsom
AR
Rose
LM
Franco-Cereceda
A
Teichert
M
Ikram
MA
Mosley
TH
Bevan
S
Dichgans
M
Rothwell
PM
Sudlow
CL
Hopewell
JC
Chambers
JC
Saleheen
D
Kooner
JS
Danesh
J
Nelson
CP
Erdmann
J
Reilly
MP
Kathiresan
S
Schunkert
H
Morange
PE
Ferrucci
L
Eriksson
JG
Jacobs
D
Deary
IJ
Soranzo
N
Witteman
JC
de Geus
EJ
Tracy
RP
Hayward
C
Koenig
W
Cucca
F
Jukema
JW
Eriksson
P
Seshadri
S
Markus
HS
Watkins
H
Samani
NJ
Wallaschofski
H
Smith
NL
Tregouet
D
Ridker
PM
Tang
W
Strachan
DP
Hamsten
A
O'Donnell
CJ
A Multi-Ethnic Meta-Analysis of Genome-Wide Association Studies in over 100,000 subjects identifies 23 fibrinogen-associated loci but no strong evidence of a causal association between circulating fibrinogen and cardiovascular disease
Circulation
2013
, vol. 
12
 (pg. 
1310
-
1324
)
87
Kannel
WB
Schwartz
MJ
McNamara
PM
Blood pressure and risk of coronary heart disease: the Framingham study
Dis Chest
1969
, vol. 
1
 (pg. 
43
-
52
)
88
Yusuf
S
Sleight
P
Pogue
J
Bosch
J
Davies
R
Dagenais
G
Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators
N Engl J Med
2000
, vol. 
3
 (pg. 
145
-
153
)
89
Whitlock
G
Lewington
S
Sherliker
P
Clarke
R
Emberson
J
Halsey
J
Qizilbash
N
Collins
R
Peto
R
Prospective Studies Collaboration
Body-mass index and cause-specific mortality in 900 000 adults: collaborative analyses of 57 prospective studies
Lancet
2009
, vol. 
9669
 (pg. 
1083
-
1096
)
90
Nordestgaard
BG
Palmer
TM
Benn
M
Zacho
J
Tybjaerg-Hansen
A
Davey Smith
G
Timpson
NJ
The effect of elevated body mass index on ischemic heart disease risk: causal estimates from a Mendelian randomisation approach
PLos Med
2012
, vol. 
5
 pg. 
1001212
 
91
Franco
OH
Steyerberg
EW
Hu
FB
Mackenbach
J
Nusselder
W
Associations of diabetes mellitus with total life expectancy and life expectancy with and without cardiovascular disease
Arch Intern Med
2007
, vol. 
11
 (pg. 
1145
-
1151
)
92
Laakso
M
Kuusisto
J
Epidemiological evidence for the association of hyperglycaemia and atherosclerotic vascular disease in non-insulin-dependent diabetes mellitus
Ann Med
1996
, vol. 
5
 (pg. 
415
-
418
)
93
Riddle
MC
Effects of intensive glucose lowering in the management of patients with type 2 diabetes mellitus in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial
Circulation
2010
, vol. 
8
 (pg. 
844
-
846
)
94
Jansen
H
Loley
C
Lieb
W
Pencina
MJ
Nelson
CP
Kathiresan
S
Peloso
GM
Voight
BF
Reilly
MP
Assimes
TL
Boerwinkle
E
Hall
AS
Hengstenberg
C
Laaksonen
R
McPherson
R
Roberts
R
Thorsteinsdottir
U
Peters
A
Thompson
JR
König
IR
Vasan
RS
Erdmann
J
Samani
NJ
Schunkert
H
on behalf of CARDIoGRAM
Genetic variants primarily associated with type 2 diabetes also affect coronary artery disease risk
 
(in revision)
95
Preis
SR
Pencina
MJ
Hwang
S-J
D'Agostino
RB
Sr
Savage
PJ
Levy
D
Fox
CS
Trends in cardiovascular disease risk factors in individuals with and without diabetes mellitus in the Framingham Heart Study
Circulation
2009
, vol. 
3
 (pg. 
212
-
220
)
96
Preuss
M
König
IR
Thompson
JR
Erdmann
J
Absher
D
Assimes
TL
Blankenberg
S
Boerwinkle
E
Chen
L
Cupples
LA
Hall
AS
Halperin
E
Hengstenberg
C
Holm
H
Laaksonen
R
Li
M
März
W
McPherson
R
Musunuru
K
Nelson
CP
Burnett
MS
Epstein
SE
O'Donnell
CJ
Quertermous
T
Rader
DJ
Roberts
R
Schillert
A
Stefansson
K
Stewart
AF
Thorleifsson
G
Voight
BF
Wells
GA
Ziegler
A
Kathiresan
S
Reilly
MP
Samani
NJ
Schunkert
H
CARDIoGRAM Consortium
Design of the Coronary ARtery DIsease Genome-Wide Replication And Meta-Analysis (CARDIoGRAM) Study: A Genome-wide association meta-analysis involving more than 22 000 cases and 60 000 controls
Circ Cardiovasc Genet
2010
, vol. 
5
 (pg. 
475
-
483
)
97
Brouilette
S
Singh
RK
Thompson
JR
Goodall
AH
Samani
NJ
“White cell telomere length and risk of premature myocardial infarction
Arterioscler Thromb Vasc Biol
2003
, vol. 
5
 (pg. 
842
-
846
)
98
Brouilette
SW
Moore
JS
McMahon
AD
Thompson
JR
Ford
I
Shepherd
J
Packard
CJ
Samani
NJ
West of Scotland Coronary Prevention Study Group
Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland Primary Prevention Study: a nested case-control study
Lancet
2007
, vol. 
9556
 (pg. 
107
-
114
)
99
Fitzpatrick
AL
Kronmal
RA
Gardner
JP
Psaty
BM
Jenny
NS
Tracy
RP
Walston
J
Kimura
M
Aviv
A
Leukocyte telomere length and cardiovascular disease in the cardiovascular health study
Am J Epidemiol
2007
, vol. 
1
 (pg. 
14
-
21
)
100
Palmer
TM
Nordestgaard
BG
Benn
M
Tybjærg-Hansen
A
Davey Smith
G
Lawlor
DA
Timpson
NJ
Association of plasma uric acid with ischaemic heart disease and blood pressure: mendelian randomisation analysis of two large cohorts
BMJ
2013
, vol. 
347
 pg. 
4262
 
101
Stender
S
Frikke-Schmidt
R
Nordestgaard
BG
Grande
P
Tybjaerg-Hansen
A
Genetically elevated bilirubin and risk of ischaemic heart disease: three Mendelian randomization studies and a meta-analysis
J Intern Med
2013
, vol. 
1
 (pg. 
59
-
68
)
102
van Meurs
JBJ
Pare
G
Schwartz
SM
Hazra
A
Tanaka
T
Vermeulen
SH
Cotlarciuc
I
Yuan
X
Mälarstig
A
Bandinelli
S
Bis
JC
Blom
H
Brown
MJ
Chen
C
Chen
Y-D
Clarke
RJ
Dehghan
A
Erdmann
J
Ferrucci
L
Hamsten
A
Hofman
A
Hunter
DJ
Goel
A
Johnson
AD
Kathiresan
S
Kampman
E
Kiel
DP
Kiemeney
LALM
Chambers
JC
Kraft
P
Lindemans
J
McKnight
B
Nelson
CP
O'Donnell
CJ
Psaty
BM
Ridker
PM
Rivadeneira
F
Rose
LM
Seedorf
U
Siscovick
DS
Schunkert
H
Selhub
J
Ueland
PM
Vollenweider
P
Waeber
G
Waterworth
DM
Watkins
H
Witteman
JCM
den Heijer
M
Jacques
P
Uitterlinden
AG
Kooner
JS
Rader
DJ
Reilly
MP
Mooser
V
Chasman
DI
Samani
NJ
Ahmadi
KR
Common genetic loci influencing plasma homocysteine concentrations and their effect on risk of coronary artery disease
Am J Clin Nutr
2013
, vol. 
3
 (pg. 
668
-
676
)
103
Dastani
Z
Johnson
T
Kronenberg
F
Nelson
CP
Assimes
TL
März
W
Richards
JB
CARDIoGRAM Consortium, ADIPOGen Consortium
The shared allelic architecture of adiponectin levels and coronary artery disease
Atherosclerosis
2013
, vol. 
1
 (pg. 
145
-
148
)
104
Benn
M
Tybjaerg-Hansen
A
McCarthy
MI
Jensen
GB
Grande
P
Nordestgaard
BG
Nonfasting glucose, ischemic heart disease, and myocardial infarction: a Mendelian randomization study
J Am Coll Cardiol
2012
, vol. 
25
 (pg. 
2356
-
2365
)
105
Tang
WHW
Hartiala
J
Fan
Y
Wu
Y
Stewart
AFR
Erdmann
J
Kathiresan
S
Roberts
R
McPherson
R
Allayee
H
Hazen
SL
CARDIoGRAM Consortium
Clinical and genetic association of serum paraoxonase and arylesterase activities with cardiovascular risk
Arterioscler Thromb Vasc Biol
2012
, vol. 
11
 (pg. 
2803
-
2812
)

Supplementary data