Abstract

There is a strong and ever-growing body of evidence regarding the use of pharmacogenomics to inform cardiovascular pharmacology. However, there is no common position taken by international cardiovascular societies to unite diverse availability, interpretation, and application of such data, nor is there recognition of the challenges of variation in clinical practice between countries within Europe. Aside from the considerable barriers to implementing pharmacogenomic testing and the complexities of clinically actioning results, there are differences in the availability of resources and expertise internationally within Europe. Diverse legal and ethical approaches to genomic testing and clinical therapeutic application also require serious thought. As direct-to-consumer genomic testing becomes more common, it can be anticipated that data may be brought in by patients themselves, which will require critical assessment by the clinical cardiovascular prescriber. In a modern, pluralistic and multi-ethnic Europe, self-identified race/ethnicity may not be concordant with genetically detected ancestry and thus may not accurately convey polymorphism prevalence. Given the broad relevance of pharmacogenomics to areas, such as thrombosis and coagulation, interventional cardiology, heart failure, arrhythmias, clinical trials, and policy/regulatory activity within cardiovascular medicine, as well as to genomic and pharmacology subspecialists, this position statement attempts to address these issues at a wide-ranging level.

Introduction

The completion of the human genome project in 2003 heralded an age of genomic promise.1,2 Advances in genomic knowledge and sequencing technology have provided vital tools for the implementation of personalized medicine; i.e. the ability to use genetic information to select the right pharmacological agent, to be used at the right dose, for the right person, and at the right time, thereby maximizing the efficacy of the intervention and minimizing adverse drug reactions (ADR).2

Pharmacogenomics (PGx) describes the role of genomic variation in drug response, which can relate to both pharmacokinetic and pharmacodynamic properties. Pharmacokinetics constitutes the effect of the body on the drug, categorized into effects on absorption, distribution, metabolism, and excretion of the drug. Pharmacodynamics reflects the effect of the drug on the body. Both pharmacokinetics and pharmacodynamics influence efficacy, effectiveness, and toxicity. Many drugs are metabolized in the liver by cytochrome P450 (CYP) enzymes. These enzymes are encoded by various genes named according to their family (e.g. ‘2’) and subfamily (e.g. ‘C’), based on the amino acid structure. A full code indicates a specific gene (e.g. CYP2C9). Other drug metabolizing enzymes, receptors, and transporters also play an important role in drug response. Indeed, whole-genome approaches, including sequencing, are likely to identify new pharmacogenes, including in therapeutic target genes, which determine drug efficacy and/or safety. It is inevitable that the number of pharmacogenes with evidence of clinical utility will continue to increase, which makes pharmacogenomics a dynamic area.

The cost of genome sequencing has decreased exponentially with the emergence of next-generation and third-generation sequencing technology, from 2.7 billion USD in 2003 to sequence the first human genome (which took 13 years) to <1000 USD to sequence a whole genome in 2019.3–5 In order to implement personalized medicine, three main tools to target discovery in PGx studies are used: genome-wide association studies (GWASs), candidate gene analyses, and next-generation sequencing (high throughput parallel sequencing).6 The first is an agnostic approach that locates a genome-wide signal to a locus, while the candidate gene approach starts from a gene hypothesized to be significant. This will often be a gene that encodes protein targets of the drug, or proteins that are involved in drug pharmacokinetics. When genetic loci or single-nucleotide polymorphisms (SNPs) of interest are identified, this should be replicated and may be followed by functional validation studies, although this is not always necessary, depending on the predictive accuracy of the variant. Evidence of clinical utility needs to be gathered which depends on the clinical phenotype and the gene-drug pair. The present rate of big data-based discovery, and the scale and complexity of genomic information interpretation and translation into useful therapeutic interventions present considerable challenges. The expectations of gold-standard evidence from randomized controlled trials (RCTs) for PGx may not be realistic (or indeed necessary) in PGx, as each trial would look at one or a few gene variant-drug pairs.7,8

Though PGx guided therapy has been endorsed by both regulators and international consortia, there is discordance in recommendations, and PGx has remained an area of specialist interest.9

Given the rapid advances which are occurring, there is a need for professional bodies and health systems to support prescribers in understanding, interpreting, and implementing evidence-based PGx in cardiovascular medicine. This is particularly timely as many prescribers begin to find themselves increasingly confronted with patients able to access PGx information through direct-to-consumer genetic testing.10 Moreover, prescribers may not have access to confirmatory testing within their place of work. Given the broad relevance of PGx to areas such as thrombosis and coagulation, interventional cardiology, heart failure, arrhythmias, clinical trials, and policy/regulatory activity within cardiovascular medicine, as well as to genomic and pharmacology subspecialists, this position statement attempts to address these issues at a wide-ranging level. The following sections address evidence for specific drug-gene pairs, as well as broad genomic approaches in cardiovascular medicine, and the potential role of genomics in clinical trials. The focus is on translational aspects most likely to be encountered by non-genomically trained practitioners in day-to-day practice.

Anticoagulants

Warfarin and other vitamin K antagonists

The coumarin derivatives, warfarin, acenocoumarol, and phenprocoumon, are used to prevent or treat thromboembolism. There are differences across the World and between European countries regarding which coumarin oral anticoagulant is preferred, but collectively they remain commonly prescribed, despite the rapidly increasing use of direct oral anticoagulants (DOACs).11,12 Coumarins inhibit vitamin K epoxide reductase complex subunit 1 (VKORC1), leading to hypofunctional clotting factors II, VII, IX, and X.13 Coumarin derivatives are all administered as a racemate. For warfarin, the S-warfarin enantiomer is preferentially metabolized by CYP2C9 and is ∼3–5x more potent than R-warfarin.14–16 Both acenocoumarol enantiomers are principally metabolized by CYP2C9 and, while S-acenocoumarol is more active, its short elimination half-life (1.8 h) means the majority of anticoagulant effect is through R-acenocoumarol.13 For phenprocoumon, both CYP2C9 and CYP3A4 are involved in its metabolism.17 Coumarin dose requirements to maintain an international normalized ratio (INR) between 2.0 and 3.0 are highly variable between individuals. For example, warfarin stable dose (WSD) ranges at least 25-fold (0.6–15.5 mg/day), and real-world registry data of patients anticoagulated for atrial fibrillation demonstrated a mean time in the therapeutic INR range (TTR) of 65% (±20%).18 This high inter-individual variation and narrow therapeutic index increase susceptibility to adverse events. Notably, 6–7% of patients prescribed warfarin were hospitalized due to bleeding over a mean follow-up of 425 days, with supra-therapeutic INRs increasing bleeding risk.19–21 Decreased TTR is also a predictor for increased ischaemic stroke, other thromboembolic events, major bleeding, and mortality.22,23

For warfarin, approximately ∼55–60% of variation in WSD can be principally explained by genetic variation in VKORC1 (∼25%) and CYP2C9 (∼15%), supplemented by CYP4F2*3 (∼1–7%) and clinical factors (e.g. age, body mass index, smoking, interacting drugs, collectively <20%)24–26 (Table 1).

Table 1

Clinical trials testing genotype-guided warfarin dosing 

GeneVariant(s)Functional effectClinical effect
VKORC1−1639G>A (rs9923231)Reduced VKORC1 transcriptionReduced warfarin requirments41
CYP2C9*2, *3, *5, *6, *8, *11Decreased CYP2C9 metabolic activityReduced warfarin requirements42,43
CYP4F2*3 (V433M)Decreased hepatic CYP4F2 levels and reduced vitamin K hydroxylationHigher warfarin requirments44
GeneVariant(s)Functional effectClinical effect
VKORC1−1639G>A (rs9923231)Reduced VKORC1 transcriptionReduced warfarin requirments41
CYP2C9*2, *3, *5, *6, *8, *11Decreased CYP2C9 metabolic activityReduced warfarin requirements42,43
CYP4F2*3 (V433M)Decreased hepatic CYP4F2 levels and reduced vitamin K hydroxylationHigher warfarin requirments44
Table 1

Clinical trials testing genotype-guided warfarin dosing 

GeneVariant(s)Functional effectClinical effect
VKORC1−1639G>A (rs9923231)Reduced VKORC1 transcriptionReduced warfarin requirments41
CYP2C9*2, *3, *5, *6, *8, *11Decreased CYP2C9 metabolic activityReduced warfarin requirements42,43
CYP4F2*3 (V433M)Decreased hepatic CYP4F2 levels and reduced vitamin K hydroxylationHigher warfarin requirments44
GeneVariant(s)Functional effectClinical effect
VKORC1−1639G>A (rs9923231)Reduced VKORC1 transcriptionReduced warfarin requirments41
CYP2C9*2, *3, *5, *6, *8, *11Decreased CYP2C9 metabolic activityReduced warfarin requirements42,43
CYP4F2*3 (V433M)Decreased hepatic CYP4F2 levels and reduced vitamin K hydroxylationHigher warfarin requirments44

At least 14 clinical trials testing genotype-guided warfarin dosing have been conducted, employing various algorithms that incorporate genetic (VKORC1, CYP2C9 ± CYP4F2) and clinical covariates.25,27,28 The largest European RCT of warfarin PGx was conducted by the EU-PACT collaboration and recruited patients starting warfarin for atrial fibrillation or venous thromboembolism (VTE).29 EU-PACT compared genotype-guided warfarin dosing on days 1–5 in 227 patients to standard warfarin loading in 228 patients, with all subsequent dosing in both arms according to routine practice, and found that the primary endpoint of 12-week TTR was significantly higher in the genotype-guided arm (67.4% vs. 60.3%, P < 0.001).29 There was no significant difference in bleeding or thromboembolic events. In EU-PACT, 99% of participants were Caucasian, and point-of-care (POC) genotyping was utilized.29 A subsequent UK-based real-world implementation study with POC genotyping similarly found genotype-guided dosing increased warfarin TTR by 7%.30 A recent meta-analysis incorporating EU-PACT data shows that genotype-guided warfarin dosing compares favourably to DOACs in cluster rank plots considering both thromboembolism and bleeding.31

In contrast, the US COAG trial did not find any significant difference between genotype-guided and clinically guided dosing algorithms (4-week TTR 45.2% vs. 45.4%, P = 0.91).32 An important distinction between the two trials is that 27% of patients recruited to COAG were African-American, but only the CYP2C9 alleles common in Caucasians, *2 and *3, were assessed. Genotype-guided African-American patients in COAG fared less well than those clinically dosed (4-week TTR 35.2% vs. 43.5%, P = 0.01), highlighting the importance of patient ethnicity in considering CYP2C9 variants.32 Another difference between the trials is that the US COAG trial comparator arm used a clinical dosing algorithm, whereas EU-PACT used standard care. However, clinical dosing algorithms have not been validated themselves vs. usual care and the comparator arm TTR was lower in COAG than in EU-PACT.

Genetics informatics trial was the largest and most recent RCT; it recruited patients undergoing elective hip or knee arthroplasty treated with perioperative warfarin and randomized them to genotype- (n = 808) or clinically guided (n = 789) dosing on days 1–11. Patients were genotyped for VKORC1 −1639G>A, CYP2C9*2, CYP2C9*3, and CYP4F2 V433M. The primary composite endpoint of INR ≥ 4, major bleeding, death (all within 30 days) plus VTE within 60 days, occurred in 10.8% and 14.7% of patients in the genotype and clinically guided groups, respectively (P = 0.02). This result was driven mainly by a decrease in INR ≥ 4 (P = 0.04) and borderline reduction in major bleeding (P = 0.06).27 Given the continued high real-world use of warfarin, these results are considered clinically relevant.

To aid implementation, there is supportive evidence for the cost-effectiveness of genotype-guided warfarin dosing, and multigene-drug testing will likely be even more advantageous.33,34 Clinical guidelines for genotype-guided warfarin dosing have been developed by both the Dutch Pharmacogenetics Working Group (DPWG) and Clinical Pharmacogenetics Implementation Consortium (CPIC).35,36

The EU-PACT collaboration also conducted RCTs of acenocoumarol and phenprocoumon PGx.37 Due to low enrolment, they were combined for analysis. Though the primary outcome of 12-week TTR in the 484 eligible participants did not differ between the genotype-guided and clinically guided dosing arms (61.6% vs. 60.2%, P = 0.52), follow-up analysis demonstrated that the acenocoumarol dosing algorithm, designed for a Dutch population, overestimated dose in Greek ancestry patients.37–39

On balance, we advocate genotype-guided dosing when commencing warfarin. To optimize the effectiveness of a genotype-informed strategy, genetic results should be available on the first day of warfarin dosing. This may require the use of point-of-care testing, or pre-emptive genotyping.

Although patient ethnicity is important in warfarin PGx, self-reported ethnicity may not always adequately represent genetic ancestry, for instance in admixed individuals. Genetic characterization to facilitate appropriate use of ethnicity-specific warfarin algorithms may be preferable, but arguably difficult to achieve in clinical practice.40 Therefore, rather than genotype for specific alleles in patients from different ethnic backgrounds, a better approach would be to genotype for all the relevant CYP2C9, VKORC1, and CYP4F2 alleles, irrespective of ethnicity/ancestry, and determine dose requirements using a ‘universal’ algorithm, which will require development.

Key clinical positions:

  • Prospective genotyping prior to warfarin initiation is advised where possible.

  • If genotypic information is already available, it should be used to guide warfarin initiation dosing.

Antiplatelets

Antiplatelet therapy is the backbone of treatment for atherothrombotic diseases, such as coronary artery disease and stroke. In acute coronary syndromes and patients undergoing percutaneous coronary intervention, dual antiplatelet therapy with aspirin and a P2Y12 receptor antagonist is usually recommended.45,46 P2Y12 receptor antagonists include clopidogrel, prasugrel, and ticagrelor. The more potent P2Y12 receptor antagonists, prasugrel, and ticagrelor, are usually preferred over clopidogrel in high-risk cases on the basis of improved cardiovascular outcomes, albeit with an increased bleeding risk.47 In stroke, single-antiplatelet therapy with clopidogrel alone is usually preferred for long-term secondary prevention.48 However, the effectiveness of antiplatelet therapy is limited by variability in patient response; particularly to clopidogrel, partly attributed to genetic variation.

Clopidogrel is a prodrug, transformed to its active metabolite in two sequential CYP2C19 dependent steps. Up to a third of patients have reduced enzymatic activity secondary to a loss of function variant in CYP2C19 (e.g. *2 or *3).49 This is associated with high on-treatment platelet reactivity and an increased risk of ischaemic events.50,51

Knowledge of an individual’s genetic profile can allow personalized antiplatelet strategies in order to optimize the balance of beneficial to adverse outcomes. However, genotypic variation does not always translate into adverse clinical outcomes as, unlike platelet function testing, genotyping does not provide a direct measure of response to therapy or an assessment of non-genetic factors on platelet function. Yet platelet function testing is limited by the need to perform testing while already on treatment and lacks standard reference values and thresholds. Given the large number of patients prescribed antiplatelet agents, use of PGx to personalize treatment could have a significant effect at a population level. A PGx polygenic risk score has also been proposed as an effective risk stratification tool, and merits further investigation in a prospective clinical setting.52

Clinical trials have used both phenotype-guided (platelet function testing while on treatment) and genotype-guided (CYP2C19 status assessed before/during treatment) approaches. Meta-analysis has shown phenotype-guided approaches in patients presenting with acute coronary syndrome or with coronary artery disease undergoing percutaneous coronary intervention are not superior to standard care.53 This is unsurprising given the limitations discussed. In contrast, meta-analysis of six randomized controlled trials using genotypic-based treatment, with 3764 patients undergoing percutaneous coronary intervention or with acute coronary syndromes, has shown a significant reduction in net adverse clinical events [hazard ratio (HR) 0.65; 95% confidence interval (CI) 0.45–0.95; P = 0.03], including reduced major adverse cardiovascular events (HR 0.59; 95% CI 0.43–0.82; P < 0.01) and reduced bleeding events (HR 0.75; 95% CI 0.61–0.93; P < 0.01).54 This meta-analysis pooled studies that use different strategies and different patient populations, including those with stable and acute coronary syndromes.

Notably, the large POPular Genetics RCT found that genotype-guided therapy in patients undergoing primary PCI was non-inferior to standard treatment with prasugrel or ticagrelor in terms of a combined primary outcome of thrombotic and bleeding events, and risk of bleeding was reduced.55 The recent TAILOR-PCI trial randomized 5302 PCI patients, 1849 of whom carried loss of function CYP2C19 alleles, to genotype-guided antiplatelet therapy vs. conventional therapy with clopidogrel.56 The composite cardiovascular primary endpoint was 4.0% in the genotype-guided and 5.9% in the non-genotype-guided cohort at 1-year follow-up (HR 0.66; 95% CI 0.43–1.02; P = 0.06).56 Unfortunately, the study was underpowered due to lower event rates than expected. However, a post hoc analysis found that the primary outcome was significantly reduced during the first 3 months after PCI (HR 0.21; 95% CI 0.08–0.54; P = 0.001), suggesting higher modifiable risk in the months immediately following stenting.56 This includes the highest risk period for early in-stent thrombosis, which is the first 30 days.57

This evidence base suggests that the genotype-guided approach could be superior to standard care in terms of both cardiovascular outcomes and reduced bleeding events, particularly in the months immediately following PCI. The availability of rapid diagnostic genetic tests means such strategies are a viable option in routine clinical practice. In other settings, such as stroke, there is still a lack of prospective large-scale RCT data. However, similar benefits might be anticipated, a hypothesis supported by systematic review and meta-analysis data.58,59

Clopidogrel PGx profiling for CYP2C19 remains only recommended in specific high-risk situations, such as patients with acute coronary syndrome undergoing percutaneous coronary intervention, or presenting with recurrent adverse events, and only if results are likely to change the treatment strategy. If patients are considered intermediate or poor metabolizers of clopidogrel, alternative antiplatelet agents are recommended.60 More large-scale trials in patients with coronary artery disease (e.g. GUARANTEE study) are ongoing and will inform this recommendation further. Additional prospective data from large-scale trials for other indications, such as stroke, are awaited before clear recommendations can be made (e.g. PLATELET study). Given the disease prevalence, even a small improvement with PGx use may translate to meaningful population-level health gains.

Key clinical positions:

  • Clopidogrel should be avoided in patients who are known to be intermediate or poor metabolisers from existing genotypic information.

  • Genotyping high-risk cardiovascular patients (either high risk of thrombosis or bleed) prior to prescribing clopidogrel should be considered where possible, particularly to prevent post-stent thrombosis.

Statins

Statins are the most commonly prescribed lipid-lowering medication for the treatment and prevention of cardiovascular disease (CVD).61,62 Meta-analyses show a consistent relationship between the extent of low-density lipoprotein cholesterol (LDL-C) reduction and CVD risk reduction, irrespective of statin choice, dose, or baseline risk.63–65 Statins are well-tolerated in most individuals but can rarely cause severe hepatotoxicity, and are associated with the development of diabetes mellitus.66 Statin-associated muscle symptoms (SAMS) are the most commonly reported side-effect and are a common reason for treatment discontinuation.67,68 SAMS range from mild myalgia without an elevation in creatine kinase to life-threatening rhabdomyolysis or autoimmune-necrotizing myositis.69,70 The underlying mechanism of muscle injury is increased systemic statin exposure with intracellular skeletal myocyte entry and disruption of muscle function.71

Statin disposition is dependent on multiple enzymes and transporters, with their relative importance varying between drugs. Solute carrier anion transporter family 1B1 (SLCO1B1) encodes organic anion transporting polypeptide 1B1 (OATP1B1) which is central to the hepatic uptake and consequent elimination of statins. A GWAS in patients with simvastatin-induced myopathy identified an increased risk of myopathy in heterozygous and particularly homozygous carriers of the reduced function SLCO1B1*5 variant, which significantly increases plasma concentrations of all statins, except fluvastatin (Table 2).72 In addition to this gene dose trend, a simvastatin dose trend was observed with the risk of myopathy increasing by 2.6 and 4.3 per copy of SLCO1B1*5 in patients on simvastatin 40 and 80 mg daily, respectively.72

Table 2

Clinical trials testing genotype-guided warfarin dosing 

StatinIncrease in area under the curve in SLCO1B1*5 (loss-of-function) homozygotes87–91
Simvastatin221%
Lovastatin186%
Pitavastatin208%
Atorvastatin145%
Pravastatin91%
Rosuvastatin65%
Fluvastatin19%, non-significant
StatinIncrease in area under the curve in SLCO1B1*5 (loss-of-function) homozygotes87–91
Simvastatin221%
Lovastatin186%
Pitavastatin208%
Atorvastatin145%
Pravastatin91%
Rosuvastatin65%
Fluvastatin19%, non-significant
Table 2

Clinical trials testing genotype-guided warfarin dosing 

StatinIncrease in area under the curve in SLCO1B1*5 (loss-of-function) homozygotes87–91
Simvastatin221%
Lovastatin186%
Pitavastatin208%
Atorvastatin145%
Pravastatin91%
Rosuvastatin65%
Fluvastatin19%, non-significant
StatinIncrease in area under the curve in SLCO1B1*5 (loss-of-function) homozygotes87–91
Simvastatin221%
Lovastatin186%
Pitavastatin208%
Atorvastatin145%
Pravastatin91%
Rosuvastatin65%
Fluvastatin19%, non-significant

The clinical association between simvastatin-induced myopathy and SLCO1B1*5 has been confirmed in a recent meta-analysis, which failed to identify any other genetic loci.73 There is ongoing uncertainty over the effect of SLCO1B1*5 on other statins as, in the example of atorvastatin, alternative anion transporters are partially responsible for hepatic uptake and its intrinsic myotoxicity may be less than compared to simvastatin.74,75 Interestingly, recent work has associated SLCO1B1*5 with rosuvastatin-induced myotoxicity in Han Chinese patients, despite it not previously being associated with myalgia in patients of European descent.76–78 This may be secondary to increased rosuvastatin exposure in individuals of Asian ancestry, which is partially explained by polymorphisms in ABCG2, an efflux transporter encoding breast cancer resistance protein (BCRP).79

Other factors, such as drug-drug interactions, exercise, neuromuscular disorders, mitochondrial impairment, mevalonate pathway perturbation, immune-mediated toxicity, muscle transcriptomics, and vitamin D deficiency can all contribute to myotoxicity.71 It is likely that SAMS often result from a combination of these and genetic factors.

There is little prospective data available to inform clinical decision making. Peyser et al.80 randomized 159 patients with prior statin myalgia to receive SLCO1B1 genotype informed statin therapy (GIST) vs. usual care. Rosuvastatin, pravastatin, or fluvastatin were recommended for SLCO1B1*5 carriers, whereas non-carriers were recommended to try any statin that they had not tried in the past. Over an 8-month follow-up, those who were randomized to GIST showed increased statin re-initiation and reduced LDL-C, but without an increase in self-reported medication adherence.80 Vassy et al.81 recently completed an RCT of 408 patients treated in primary care settings, to assess potential unintended harms of PGx testing. The findings were reassuring, showing that knowledge of SLCO1B1 genotype did not deter from appropriate initiation of statins in accordance with guidance, and did not result in inferior LDL-C at the 1 year endpoint.81

The minor allele frequency of rs4149056 (SLCO1B1*5) is approximately 1%, 8%, and 16% in African, Asian, and European populations, respectively,82 and so a substantial proportion of the population may be at risk. Both CPIC and DPWG, as well as the European summary of product characteristics, recommend that individuals carrying SLCO1B1*5 avoid high-dose simvastatin, but the suggested clinical approach differs.83,84 The CPIC guidelines recommend a lower simvastatin starting dose or an alternative statin in heterozygous carriers, alongside consideration of routine creatine kinase surveillance in those who are homozygous.85 In contrast, the DPWG recommends that homozygotes avoid simvastatin entirely.86 The DPWG guideline also recommends avoiding atorvastatin for individuals with other clinical risk factors for SAMS.86 The EU summary of product characteristic for simvastatin states ‘Where available, genotyping for the presence of the C allele should be considered as part of the benefit-risk assessment prior to prescribing 80 mg simvastatin for individual patients and high doses avoided in those found to carry the CC genotype.’

As the current evidence base is limited to specific drug-gene pairs and largely retrospective, PGx testing to assess the risk of SAMS has not yet transitioned into routine clinical care, and PGx testing while on statin therapy is not usually recommended.

Key clinical positions:

  • Avoid high-dose simvastatin (80 mg) and consider an alternative statin of equivalent LDL lowering efficacy in patients known to be homozygous for the SLCO1B1*5 reduced function variant.

Beta-blockers

Several beta-blockers, including metoprolol, carvedilol, and propranolol are metabolized by CYP2D6, a phase I drug-metabolizing enzyme implicated in the metabolism of 20–25% of clinically prescribed drugs,92–94 and encoded by the highly polymorphic gene, CYP2D6. Of these, metoprolol is most highly dependent on CYP2D6 for its elimination, with 70–80% of an oral dose undergoing CYP2D6-mediated biotransformation92 (Table 3). Other beta-blockers, such as atenolol and bisoprolol, undergo either no or negligible CYP2D6 metabolism.92,95

Table 3

Clinical trials testing genotype-guided warfarin dosing 

CYP2D6 metabolizer phenotypeFold difference in metoprolol exposure (compared with normal metabolizer)109
Ultra-rapid∼0.4
Intermediate∼2.5
Poor∼4.9
CYP2D6 metabolizer phenotypeFold difference in metoprolol exposure (compared with normal metabolizer)109
Ultra-rapid∼0.4
Intermediate∼2.5
Poor∼4.9
Table 3

Clinical trials testing genotype-guided warfarin dosing 

CYP2D6 metabolizer phenotypeFold difference in metoprolol exposure (compared with normal metabolizer)109
Ultra-rapid∼0.4
Intermediate∼2.5
Poor∼4.9
CYP2D6 metabolizer phenotypeFold difference in metoprolol exposure (compared with normal metabolizer)109
Ultra-rapid∼0.4
Intermediate∼2.5
Poor∼4.9

Several studies have investigated the clinical effects of CYP2D6 in patients taking metoprolol, mostly for heart failure (HF) or hypertension (HTN). The tolerated maintenance metoprolol dose in CYP2D6 poor metabolizers (PMs) relative to extensive (normal) metabolizers (EMs) is lower in some, but not all studies.96–101 A decreased heart rate (HR) has been frequently reported in intermediate metabolizers (IMs) and particularly PMs relative to EMs, and an increased incidence of (mostly asymptomatic) bradycardia (HR < 60 b.p.m.) has been reported in CYP2D6 PMs and occasionally IMs in some but not all studies.96,98,99,101–104 No association between CYP2D6 and systolic blood pressure in metoprolol users has been identified.96,98,101,103 However, at least three studies, including the two largest to date have linked CYP2D6 PM status to a ∼5 mmHg reduction in diastolic blood pressure relative to EMs, although this has not been replicated in all studies.96,98,99,101–103,105 Bar an early small case–control study, clinically apparent adverse events have not been associated with CYP2D6 PMs or IMs.98,99,101,102,105–107 One limitation of some of these studies is a reliance on a single, albeit common, loss-of-function variant, CYP2D6*4 (rs3892097) for assigning CYP2D6 predicted function.96,97,101

Due to the lower prevalence of CYP2D6 ultra-rapid metabolizers (UMs) compared to IM/PMs, less research has specifically focused on this group, although HR and blood pressure do not appear to be notably different from EMs with metoprolol.102,103 One study of 187 patients post myocardial infarction related CYP2D6 duplications to ventricular rhythm disturbances, although this group also had a higher proportion of ST-elevation myocardial infarction.104

The US Food & Drug Administration (FDA)-approved metoprolol product label states that CYP2D6 (genotype)-dependent metabolism seems to have little influence on the safety or tolerability of metoprolol.108 Nonetheless, the label also notes that strong CYP2D6 drug inhibitors are expected to mimic CYP2D6 PMs and recommends caution with co-administration of potent CYP2D6 inhibitors.108 This is one example of a trend in drug labelling that urges more (appropriate) caution for drug-drug interactions than for genetic polymorphisms with the same effect. The DPWG guidance for metoprolol-CYP2D6 recommends slower dose titration and reduced maximal doses in IMs and PMs in the event of symptomatic bradycardia or when a gradual reduction in heart rate is indicated, and suggests increasing metoprolol dose beyond the usual maximum or an alternative beta-blocker in UMs if effectiveness remains insufficient.35

On balance, if patients known to be a CYP2D6 PM or UM are started on a beta-blocker, avoiding metoprolol seems prudent. Similarly, CYP2D6 genotyping in patients that experience an ADR on metoprolol appears justifiable to minimize risk of future ADRs from other CYP2D6-metabolized drugs that the patient might subsequently be prescribed, and to refine the metoprolol-CYP2D6 evidence base.

Key clinical positions:

  • If genotype is known metoprolol should be avoided in CYP2D6 poor and ultrarapid metabolizers, and an appropriate substitute made (e.g. bisoprolol).

  • Prescribers should be aware that co-administration of potent CYP2D6 inhibiting drugs can lead to variable phenoconversion, the extent of which will be influenced by the underlying CYP2D6 genotype.

Hypertension

Primary hypertension (HTN) is highly prevalent in Europe and a leading cause of morbidity and mortality despite the existence of many pharmacologic therapies and lifestyle modification recommendations.110,111 There are now >1000 genetic loci linked with blood pressure regulation through large GWASs and meta-analyses, which supports a complex polygenic mechanism of partial heritability, with most common risk alleles exerting a small effect on blood pressure.112,113

Interestingly, one of the largest multi-ethnic meta-analysis of blood pressure traits to-date included a phenome-wide association study which demonstrated uniform distribution of clinical risk based on identified genetic loci across ethnic groups, including Caucasians, Hispanics, and persons of African descent.113 Though PGx trials have not provided an evidence base to support genotype-guided HTN pharmacotherapy, therapeutic recommendations remain stratified by self-reported ethnicity, with ‘black’ Europeans recommended a different treatment regime (extrapolated from African American population data from the USA).110

There is some evidence from the GenHAT study (a sub-study of ALLHAT), the largest RCT to explore gene panel risk prediction in stratified anti-hypertensive (anti-HTN) therapies that genetic panels can predict response to different anti-HTN therapies. However, this adequately powered study (N > 39 000) did not demonstrate clinically significant benefit to genetically stratified therapeutic choice.114 It also failed to demonstrate different clinical outcomes for use of the studied anti-HTN therapeutics in Caucasians or participants of African descent. This trial did not include beta-blockers, which are less routinely used to treat HTN.

Thus, current evidence does not support the routine use of any genomic targeted treatment or change to therapy for any variant reported for essential HTN (beta-blockers discussed separately above). There remains a need for further prospective clinical studies to either validate or discard the current practice of using self-reported ethnicity (black or white) to guide initial therapeutic choice. Prior assessment of evidence has concluded that this may be dubious.115

Key clinical positions:

  • No clinical action advised based on current evidence

Arrhythmias

The main arrhythmia-predisposing condition related to pharmacotherapy and amenable to genetically guided intervention is long QT.116

A prolonged QT interval is a well-known risk factor for ventricular arrhythmias [including torsade de pointes (TdP), a rare and unstable polymorphic ventricular tachycardia] and sudden cardiac death (SCD), and can be congenital or acquired.117–121 Acquired QT prolongation can be caused by cardiac disease, such as coronary heart disease and HF, but it is commonly caused by certain drugs [drug-induced long QT syndrome (diLQTS)].120,122 QT interval prolongation is one of the leading causes of therapeutic relabelling or withdrawal from the market. To date, >200 drugs are listed on CredibleMeds.org (a curated database maintained by AZCERT) as associated with QT prolongation and/or TdP.123

Susceptibility to acquired QT interval prolongation can be influenced by genetic variation; the heritability of QT interval duration in the general population (excluding congenital LQTS patients) is estimated to be ∼35%, and first-degree relatives of patients with congenital LQTS have a higher risk of drug-induced QT prolongation than non-related individuals.124–127 A large number of genes associated with QT interval duration have been identified by GWAS.128–130 The gene with the strongest signal related to QT interval duration is the nitric oxide synthase 1 adaptor protein gene (NOS1AP), located on chromosome 1.128–134 This gene also influences impulse propagation.135 Other key findings from GWAS included polymorphisms within genes known to be mutated in congenital LQTS, genes associated with intracellular calcium handling, as well as genes previously not known to influence cardiac repolarization.124,129,130 A polygenic risk score comprised of 61 variants previously associated with baseline QT interval at genome-wide level was also associated with drug-induced QT prolongation, and was a significant predictor of drug-induced TdP.136,137

Several other exploratory studies have sought to identify the underlying genetic architecture predisposing to diLQTS.136,138–140 One candidate gene study evaluated the genetic predisposition to diLQTS among 176 cases and 1044 controls and identified a nonsynonymous variant in KCNE1 (rs1805128, D85N) that conferred risk of diLQTS (odds ratio = 9.0, 95% CI 3.5–22.9).138 In an exome sequencing study of 65 diLQTS cases and 148 controls, a significant excess of rare variants in KCNE1 and ACN9 were identified. Moreover, 37% of diLQTS cases were also carriers of a rare variant in potassium channel genes compared with 21% of controls.140 These studies highlight the importance of individualized treatment and risk stratification in the setting of ADRs, but also the need for further work to translate this data to the clinical realm with thorough validation studies.

The most common mechanism by which drugs lead to acquired LQTS is blockade of the rapid component of the delayed rectifier potassium current, Ikr, encoded by KCNH2, also referred to as human ether-a-go-go-related gene.141 The risk of TdP seems to increase with higher concentrations of Ikr blocking drugs. Thus, genetic variants that alter drug metabolism and result in higher drug concentrations can predispose to diLQTS. The reduced repolarization reserve hypothesis proposes that patients remain asymptomatic until multiple hits (e.g. hypokalaemia, bradycardia, and/or exposure to a drug) unmask an extreme drug response phenotype (i.e. QT prolongation) that can lead to the development of fatal arrhythmias.117

Future prospects

Potentially dangerous effects of drugs may be masked when administered to a large number of patients. The risk may become evident only when genetic and environmental risk factors combine; for example, when an Ikr blocking drug is given to an individual with a silent mutation in one of the congenital LQTS genes in the setting of hypokalaemia.

Therefore, therapeutics that are both safe and efficacious for the majority of the population may be withdrawn due to unacceptable risk in a small minority of patients. With the current emphasis on avoiding QT prolongation in drug development, it is likely that drugs that would never result in TdP in a selected population are being abandoned due to perceived risk. The challenge lies in identifying which patients, in which settings, and with which drugs or combination of drugs (and at what doses) will develop diLQTS and avoiding the combination of factors that promote TdP.

Key clinical positions:

  • Medications known to prolong the QT interval should be avoided in those known to harbour long-QT-associated genetic variants.

  • Prospective genotyping in absence of phenotype or family history is not advised.

Chemotherapy-induced cardiomyopathy

Cardiomyopathy and HF resultant from chemotherapeutic myocyte toxicity [chemotherapy-induced cardiomyopathy (CCM)] is a feared side effect of several common chemotherapeutic agents, most prominently: doxorubicin, trastuzumab, paclitaxel, and 5-fluorouracil.142 The most studied amongst these is doxorubicin, with anthracycline-induced cardiotoxicity the subject of the sole existing PGx recommendation: The Canadian Pharmacogenomics Network for Drug Safety (CPNDS) recommends genetic testing for 3 SNPs (RARG rs2229774, SLC28A3 rs7853758, UGT1A6*4 rs17863783) prior to initiating doxorubicin or daunorubicin in children, endorsed by a moderate level B evidence-based support.143 Though these variants were each replicated in separate cohorts within a single published study, they have not yet been validated by further independent research. This recommendation was met with some consternation, as these SNPs lack functional validation as well as mechanistic plausibility (two are not expressed in cardiac tissue and don’t alter protein coding), and has not been incorporated into other guidelines or mainstream practice.144 A recent study from the USA found that a polymorphism present in both African and European ancestry populations was associated with higher risk of cardiomyopathy in childhood cancer survivors, with disproportionately higher risk to those of African ancestry (5.43- vs. 1.31-fold); however, this population included exposure to radiotherapy and anthracyclines.145

While PGx studies have not yet evolved to clinical utility in risk prediction within mainstream care, recent advances have localized additional risk to those cancer patients with TTN truncating variants (TTNtvs). The massive titin protein, coded for by TTN, is integral to sarcomere function, and implicated in familial forms of DCM.146,147 These genetic changes are hypothesized to create a stress cardiomyopathy phenotype that may also be expected in states such as pregnancy or alcohol excess.146

Nevertheless, the study that linked TTNtvs to CCM identified TTNtvs in only 7.5% of those with CCM, and not all the at-risk participants developed cardiomyopathy.146 However, this study sets the stage for the possibility of improved risk stratification which could prospectively investigate risk modification strategies for a genetically at-risk population. Work is urgently needed in this area to advance scientific gains to clinical care.

Key clinical positions:

  • No clinical action advised based on current evidence.

Clinical trials

Despite the growing worldwide burden of CVD, drug development in the field has been on a downward trend due to the numerous challenges associated with successfully bringing a new drug to the market. Compared to other therapeutic areas, developing new cardiovascular medicines is more expensive for many reasons including the requirement for trials with large sample sizes, new therapies usually add only incremental benefit to individuals on very effective polypharmacy, and increased late-phase failures. Therefore, to ensure the future of cardiovascular drug development, clinical trials need to evolve and embrace new advances to overcome these challenges.148

Pharmacogenomics could help address these difficulties by facilitating mechanism-based approaches for drug development in defined populations. This approach can be utilized at various stages in the drug development pathway.

Challenges for conducting PGx RCTs for drugs that are already licensed include each trial only being able to assess one or a few gene-drug pairs, and large numbers of participants needing to be recruited to include sufficient with the variant allele(s) of interest in the absence of accessible genomic data prior to recruitment.56 Moreover, there are specific concerns over the generalizability of PGx RCT evidence to ethnicities inadequately represented in such trials, as they may carry different gene variants relevant to the studied drug. Furthermore, there is a lack of consensus over the evidential threshold required for PGx biomarkers whose role is to guide dose/drug selection akin to prescribing adjustments based on renal/liver function tests.149

Phase II and III trials

Randomized controlled trials tend to require large patient numbers to show a clinical effect. However, there has been interested in improving patient selection to identify those who are most likely to benefit from the proposed treatment, which may potentially reduce the sample size requirements.

Post hoc subgroup analyses attempt to identify PGx markers from already completed RCTs. This has clear cost saving benefits, but studies risk being under-powered and results inconclusive.

Prospective study design takes adequate powering into consideration to produce more robust evidence. Examples of this design include the following150:

  • Enrichment design, where only patients who have the feature predicted to have clinical benefit are recruited or analysed. An example of this is the TAILOR-PCI trial discussed above, where the analysis was limited to those with CYP2C19 loss of function (*2, *3) variants.56

  • Adaptive trial design is categorized by adjusting trial features based on data amassed during the trial itself. An example of this is the GENETIC-AF trial.151

  • Biomarker based all-comers design which aims to investigate the interaction between treatment effect and biomarker status, more commonly employed in oncology.

  • Hybrid or combination biomarker trials, which are typically an enrichment study extending to include another technique such as adaptive design.

Successes, failures, and opportunities—lipid modulation trials

The discovery that mutations in PCSK9 were a rare cause of familial hypercholesterolaemia paved the way for the development of PCSK9 inhibitors for lipid management. Yet, cholesteryl ester transfer protein (CETP) inhibitors have proven that identification of a PGx marker does not guarantee successful drug development, and multiple CETP inhibitors have failed phase III trials.152 However, persistence with this therapeutic target is proving fruitful. The REVEAL trial, with the CETP inhibitor Anacetrapib showed a reduction in cardiovascular events.153 Additionally, the dal-GenE trial suggests improved cardiovascular outcomes when the CETP inhibitor dalcetrapib is used with a statin in the subset of patients with the AA genotype at rs1967309 in the ADCY9 gene.154 This further demonstrates the complexities of PGx application in trials as well as the need for robust research to ensure that potentially useful therapies are not wasted.

Pharmacogenomics has the potential to enrich the development of new therapies in cardiovascular medicine, resulting in a more efficient and cost saving drug development route. Significant challenges exist and well-designed trials are required to ensure we have robust evidence.

Real-world evidence to evaluate pharmacogenomics utility

Due to limitations in feasibility for RCT trials for each drug-gene pair, real-world evidence which uses the power of big data contained within electronic health records may prove useful. This approach would also be more likely to include more diverse populations, limiting concerns about external validity of data and health equality. An example of such an approach is the IGNITE study, which assessed outcomes for use of PGx-guided antiplatelet therapy following PCI (and found increased risk of cardiovascular events in those with a CYP2C19 loss-of-function allele who were prescribed clopidogrel).155

Key position:

  • Pharmacogenomics offers tools that can revolutionize drug design and clinical trials and make both more economically efficient. However, prospective designs should be optimized and post hoc subgroup analysis limitations recognized. Real-world evidence may fill important gaps.

Key paper positions

  • Thrombosis, ischaemic heart disease, and stroke: Evidence is strongest for pre-emptive genotyping prior to warfarin use. There is also enough evidence to advise genotype-guided antiplatelet therapy in selected high-risk CV patients.

  • Hypertension: Further research is needed to translate scientific gains to the bedside. Self-identified black/white racial stratification in HTN therapeutic algorithms is not supported by genetic evidence to-date.

  • Arrhythmias: Further research is needed to advance our understanding of genetic risk loci for drug-induced long QT and to trial a PGx approach in clinical practice. Known carriers of pathogenic LQT variants should not be given medication known to prolong the QTC.

  • Chemotherapy-induced cardiomyopathy: Further research is needed to translate scientific gains to the bedside.

  • Clinical trials: PGx has the potential to improve the success, efficacy, and cost profile of drug development and clinical trials but is in early stages of development.

Summary graphic of ESC WG clinical PGx advice. This flow graphic illustrates the modes in which patients may have genetic testing, either prior to clinical encounter or indicated during a clinical encounter (where genotyping may be indicated prior to warfarin initiation or to guide antiplatelet therapy in high-risk patients). This information from diverse sources may then impact on the prescribing of metoprolol, simvastatin, clopidogrel, and warfarin, to increase therapeutic efficacy and decrease the probability of adverse drug reactions. ADR, adverse drug reactions; DTC, direct-to-consumer.
Figure 1

Summary graphic of ESC WG clinical PGx advice. This flow graphic illustrates the modes in which patients may have genetic testing, either prior to clinical encounter or indicated during a clinical encounter (where genotyping may be indicated prior to warfarin initiation or to guide antiplatelet therapy in high-risk patients). This information from diverse sources may then impact on the prescribing of metoprolol, simvastatin, clopidogrel, and warfarin, to increase therapeutic efficacy and decrease the probability of adverse drug reactions. ADR, adverse drug reactions; DTC, direct-to-consumer.

Areas with promise for PGx translation—key opportunities for research. Key opportunities for research to advance pharmacogenomics and facilitate translation to the bedside.
Figure 2

Areas with promise for PGx translation—key opportunities for research. Key opportunities for research to advance pharmacogenomics and facilitate translation to the bedside.

Acknowledgements

M.J.C. is an NIHR Senior Investigator and this work forms part of the research portfolio of the NIHR Biomedical Research Centre at Barts. M.P. is an Emeritus NIHR Senior Investigator, and wishes to thank the MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine for infrastructure support.

Conflict of interest: M.J.C. is seconded to Genomics England as a Chief Scientist and part of his salary is funded by Genomics England Ltd, a wholly owned Department of Health and Social Care Company. M.P. receives research funding from various organizations including the MRC, NIHR, EU Commission, HDR UK, and Health Education England. He has also received partnership funding for the following: MRC Clinical Pharmacology Training Scheme (co-funded by MRC and Roche, UCB, Eli Lilly, and Novartis); a PhD studentship jointly funded by EPSRC and Astra Zeneca; and grant funding from Vistagen Therapeutics. He has also unrestricted educational grant support for the UK Pharmacogenetics and Stratified Medicine Network from Bristol-Myers Squibb and UCB. He has developed an HLA genotyping panel with MC Diagnostics, but does not benefit financially from this. MP is also a member of the IMI Consortium ARDAT (www.ardat.org). None of the funding declared above has been directly used for the current review paper. S.A. receives Speaker fees and advisory board fees from: Boehringer-Ingelheim, Daiichi Sankyo, Bayer, Thermofisher. A.G.S. has received speaker honoraria and/or consulting fees from Sanofi, AbbVie, Novartis, Bayer, and Lilly. G.S. reports grants and personal fees from Vifor, grants and non-financial support from Boehringer Ingelheim, personal fees from Societa’ Prodotti Antibiotici, grants and personal fees from AstraZeneca, personal fees from Roche and Servier, grants from Novartis, personal fees from GENESIS, Cytokinetics, and Medtronic, grants from Boston Scientific, outside the submitted work. P.S. reports grants from Daiichi Sankyo, grants and personal fees from Boehringer-Ingelheim outside the submitted work.

References

1

Initial sequencing and analysis of the human genome
.
Nature
 
2001
;
409
:
860
921
.

2

Green
 
ED
,
Guyer
 
MS
;
National Human Genome Research Institute
.
Charting a course for genomic medicine from base pairs to bedside
.
Nature
 
2011
;
470
:
204
213
.

3

National Human Genome Research Institute
. DNA Sequencing Costs: Data.
2019
. https://www.genome.gov/about-genomics/fact-sheets/DNA-Sequencing-Costs-Data (10 August 2020).

4

Schwarze
 
K
,
Buchanan
 
J
,
Taylor
 
JC
,
Wordsworth
 
S.
 
Are whole-exome and whole-genome sequencing approaches cost-effective? A systematic review of the literature
.
Genet Med
 
2018
;
20
:
1122
1130
.

5

NIH National Human Genome Research Institute
; Human Genome Project. https://www.genome.gov/human-genome-project/Completion-FAQ (20 October
2020
).

6

Relling
 
MV
,
Evans
 
WE.
 
Pharmacogenomics in the clinic
.
Nature
 
2015
;
526
:
343
350
.

7

Pirmohamed
 
M
,
Hughes
 
DA.
 
Pharmacogenetic tests: the need for a level playing field
.
Nat Rev Drug Discov
 
2013
;
12
:
3
4
.

8

Ch van der
 
W
,
Swen
 
JJ
,
Samwald
 
M
,
Mitropoulou
 
C
,
Schwab
 
M
,
Guchelaar
 
H-J
; on behalf of the Ubiquitous-Pharmacogenomics Consortium.
A brighter future for the implementation of pharmacogenomic testing
.
Eur J Hum Genet
 
2016
;
24
:
1658
1660
.

9

Shekhani
 
R
,
Steinacher
 
L
,
Swen
 
JJ
,
Ingelman‐Sundberg
 
M.
 
Evaluation of current regulation and guidelines of pharmacogenomic drug labels: opportunities for improvements
.
Clin Pharmacol Ther
 
2020
;
107
:
1240
1255
.

10

Howard
 
HC
,
Borry
 
P.
 
Direct-to-consumer pharmacogenomic testing
.
Pharmacogenomics
 
2011
;
12
:
1367
1370
.

11

Pengo
 
V
,
Pegoraro
 
C
,
Cucchini
 
U
,
Iliceto
 
S.
 
Worldwide management of oral anticoagulant therapy: the ISAM study
.
J Thromb Thrombolysis
 
2006
;
21
:
73
77
.

12

Lutsey
 
PL
,
Walker
 
RF
,
MacLehose
 
RF
,
Alonso
 
A
,
Adam
 
TJ
,
Zakai
 
NA.
 
Direct oral anticoagulants and warfarin for venous thromboembolism treatment: trends from 2012 to 2017
.
Res Pract Thromb Haemost
 
2019
;
3
:
668
673
.

13

Verhoef
 
TI
,
Redekop
 
WK
,
Daly
 
AK
,
Van Schie
 
RMF
,
De Boer
 
A
,
Maitland-Van Der Zee
 
A-H.
 
Pharmacogenetic-Guided. dosing of coumarin anticoagulants: algorithms for warfarin, acenocoumarol and phenprocoumon
.
Br J Clin Pharmacol
 
2014
;
77
:
626
641
.

14

Reilly
 
RAO.
 
Studies on the optical enantiomorphs of warfarin in man
.
Clin Pharmacol Ther
 
1974
;
16
:
348
354
.

15

Wadelius
 
M
,
Sörlin
 
K
,
Wallerman
 
O
,
Karlsson
 
J
,
Yue
 
QY
,
Magnusson
 
PKE
,
Wadelius
 
C
,
Melhus
 
H.
 
Warfarin sensitivity related to CYP2C9, CYP3A5, ABCB1 (MDR1) and other factors
.
Pharmacogenomics J
 
2004
;
4
:
40
48
.

16

Kaminsky
 
LS
,
Zhang
 
ZY.
 
Human P450 metabolism of warfarin
.
Pharmacol Ther
 
1997
;
73
:
67
74
.

17

Ufer
 
M
,
Svensson
 
JO
,
Krausz
 
KW
,
Gelboin
 
HV
,
Rane
 
A
,
Tybring
 
G.
 
Identification of cytochromes P450 2C9 and 3A4 as the major catalysts of phenprocoumon hydroxylation in vitro
.
Eur J Clin Pharmacol
 
2004
;
60
:
173
182
.

18

Pokorney
 
SD
,
Simon
 
DN
,
Thomas
 
L
,
Fonarow
 
GC
,
Kowey
 
PR
,
Chang
 
P
,
Singer
 
DE
,
Ansell
 
J
,
Blanco
 
RG
,
Gersh
 
B
,
Mahaffey
 
KW
,
Hylek
 
EM
,
Go
 
AS
,
Piccini
 
JP
,
Peterson
 
ED.
 
Patients’ time in therapeutic range on warfarin among US patients with atrial fibrillation: results from ORBIT-AF registry
.
Am Heart J
 
2015
;
170
:
141
148.e1
.

19

Lau
 
WCY
,
Li
 
X
,
Wong
 
ICK
,
Man
 
KKC
,
Lip
 
GYH
,
Leung
 
WK
,
Siu
 
CW
,
Chan
 
EW.
 
Bleeding-related hospital admissions and 30-day readmissions in patients with non-valvular atrial fibrillation treated with dabigatran versus warfarin
.
J Thromb Haemost
 
2017
;
15
:
1923
1933
.

20

Hylek
 
EM
,
Evans-Molina
 
C
,
Shea
 
C
,
Henault
 
LE
,
Regan
 
S.
 
Major hemorrhage and tolerability of warfarin in the first year of therapy among elderly patients with atrial fibrillation
.
Circulation
 
2007
;
115
:
2689
2696
.

21

Palareti
 
G
,
Cosmi
 
B.
 
Bleeding with anticoagulation therapy—who is at risk, and how best to identify such patients
.
Thromb Haemost
 
2009
;
102
:
268
278
.

22

Jones
 
M
,
McEwan
 
P
,
Morgan
 
CL
,
Peters
 
JR
,
Goodfellow
 
J
,
Currie
 
CJ.
 
Evaluation of the pattern of treatment, level of anticoagulation control, and outcome of treatment with warfarin in patients with non-valvar atrial fibrillation: a record linkage study in a large British population
.
Heart
 
2005
;
91
:
472
477
.

23

Cancino
 
RS
,
Hylek
 
EM
,
Reisman
 
JI
,
Rose
 
AJ.
 
Comparing patient-level and site-level anticoagulation control as predictors of adverse events
.
Thromb Res
 
2014
;
133
:
652
656
.

24

Bourgeois
 
S
,
Jorgensen
 
A
,
Zhang
 
EJ
,
Hanson
 
A
,
Gillman
 
MS
,
Bumpstead
 
S
,
Toh
 
CH
,
Williamson
 
P
,
Daly
 
AK
,
Kamali
 
F
,
Deloukas
 
P
,
Pirmohamed
 
M.
 
A multi-factorial analysis of response to warfarin in a UK prospective cohort
.
Genome Med
 
2016
;
8
:
2
–14.

25

Pirmohamed
 
M
,
Kamali
 
F
,
Daly
 
AK
,
Wadelius
 
M.
 
Oral anticoagulation: a critique of recent advances and controversies
.
Trends Pharmacol Sci
 
2015
;
36
:
153
163
.

26

Borgiani
 
P
,
Ciccacci
 
C
,
Forte
 
V
,
Siríanni
 
E
,
Novelli
 
L
,
Bramanti
 
P
,
Novelli
 
G.
 
CYP4F2 genetic variant (rs2108622) significantly contributes to warfarin dosing variability in the Italian population
.
Pharmacogenomics
 
2009
;
10
:
261
266
.

27

Gage
 
BF
,
Bass
 
AR
,
Lin
 
H
,
Woller
 
SC
,
Stevens
 
SM
,
Al-Hammadi
 
N
,
Li
 
J
,
Rodríguez
 
T
,
Miller
 
JP
,
McMillin
 
GA
,
Pendleton
 
RC
,
Jaffer
 
AK
,
King
 
CR
,
Whipple
 
BDV
,
Porche-Sorbet
 
R
,
Napoli
 
L
,
Merritt
 
K
,
Thompson
 
AM
,
Hyun
 
G
,
Anderson
 
JL
,
Hollomon
 
W
,
Barrack
 
RL
,
Nunley
 
RM
,
Moskowitz
 
G
,
Dávila-Román
 
V
,
Eby
 
CS.
 
Effect of genotype-guided warfarin dosing on clinical events and anticoagulation control among patients undergoing hip or knee arthroplasty: the GIFT randomized clinical trial
.
JAMA
 
2017
;
318
:
1115
1124
.

28

Xu
 
Z
,
Zhang
 
S
,
Yu Huang
 
M
,
Hu
 
R
,
Li
 
J
,
Li Cen
 
H
,
Jin Wang
 
Z
,
Ping Ou
 
J
,
Song Yin
 
S
,
Li Xu
 
Y
,
Qi Wu
 
Z
,
Kai Zhang
 
X.
 
Genotype-Guided Warfarin Dosing in Patients With Mechanical Valves: a Randomized Controlled Trial
.
Ann Thorac Surg
 
2018
;
106
:
1774
1781
.

29

Pirmohamed
 
M
,
Burnside
 
G
,
Eriksson
 
N
,
Jorgensen
 
AL
,
Toh
 
CH
,
Nicholson
 
T
,
Kesteven
 
P
,
Christersson
 
C
,
Wahlström
 
B
,
Stafberg
 
C
,
Zhang
 
JE
,
Leathart
 
JB
,
Kohnke
 
H
,
Maitland-van der Zee
 
AH
,
Williamson
 
PR
,
Daly
 
AK
,
Avery
 
P
,
Kamali
 
F
,
Wadelius
 
M.
 
A randomized trial of genotype-guided dosing of warfarin
.
N Engl J Med
 
2013
;
369
:
2294
2303
.

30

Jorgensen
 
AL
,
Prince
 
C
,
Fitzgerald
 
G
,
Hanson
 
A
,
Downing
 
J
,
Reynolds
 
J
,
Zhang
 
JE
,
Alfirevic
 
A
,
Pirmohamed
 
M.
 
Implementation of genotype-guided dosing of warfarin with point-of-care genetic testing in three UK clinics: a matched cohort study
.
BMC Med
 
2019
;
17
:
76
–87.

31

Ng
 
SS
,
Lai
 
NM
,
Nathisuwan
 
S
,
Jahan
 
NK
,
Dilokthornsakul
 
P
,
Kongpakwattana
 
K
,
Hollingworth
 
W
,
Chaiyakunapruk
 
N.
 
Comparative efficacy and safety of warfarin care bundles and novel oral anticoagulants in patients with atrial fibrillation: a systematic review and network meta-analysis
.
Sci Rep
 
2020
;
10
:
1
11
.

32

Kimmel
 
SE
,
French
 
B
,
Kasner
 
SE
,
Johnson
 
JA
,
Anderson
 
JL
,
Gage
 
BF
,
Rosenberg
 
YD
,
Eby
 
CS
,
Madigan
 
RA
,
McBane
 
RB
,
Abdel-Rahman
 
SZ
,
Stevens
 
SM
,
Yale
 
S
,
Mohler
 
ER
,
Fang
 
MC
,
Shah
 
V
,
Horenstein
 
RB
,
Limdi
 
NA
,
Muldowney
 
JAS
,
Gujral
 
J
,
Delafontaine
 
P
,
Desnick
 
RJ
,
Ortel
 
TL
,
Billett
 
HH
,
Pendleton
 
RC
,
Geller
 
NL
,
Halperin
 
JL
,
Goldhaber
 
SZ
,
Caldwell
 
MD
,
Califf
 
RM
,
Ellenberg
 
JH.
 
A pharmacogenetic versus a clinical algorithm for warfarin dosing
.
N Engl J Med
 
2013
;
369
:
2283
2293
.

33

Zhu
 
Y
,
Swanson
 
KM
,
Rojas
 
RL
,
Wang
 
Z
,
Sauver
 
JL
,
St Visscher
 
SL
,
Prokop
 
LJ
,
Bielinski
 
SJ
,
Wang
 
L
,
Weinshilboum
 
R
,
Borah
 
BJ.
 
Systematic review of the evidence on the cost-effectiveness of pharmacogenomics-guided treatment for cardiovascular diseases
.
Genet Med
 
2020
;
22
:
475
486
.

34

Dong
 
OM
,
Wheeler
 
SB
,
Cruden
 
G
,
Lee
 
CR
,
Voora
 
D
,
Dusetzina
 
SB
,
Wiltshire
 
T.
 
Cost-effectiveness of multigene pharmacogenetic testing in patients with acute coronary syndrome after percutaneous coronary intervention
.
Value Heal
 
2020
;
23
:
61
73
.

35

Dutch Pharmacogenetics Working Group (DPWG)
. Pharmacogenetics recommendations. Eur J Hum Genet 2020;
28
:
508
517
.

36

Johnson
 
JA
,
Caudle
 
KE
,
Gong
 
L
,
Whirl-Carrillo
 
M
,
Stein
 
CM
,
Scott
 
SA
,
Lee
 
MT
,
Gage
 
BF
,
Kimmel
 
SE
,
Perera
 
MA
,
Anderson
 
JL
,
Pirmohamed
 
M
,
Klein
 
TE
,
Limdi
 
NA
,
Cavallari
 
LH
,
Wadelius
 
M.
 
Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for pharmacogenetics-guided warfarin dosing: 2017 update
.
Clin Pharmacol Ther
 
2017
;
102
:
397
404
.

37

Verhoef
 
TI
,
Ragia
 
G
,
Boer
 
A
,
de Barallon
 
R
,
Kolovou
 
G
,
Kolovou
 
V
,
Konstantinides
 
S
,
Cessie
 
S
,
Le Maltezos
 
E
,
Meer
 
FJM
,
van der Redekop
 
WK
,
Remkes
 
M
,
Rosendaal
 
FR
,
Schie
 
RMF
,
van Tavridou
 
A
,
Tziakas
 
D
,
Wadelius
 
M
,
Manolopoulos
 
VG
,
Maitland-van der Zee
 
AH.
 
A randomized trial of genotype-guided dosing of acenocoumarol and phenprocoumon
.
N Engl J Med
 
2013
;
369
:
2304
2312
.

38

Ragia
 
G
,
Kolovou
 
V
,
Kolovou
 
G
,
Konstantinides
 
S
,
Maltezos
 
E
,
Tavridou
 
A
,
Tziakas Di Maitland-Van Der Zee
 
AH
,
Manolopoulos
 
VG.
 
A novel acenocoumarol pharmacogenomic dosing algorithm for the Greek population of EU-PACT trial
.
Pharmacogenomics
 
2017
;
18
:
23
34
.

39

Ragia
 
G
,
Manolopoulos
 
VG.
 
Pharmacogenomics of anticoagulation therapy: the last 10 years
.
Pharmacogenomics
 
2019
;
20
:
1113
1117
.

40

Hernandez
 
W
,
Danahey
 
K
,
Pei
 
X
,
Yeo
 
KTJ
,
Leung
 
E
,
Volchenboum
 
SL
,
Ratain
 
MJ
,
Meltzer
 
DO
,
Stranger
 
BE
,
Perera
 
MA
,
O’Donnell
 
PH.
 
Pharmacogenomic genotypes define genetic ancestry in patients and enable population-specific genomic implementation
.
Pharmacogenomics
 
2020
;
20
:
126
135
.

41

Yuan
 
HY
,
Chen
 
JJ
,
Lee
 
MTM
,
Wung
 
JC
,
Chen
 
YF
,
Charng
 
MJ
,
Lu
 
MJ
,
Hung
 
CR
,
Wei
 
CY
,
Chen
 
CH
,
Wu
 
JY
,
Chen
 
YT.
 
A novel functional VKORC1 promoter polymorphism is associated with inter-individual and inter-ethnic differences in warfarin sensitivity
.
Hum Mol Genet Hum Mol Genet
 
2005
;
14
:
1745
1751
.

42

Asiimwe
 
IG
,
Zhang
 
EJ
,
Osanlou
 
R
,
Krause
 
A
,
Dillon
 
C
,
Suarez‐Kurtz
 
G
,
Zhang
 
H
,
Perini
 
JA
,
Renta
 
JY
,
Duconge
 
J
,
Cavallari
 
LH
,
Marcatto
 
LR
,
Beasly
 
MT
,
Perera
 
MA
,
Limdi
 
NA
,
Santos
 
PCJL
,
Kimmel
 
SE
,
Lubitz
 
SA
,
Scott
 
SA
,
Kawai
 
VK
,
Jorgensen
 
AL
,
Pirmohamed
 
M.
 
Genetic factors influencing warfarin dose in Black‐African patients: a systematic review and meta‐analysis
.
Clin Pharmacol Ther
 
2020
;
107
:
1420
1433
.

43

Cavallari
 
LH
,
Langaee
 
TY
,
Momary
 
KM
,
Shapiro
 
NL
,
Nutescu
 
EA
,
Coty
 
WA
,
Viana
 
MAG
,
Patel
 
SR
,
Johnson
 
JA.
 
Genetic and clinical predictors of warfarin dose requirements in African Americans
.
Clin Pharmacol Ther
 
2010
;
87
:
459
464
.

44

McDonald
 
MG
,
Rieder
 
MJ
,
Nakano
 
M
,
Hsia
 
CK
,
Rettie
 
AE.
 
CYP4F2 is a vitamin K1 oxidase: an explanation for altered warfarin dose in carriers of the V433M variant
.
Mol Pharmacol
 
2009
;
75
:
1337
1346
.

45

Ibanez
 
B
,
James
 
S
,
Agewall
 
S
,
Antunes
 
MJ
,
Bucciarelli-Ducci
 
C
,
Bueno
 
H
,
Caforio
 
ALP
,
Crea
 
F
,
Goudevenos
 
JA
,
Halvorsen
 
S
,
Hindricks
 
G
,
Kastrati
 
A
,
Lenzen
 
MJ
,
Prescott
 
E
,
Roffi
 
M
,
Valgimigli
 
M
,
Varenhorst
 
C
,
Vranckx
 
P
,
Widimský
 
P
,
Collet
 
J-P
,
Kristensen
 
SD
,
Aboyans
 
V
,
Baumbach
 
A
,
Bugiardini
 
R
,
Coman
 
IM
,
Delgado
 
V
,
Fitzsimons
 
D
,
Gaemperli
 
O
,
Gershlick
 
AH
,
Gielen
 
S
;
ESC Scientific Document Group
.
2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation
.
Eur Heart J
 
2018
;
39
:
119
177
.

46

Roffi
 
M
,
Patrono
 
C
,
Collet
 
J-P
,
Mueller
 
C
,
Valgimigli
 
M
,
Andreotti
 
F
,
Bax
 
JJ
,
Borger
 
MA
,
Brotons
 
C
,
Chew
 
DP
,
Gencer
 
B
,
Hasenfuss
 
G
,
Kjeldsen
 
K
,
Lancellotti
 
P
,
Landmesser
 
U
,
Mehilli
 
J
,
Mukherjee
 
D
,
Storey
 
RF
,
Windecker
 
S
;
ESC Scientific Document Group
.
2015 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation
.
Eur Heart J
 
2016
;
37
:
267
315
.

47

Wallentin
 
L
,
Becker
 
RC
,
Budaj
 
A
,
Cannon
 
CP
,
Emanuelsson
 
H
,
Held
 
C
,
Horrow
 
J
,
Husted
 
S
,
James
 
S
,
Katus
 
H
,
Mahaffey
 
KW
,
Scirica
 
BM
,
Skene
 
A
,
Steg
 
PG
,
Storey
 
RF
,
Harrington
 
RA.
 
Ticagrelor versus clopidogrel in patients with acute coronary syndromes
.
N Engl J Med
 
2009
;
361
:
1045
1057
.

48

Powers
 
WJ
,
Rabinstein
 
AA
,
Ackerson
 
T
,
Adeoye
 
OM
,
Bambakidis
 
NC
,
Becker
 
K
,
Biller
 
J
,
Brown
 
M
,
Demaerschalk
 
BM
,
Hoh
 
B
,
Jauch
 
EC
,
Kidwell
 
CS
,
Leslie-Mazwi
 
TM
,
Ovbiagele
 
B
,
Scott
 
PA
,
Sheth
 
KN
,
Southerland
 
AM
,
Summers
 
DV
,
Tirschwell
 
DL.
 
Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association
.
Stroke 2018
 
2018
;
49
:
e46
e99
.

49

Scott
 
SA
,
Sangkuhl
 
K
,
Gardner
 
EE
,
Stein
 
CM
,
Hulot
 
J-S
,
Johnson
 
JA
,
Roden
 
DM
,
Klein
 
TE
,
Shuldiner
 
AR
;
Clinical Pharmacogenetics Implementation Consortium
.
Clinical pharmacogenetics implementation consortium guidelines for cytochrome P450-2C19 (CYP2C19) genotype and clopidogrel therapy
.
Clin Pharmacol Ther
 
2011
;
90
:
328
332
.

50

Shuldiner
 
AR.
 
Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy
.
JAMA
 
2009
;
302
:
849
.

51

Sofi
 
F
,
Giusti
 
B
,
Marcucci
 
R
,
Gori
 
AM
,
Abbate
 
R
,
Gensini
 
GF.
 
Cytochrome P450 2C192 polymorphism and cardiovascular recurrences in patients taking clopidogrel: a meta-analysis
.
Pharmacogenomics J
 
2011
;
11
:
199
206
.

52

Lewis
 
JP
,
Backman
 
JD
,
Reny
 
J-L
,
Bergmeijer
 
TO
,
Mitchell
 
BD
,
Ritchie
 
MD
,
Déry
 
J-P
,
Pakyz
 
RE
,
Gong
 
L
,
Ryan
 
K
,
Kim
 
E-Y
,
Aradi
 
D
,
Fernandez-Cadenas
 
I
,
Lee
 
MTM
,
Whaley
 
RM
,
Montaner
 
J
,
Gensini
 
GF
,
Cleator
 
JH
,
Chang
 
K
,
Holmvang
 
L
,
Hochholzer
 
W
,
Roden
 
DM
,
Winter
 
S
,
Altman
 
RB
,
Alexopoulos
 
D
,
Kim
 
H-S
,
Gawaz
 
M
,
Bliden
 
KP
,
Valgimigli
 
M
,
Marcucci
 
R
,
Campo
 
G
,
Schaeffeler
 
E
,
Dridi
 
NP
,
Wen
 
M-S
,
Shin
 
JG
,
Fontana
 
P
,
Giusti
 
B
,
Geisler
 
T
,
Kubo
 
M
,
Trenk
 
D
,
Siller-Matula
 
JM
,
ten Berg
 
JM
,
Gurbel
 
PA
,
Schwab
 
M
,
Klein
 
TE
,
Shuldiner
 
AR
;
for the ICPC Investigators
.
Pharmacogenomic polygenic response score predicts ischaemic events and cardiovascular mortality in clopidogrel-treated patients
.
Eur Heart J Cardiovasc Pharmacother
 
2020
;
6
:
203
210
.

53

Kheiri
 
B
,
Osman
 
M
,
Abdalla
 
A
,
Haykal
 
T
,
Pandrangi
 
PV
,
Chahine
 
A
,
Ahmed
 
S
,
Osman
 
K
,
Bachuwa
 
G
,
Hassan
 
M
,
Bhatt
 
DL.
 
CYP2C19 pharmacogenetics versus standard of care dosing for selecting antiplatelet therapy in patients with coronary artery disease: a meta-analysis of randomized clinical trials
.
Catheter Cardiovasc Interv
 
2019
;
93
:
1246
1252
.

54

Kheiri
 
B
,
Simpson
 
TF
,
Osman
 
M
,
Kumar
 
K
,
Przybylowicz
 
R
,
Merrill
 
M
,
Golwala
 
H
,
Rahmouni
 
H
,
Cigarroa
 
JE
,
Zahr
 
F.
 
Genotype-guided strategy for P2Y12 inhibitors in coronary artery disease
.
JACC Cardiovasc Interv
 
2020
;
13
:
659
661
.

55

Claassens
 
DMF
,
Vos
 
GJA
,
Bergmeijer
 
TO
,
Hermanides
 
RS
,
’t Hof
 
AWJ
,
van Harst
 
P
,
van der Barbato
 
E
,
Morisco
 
C
,
Tjon Joe Gin
 
RM
,
Asselbergs
 
FW
,
Mosterd
 
A
,
Herrman
 
J-PR
,
Dewilde
 
WJM
,
Janssen
 
PWA
,
Kelder
 
JC
,
Postma
 
MJ
,
Boer
 
A
,
de Boersma
 
C
,
Deneer
 
VHM
,
ten Berg
 
JM.
 
A genotype-guided strategy for oral P2Y 12 inhibitors in primary PCI
.
N Engl J Med
 
2019
;
381
:
1621
1631
.

56

Pereira
 
NL
,
Farkouh
 
ME
,
So
 
D
,
Lennon
 
R
,
Geller
 
N
,
Mathew
 
V
,
Bell
 
M
,
Bae
 
J-H
,
Jeong
 
MH
,
Chavez
 
I
,
Gordon
 
P
,
Abbott
 
JD
,
Cagin
 
C
,
Baudhuin
 
L
,
Fu
 
Y-P
,
Goodman
 
SG
,
Hasan
 
A
,
Iturriaga
 
E
,
Lerman
 
A
,
Sidhu
 
M
,
Tanguay
 
J-F
,
Wang
 
L
,
Weinshilboum
 
R
,
Welsh
 
R
,
Rosenberg
 
Y
,
Bailey
 
K
,
Rihal
 
C.
 
Effect of genotype-guided oral P2Y12 inhibitor selection vs conventional clopidogrel therapy on ischemic outcomes after percutaneous coronary intervention
.
JAMA
 
2020
;
324
:
761
771
.

57

Byrne
 
RA
,
Joner
 
M
,
Kastrati
 
A.
 
Stent thrombosis and restenosis: what have we learned and where are we going? The Andreas Grüntzig Lecture ESC 2014
.
Eur Heart J
 
2015
;
36
:
3320
3331
.

58

Pan
 
Y
,
Chen
 
W
,
Xu
 
Y
,
Yi
 
X
,
Han
 
Y
,
Yang
 
Q
,
Li
 
X
,
Huang
 
L
,
Johnston
 
SC
,
Zhao
 
X
,
Liu
 
L
,
Zhang
 
Q
,
Wang
 
G
,
Wang
 
Y
,
Wang
 
Y.
 
Genetic polymorphisms and clopidogrel efficacy for acute ischemic stroke or transient ischemic attack
.
Circulation
 
2017
;
135
:
21
33
.

59

Simon
 
T
,
Danchin
 
N.
 
Clinical impact of pharmacogenomics of clopidogrel in stroke
.
Circulation
 
2017
;
135
:
34
37
.

60

Scott
 
SA
,
Sangkuhl
 
K
,
Stein
 
CM
,
Hulot
 
J-S
,
Mega
 
JL
,
Roden
 
DM
,
Klein
 
TE
,
Sabatine
 
MS
,
Johnson
 
JA
,
Shuldiner
 
AR
;
Clinical Pharmacogenetics Implementation Consortium
.
Clinical pharmacogenetics implementation consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update
. Clin Pharmacol Ther
 
2013
;
94
:
317
323
.

61

Catapano
 
AL
,
Graham
 
I
,
De Backer
 
G
,
Wiklund
 
O
,
Chapman
 
MJ
,
Drexel
 
H
,
Hoes
 
AW
,
Jennings
 
CS
,
Landmesser
 
U
,
Pedersen
 
TR
,
Reiner
 
Ž
,
Riccardi
 
G
,
Taskinen
 
M-R
,
Tokgozoglu
 
L
,
Verschuren
 
WMM
,
Vlachopoulos
 
C
,
Wood
 
DA
,
Zamorano
 
JL
,
Cooney
 
M-T
;
ESC Scientific Document Group
.
2016 ESC/EAS guidelines for the management of dyslipidaemias
.
Eur Heart J
 
2016
;
37
:
2999
3058
.

62

Audi
 
S
,
Burrage
 
DR
,
Lonsdale
 
DO
,
Pontefract
 
S
,
Coleman
 
JJ
,
Hitchings
 
AW
,
Baker
 
EH.
 
The ‘top 100’ drugs and classes in England: an updated ‘starter formulary’ for trainee prescribers
.
Br J Clin Pharmacol
 
2018
;
84
:
2562
2571
.

63

The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials
.
Lancet
 
2012
;
380
:
581
590
.

64

Collins
 
R
,
Reith
 
C
,
Emberson
 
J
,
Armitage
 
J
,
Baigent
 
C
,
Blackwell
 
L
,
Blumenthal
 
R
,
Danesh
 
J
,
Smith
 
GD
,
DeMets
 
D
,
Evans
 
S
,
Law
 
M
,
MacMahon
 
S
,
Martin
 
S
,
Neal
 
B
,
Poulter
 
N
,
Preiss
 
D
,
Ridker
 
P
,
Roberts
 
I
,
Rodgers
 
A
,
Sandercock
 
P
,
Schulz
 
K
,
Sever
 
P
,
Simes
 
J
,
Smeeth
 
L
,
Wald
 
N
,
Yusuf
 
S
,
Peto
 
R.
 
Interpretation of the evidence for the efficacy and safety of statin therapy
.
Lancet
 
2016
;
388
:
2532
2561
.

65

Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90 056 participants in 14 randomised trials of statins
.
Lancet
 
2005
;
366
:
1267
1278
.

66

Newman
 
CB
,
Preiss
 
D
,
Tobert
 
JA
,
Jacobson
 
TA
,
Page
 
RL
,
Goldstein
 
LB
,
Chin
 
C
,
Tannock
 
LR
,
Miller
 
M
,
Raghuveer
 
G
,
Duell
 
PB
,
Brinton
 
EA
,
Pollak
 
A
,
Braun
 
LT
,
Welty
 
FK.
 
Statin safety and associated adverse events: a scientific statement from the American Heart Association
.
Arterioscler Thromb Vasc Biol
 
2019
;
39
.

67

Rasmussen
 
JN
,
Chong
 
A
,
Alter
 
DA.
 
Relationship between adherence to evidence-based pharmacotherapy and long-term mortality after acute myocardial infarction
.
JAMA
 
2007
;
297
:
177
.

68

Cohen
 
JD
,
Brinton
 
EA
,
Ito
 
MK
,
Jacobson
 
TA.
 
Understanding Statin Use in America and Gaps in Patient Education (USAGE): an internet-based survey of 10,138 current and former statin users
.
J Clin Lipidol
 
2012
;
6
:
208
215
.

69

Alfirevic
 
A
,
Neely
 
D
,
Armitage
 
J
,
Chinoy
 
H
,
Cooper
 
RG
,
Laaksonen
 
R
,
Carr
 
DF
,
Bloch
 
KM
,
Fahy
 
J
,
Hanson
 
A
,
Yue
 
Q-Y
,
Wadelius
 
M
,
Maitland-van Der Zee
 
AH
,
Voora
 
D
,
Psaty
 
BM
,
Palmer
 
CNA
,
Pirmohamed
 
M.
 
Phenotype standardization for statin-induced myotoxicity
.
Clin Pharmacol Ther
 
2014
;
96
:
470
476
.

70

Hopewell
 
JC
,
Offer
 
A
,
Haynes
 
R
,
Bowman
 
L
,
Li
 
J
,
Chen
 
F
,
Bulbulia
 
R
,
Lathrop
 
M
,
Baigent
 
C
,
Landray
 
MJ
,
Collins
 
R
,
Armitage
 
J
,
Parish
 
S.
 
Independent risk factors for simvastatin-related myopathy and relevance to different types of muscle symptom
.
Eur Heart J
 
2020
;
41
:
3336
3342
.

71

Turner
 
RM
,
Pirmohamed
 
M.
 
Statin-related myotoxicity: a comprehensive review of pharmacokinetic, pharmacogenomic and muscle components
.
J Clin Med
 
2019
;
9
:
22
.

72

SLCO1B1 variants and statin-induced myopathy—a genomewide study
.
N Engl J Med
 
2008
;
359
:
789
799
.

73

Carr
 
DF
,
Francis
 
B
,
Jorgensen
 
AL
,
Zhang
 
E
,
Chinoy
 
H
,
Heckbert
 
SR
,
Bis
 
JC
,
Brody
 
JA
,
Floyd
 
JS
,
Psaty
 
BM
,
Molokhia
 
M
,
Lapeyre‐Mestre
 
M
,
Conforti
 
A
,
Alfirevic
 
A
,
Staa
 
T
,
Pirmohamed
 
M.
 
Genomewide association study of statin‐induced myopathy in patients recruited using the <scp>UK</scp> clinical practice research datalink
.
Clin Pharmacol Ther
 
2019
;
106
:
1353
1361
.

74

Generaux
 
GT
,
Bonomo
 
FM
,
Johnson
 
M
,
Mahar Doan
 
KM.
 
Impact of SLCO1B1 (OATP1B1) and ABCG2 (BCRP) genetic polymorphisms and inhibition on LDL-C lowering and myopathy of statins
.
Xenobiotica
 
2011
;
41
:
639
651
.

75

Skottheim
 
IB
,
Gedde-Dahl
 
A
,
Hejazifar
 
S
,
Hoel
 
K
,
Åsberg
 
A.
 
Statin induced myotoxicity: the lactone forms are more potent than the acid forms in human skeletal muscle cells in vitro
.
Eur J Pharm Sci
 
2008
;
33
:
317
325
.

76

Danik
 
JS
,
Chasman
 
DI
,
MacFadyen
 
JG
,
Nyberg
 
F
,
Barratt
 
BJ
,
Ridker
 
PM.
 
Lack of association between SLCO1B1 polymorphisms and clinical myalgia following rosuvastatin therapy
.
Am Heart J
 
2013
;
165
:
1008
1014
.

77

Liu
 
J-E
,
Liu
 
X-Y
,
Chen
 
S
,
Zhang
 
Y
,
Cai
 
L-Y
,
Yang
 
M
,
Lai
 
W-H
,
Ren
 
B
,
Zhong
 
S-L.
 
SLCO1B1 521T > C polymorphism associated with rosuvastatin-induced myotoxicity in Chinese coronary artery disease patients: a nested case–control study
.
Eur J Clin Pharmacol
 
2017
;
73
:
1409
1416
.

78

Bai
 
X
,
Zhang
 
B
,
Wang
 
P
,
Wang
 
G
,
Li
 
J
,
Wen
 
D
,
Long
 
X
,
Sun
 
H
,
Liu
 
Y
,
Huang
 
M
,
Zhong
 
S.
 
Effects of SLCO1B1 and GATM gene variants on rosuvastatin-induced myopathy are unrelated to high plasma exposure of rosuvastatin and its metabolites
.
Acta Pharmacol Sin
 
2019
;
40
:
492
499
.

79

Birmingham
 
BK
,
Bujac
 
SR
,
Elsby
 
R
,
Azumaya
 
CT
,
Wei
 
C
,
Chen
 
Y
,
Mosqueda-Garcia
 
R
,
Ambrose
 
HJ.
 
Impact of ABCG2 and SLCO1B1 polymorphisms on pharmacokinetics of rosuvastatin, atorvastatin and simvastatin acid in Caucasian and Asian subjects: a class effect?
 
Eur J Clin Pharmacol
 
2015
;
71
:
341
355
.

80

Peyser
 
B
,
Perry
 
EP
,
Singh
 
K
,
Gill
 
RD
,
Mehan
 
MR
,
Haga
 
SB
,
Musty
 
MD
,
Milazzo
 
NA
,
Savard
 
D
,
Li
 
Y-J
,
Trujilio
 
G
,
Voora
 
D.
 
Effects of delivering SLCO1B1 pharmacogenetic information in randomized trial and observational settings
.
Circ Genomic Precis Med
 
2018
;
11
.

81

Vassy
 
JL
,
Gaziano
 
JM
,
Green
 
RC
,
Ferguson
 
RE
,
Advani
 
S
,
Miller
 
SJ
,
Chun
 
S
,
Hage
 
AK
,
Seo
 
S-J
,
Majahalme
 
N
,
MacMullen
 
L
,
Zimolzak
 
AJ
,
Brunette
 
CA.
 
Effect of pharmacogenetic testing for statin myopathy risk vs usual care on blood cholesterol
.
JAMA Netw Open
 
2020
;
3
:
e2027092
.

82

Yates
 
A
,
Akanni
 
W
,
Amode
 
MR
,
Barrell
 
D
,
Billis
 
K
,
Carvalho-Silva
 
D
,
Cummins
 
C
,
Clapham
 
P
,
Fitzgerald
 
S
,
Gil
 
L
,
Girón
 
CG
,
Gordon
 
L
,
Hourlier
 
T
,
Hunt
 
SE
,
Janacek
 
SH
,
Johnson
 
N
,
Juettemann
 
T
,
Keenan
 
S
,
Lavidas
 
I
,
Martin
 
FJ
,
Maurel
 
T
,
McLaren
 
W
,
Murphy
 
DN
,
Nag
 
R
,
Nuhn
 
M
,
Parker
 
A
,
Patricio
 
M
,
Pignatelli
 
M
,
Rahtz
 
M
,
Riat
 
HS
,
Sheppard
 
D
,
Taylor
 
K
,
Thormann
 
A
,
Vullo
 
A
,
Wilder
 
SP
,
Zadissa
 
A
,
Birney
 
E
,
Harrow
 
J
,
Muffato
 
M
,
Perry
 
E
,
Ruffier
 
M
,
Spudich
 
G
,
Trevanion
 
SJ
,
Cunningham
 
F
,
Aken
 
BL
,
Zerbino
 
DR
,
Flicek
 
P.
 
Ensembl 2016
.
Nucleic Acids Res
 
2016
;
44
:
D710
D716
.

83

Bank
 
P
,
Caudle
 
K
,
Swen
 
J
,
Gammal
 
R
,
Whirl‐Carrillo
 
M
,
Klein
 
T
,
Relling
 
M
,
Guchelaar
 
H.
 
Comparison of the guidelines of the clinical pharmacogenetics implementation consortium and the Dutch Pharmacogenetics Working Group
.
Clin Pharmacol Ther
 
2018
;
103
:
599
618
.

84

Summary of Product Characteristics: Simvastatin
. https://mri.cts-mrp.eu/human/downloads/IT_H_0729_001_FinalPI.pdf (20 October
2020
).

85

Wilke
 
RA
,
Ramsey
 
LB
,
Johnson
 
SG
,
Maxwell
 
WD
,
McLeod
 
HL
,
Voora
 
D
,
Krauss
 
RM
,
Roden
 
DM
,
Feng
 
Q
,
Cooper-DeHoff
 
RM
,
Gong
 
L
,
Klein
 
TE
,
Wadelius
 
M
,
Niemi
 
M
;
Clinical Pharmacogenomics Implementation Consortium (CPIC)
.
The Clinical Pharmacogenomics Implementation Consortium: CPIC Guideline for SLCO1B1 and simvastatin-induced myopathy
.
Clin Pharmacol Ther
 
2012
;
92
:
112
117
.

86

Bank
 
PCD
,
Swen
 
JJ
,
Guchelaar
 
FJ
.
Estimated nationwide impact of implementing a preemptive pharmacogenetic panel approach to guide drug prescribing in primary care in The Netherlands
.
BMC Med
 
2019
;
17
:
110
114

87

Pasanen
 
MK
,
Neuvonen
 
M
,
Neuvonen
 
PJ
,
Niemi
 
M.
 
SLCO1B1 polymorphism markedly affects the pharmacokinetics of simvastatin acid
.
Pharmacogenet Genomics
 
2006
;
16
:
873
879
.

88

Tornio
 
A
,
Vakkilainen
 
J
,
Neuvonen
 
M
,
Backman
 
JT
,
Neuvonen
 
PJ
,
Niemi
 
M.
 
SLCO1B1 polymorphism markedly affects the pharmacokinetics of lovastatin acid
.
Pharmacogenet Genomics
 
2015
;
25
:
382
387
.

89

Deng
 
JW
,
Song
 
I-S
,
Shin
 
HJ
,
Yeo
 
C-W
,
Cho
 
D-Y
,
Shon
 
J-H
,
Shin
 
J-G.
 
The effect of SLCO1B115 on the disposition of pravastatin and pitavastatin is substrate dependent: the contribution of transporting activity changes by SLCO1B115
.
Pharmacogenet Genomics
 
2008
;
18
:
424
433
.

90

Pasanen
 
MK
,
Fredrikson
 
H
,
Neuvonen
 
PJ
,
Niemi
 
M.
 
Different effects of SLCO1B1 polymorphism on the pharmacokinetics of atorvastatin and rosuvastatin
.
Clin Pharmacol Ther
 
2007
;
82
:
726
733
.

91

Niemi
 
M
,
Pasanen
 
M
,
Neuvonen
 
P.
 
SLCO1B1 polymorphism and sex affect the pharmacokinetics of pravastatin but not fluvastatin
.
Clin Pharmacol Ther
 
2006
;
80
:
356
366
.

92

Zisaki
 
A
,
Miskovic
 
L
,
Hatzimanikatis
 
V.
 
Antihypertensive drugs metabolism: an update to pharmacokinetic profiles and computational approaches
.
Curr Pharm Des
 
2015
;
21
:
806
822
.

93

Zanger
 
UM
,
Schwab
 
M.
 
Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation
.
Pharmacol Ther
 
2013
;
138
:
103
141
.

94

Ingelman-Sundberg
 
M
,
Sim
 
SC
,
Gomez
 
A
,
Rodriguez-Antona
 
C.
 
Influence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspects
.
Pharmacol Ther
 
2007
;
116
:
496
526
.

95

Leopold
 
G.
 
Balanced pharmacokinetics and metabolism of bisoprolol
.
J Cardiovasc Pharmacol J Cardiovasc
 
1986
;
8
:
S16
S20
.

96

Bijl
 
MJ
,
Visser
 
LE
,
Schaik
 
RHN
,
Van Kors
 
JA
,
Witteman
 
JCM
,
Hofman
 
A
,
Vulto
 
AG
,
Gelder
 
T
,
Van Stricker
 
BHC.
 
Genetic variation in the CYP2D6 gene is associated with a lower heart rate and blood pressure in β-blocker users
.
Clin Pharmacol Ther
 
2009
;
85
:
45
50
.

97

Luzum
 
JA
,
Sweet
 
KM
,
Binkley
 
PF
,
Schmidlen
 
TJ
,
Jarvis
 
JP
,
Christman
 
MF
,
Sadee
 
W
,
Kitzmiller
 
JP.
 
CYP2D6 genetic variation and beta-blocker maintenance dose in patients with heart failure
.
Pharm Res
 
2017
;
34
:
1615
1625
.

98

Rau
 
T
,
Wuttke
 
H
,
Michels
 
LM
,
Werner
 
U
,
Bergmann
 
K
,
Kreft
 
M
,
Fromm
 
MF
,
Eschenhagen
 
T.
 
Impact of the CYP2D6 genotype on the clinical effects of metoprolol: A prospective longitudinal study
.
Clin Pharmacol Ther Clin Pharmacol Ther
 
2009
;
85
:
269
272
.

99

Anstensrud
 
AK
,
Molden
 
E
,
Haug
 
HJ
,
Qazi
 
R
,
Muriq
 
H
,
Fosshaug
 
LE
,
Spigset
 
O
,
Øie
 
E.
 
Impact of genotype-predicted CYP2D6 metabolism on clinical effects and tolerability of metoprolol in patients after myocardial infarction—a prospective observational study
.
Eur J Clin Pharmacol
 
2020
;
76
:
673
683
.

100

Sharp
 
CF
,
Gardiner
 
SJ
,
Jensen
 
BP
,
Roberts
 
RL
,
Troughton
 
RW
,
Lainchbury
 
JG
,
Begg
 
EJ.
 
CYP2D6 genotype and its relationship with metoprolol dose, concentrations and effect in patients with systolic heart failure
.
Pharmacogenomics J
 
2009
;
9
:
175
184
.

101

Batty
 
JA
,
Hall
 
AS
,
White
 
HL
,
Wikstrand
 
J
,
Boer
 
RA
,
De Veldhuisen
 
DJ
,
Van Harst
 
P
,
Van Der Waagstein
 
F
,
Hjalmarson
 
Å
,
Kjekshus
 
J
,
Balmforth
 
AJ.
 
An investigation of CYP2D6 genotype and response to metoprolol CR/XL during dose titration in patients with heart failure: A MERIT-HF substudy
.
Clin Pharmacol Ther
 
2014
;
95
:
321
330
.

102

Hamadeh
 
IS
,
Langaee
 
TY
,
Dwivedi
 
R
,
Garcia
 
S
,
Burkley
 
BM
,
Skaar
 
TC
,
Chapman
 
AB
,
Gums
 
JG
,
Turner
 
ST
,
Gong
 
Y
,
Cooper-Dehoff
 
RM
,
Johnson
 
JA.
 
Impact of CYP2D6 polymorphisms on clinical efficacy and tolerability of metoprolol tartrate
.
Clin Pharmacol Ther
 
2014
;
96
:
175
181
.

103

Kirchheiner
 
J
,
Heesch
 
C
,
Bauer
 
S
,
Meisel
 
C
,
Seringer
 
A
,
Goldammer
 
M
,
Tzvetkov
 
M
,
Meineke
 
I
,
Roots
 
I
,
Brockmöller
 
J.
 
Impact of the ultrarapid metabolizer genotype of cytochrome P450 2D6 on metoprolol pharmacokinetics and pharmacodynamics
.
Clin Pharmacol Ther
 
2004
;
76
:
302
312
.

104

Goryachkina
 
K
,
Burbello
 
A
,
Boldueva
 
S
,
Babak
 
S
,
Bergman
 
U
,
Bertilsson
 
L.
 
CYP2D6 is a major determinant of metoprolol disposition and effects in hospitalized Russian patients treated for acute myocardial infarction
.
Eur J Clin Pharmacol
 
2008
;
64
:
1163
1173
.

105

Zineh
 
I
,
Beitelshees
 
AL
,
Gaedigk
 
A
,
Walker
 
JR
,
Pauly
 
DF
,
Eberst
 
K
,
Leeder
 
JS
,
Phillips
 
MS
,
Gelfand
 
CA
,
Johnson
 
JA.
 
Pharmacokinetics and CYP2D6 genotypes do not predict metoprolol adverse events or efficacy in hypertension
.
Clin Pharmacol Ther
 
2004
;
76
:
536
544
.

106

Wuttke
 
H
,
Rau
 
T
,
Heide
 
R
,
Bergmann
 
K
,
Böhm
 
M
,
Weil
 
J
,
Werner
 
D
,
Eschenhagen
 
T.
 
Increased frequency of cytochrome P450 2D6 poor metabolizers among patients with metoprolol-associated adverse effects
.
Clin Pharmacol Ther
 
2002
;
72
:
429
437
.

107

Fux
 
R
,
Mörike
 
K
,
Pröhmer
 
AMT
,
Delabar
 
U
,
Schwab
 
M
,
Schaeffeler
 
E
,
Lorenz
 
G
,
Gleiter
 
CH
,
Eichelbaum
 
M
,
Kivistö
 
KT.
 
Impact of CYP2D6 genotype on adverse effects during treatment with metoprolol: A prospective clinical study
.
Clin Pharmacol Ther
 
2005
;
78
:
378
387
.

108

Novartis Pharmaceuticals Corporation
. LOPRESSOR (metoprolol tartrate).
2008
. FDA. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/017963s062,018704s021lbl.pdf.

109

Blake
 
CM
,
Kharasch
 
ED
,
Schwab
 
M
,
Nagele
 
P.
 
A meta-analysis of CYP2D6 metabolizer phenotype and metoprolol pharmacokinetics
.
Clin Pharmacol Ther
 
2013
;
94
:
394
399
.

110

Williams
 
B
,
Mancia
 
G
,
Spiering
 
W
,
Agabiti Rosei
 
E
,
Azizi
 
M
,
Burnier
 
M
,
Clement
 
DL
,
Coca
 
A
,
de Simone
 
G
,
Dominiczak
 
A
,
Kahan
 
T
,
Mahfoud
 
F
,
Redon
 
J
,
Ruilope
 
L
,
Zanchetti
 
A
,
Kerins
 
M
,
Kjeldsen
 
SE
,
Kreutz
 
R
,
Laurent
 
S
,
Lip
 
GYH
,
McManus
 
R
,
Narkiewicz
 
K
,
Ruschitzka
 
F
,
Schmieder
 
RE
,
Shlyakhto
 
E
,
Tsioufis
 
C
,
Aboyans
 
V
,
Desormais
 
I
;
Authors/Task Force Members
.
2018 ESC/ESH Guidelines for the management of arterial hypertension
.
J Hypertens
 
2018
;
36
:
1953
2041
.

111

NCD Risk Factor Collaboration (NCD-RisC)
.
Worldwide trends in blood pressure from 1975 to 2015: a pooled analysis of 1479 population-based measurement studies with 19·1 million participants
.
Lancet
 
2017
;
389
:
37
55
.

112

Evangelou
 
E
,
Warren
 
HR
,
Mosen-Ansorena
 
D
,
Mifsud
 
B
,
Pazoki
 
R
,
Gao
 
H
,
Ntritsos
 
G
,
Dimou
 
N
,
Cabrera
 
CP
,
Karaman
 
I
,
Ng
 
FL
,
Evangelou
 
M
,
Witkowska
 
K
,
Tzanis
 
E
,
Hellwege
 
JN
,
Giri
 
A
,
Velez Edwards
 
DR
,
Sun
 
YV
,
Cho
 
K
,
Gaziano
 
JM
,
Wilson
 
PWF
,
Tsao
 
PS
,
Kovesdy
 
CP
,
Esko
 
T
,
Mägi
 
R
,
Milani
 
L
,
Almgren
 
P
,
Boutin
 
T
,
Debette
 
S
,
Ding
 
J
,
Giulianini
 
F
,
Holliday
 
EG
,
Jackson
 
AU
,
Li-Gao
 
R
,
Lin
 
W-Y
,
Luan
 
J
,
Mangino
 
M
,
Oldmeadow
 
C
,
Prins
 
BP
,
Qian
 
Y
,
Sargurupremraj
 
M
,
Shah
 
N
,
Surendran
 
P
,
Thériault
 
S
,
Verweij
 
N
,
Willems
 
SM
,
Zhao
 
J-H
,
Amouyel
 
P
,
Connell
 
J
,
de Mutsert
 
R
,
Doney
 
ASF
,
Farrall
 
M
,
Menni
 
C
,
Morris
 
AD
,
Noordam
 
R
,
Paré
 
G
,
Poulter
 
NR
,
Shields
 
DC
,
Stanton
 
A
,
Thom
 
S
,
Abecasis
 
G
,
Amin
 
N
,
Arking
 
DE
,
Ayers
 
KL
,
Barbieri
 
CM
,
Batini
 
C
,
Bis
 
JC
,
Blake
 
T
,
Bochud
 
M
,
Boehnke
 
M
,
Boerwinkle
 
E
,
Boomsma
 
DI
,
Bottinger
 
EP
,
Braund
 
PS
,
Brumat
 
M
,
Campbell
 
A
,
Campbell
 
H
,
Chakravarti
 
A
,
Chambers
 
JC
,
Chauhan
 
G
,
Ciullo
 
M
,
Cocca
 
M
,
Collins
 
F
,
Cordell
 
HJ
,
Davies
 
G
,
de Borst
 
MH
,
de Geus
 
EJ
,
Deary
 
IJ
,
Deelen
 
J
,
Del Greco M
 
F
,
Demirkale
 
CY
,
Dörr
 
M
,
Ehret
 
GB
,
Elosua
 
R
,
Enroth
 
S
,
Erzurumluoglu
 
AM
,
Ferreira
 
T
,
Frånberg
 
M
,
Franco
 
OH
,
Gandin
 
I
,
Gasparini
 
P
,
Giedraitis
 
V
,
Gieger
 
C
,
Girotto
 
G
,
Goel
 
A
,
Gow
 
AJ
,
Gudnason
 
V
,
Guo
 
X
,
Gyllensten
 
U
,
Hamsten
 
A
,
Harris
 
TB
,
Harris
 
SE
,
Hartman
 
CA
,
Havulinna
 
AS
,
Hicks
 
AA
,
Hofer
 
E
,
Hofman
 
A
,
Hottenga
 
J-J
,
Huffman
 
JE
,
Hwang
 
S-J
,
Ingelsson
 
E
,
James
 
A
,
Jansen
 
R
,
Jarvelin
 
M-R
,
Joehanes
 
R
,
Johansson
 
Å
,
Johnson
 
AD
,
Joshi
 
PK
,
Jousilahti
 
P
,
Jukema
 
JW
,
Jula
 
A
,
Kähönen
 
M
,
Kathiresan
 
S
,
Keavney
 
BD
,
Khaw
 
K-T
,
Knekt
 
P
,
Knight
 
J
,
Kolcic
 
I
,
Kooner
 
JS
,
Koskinen
 
S
,
Kristiansson
 
K
,
Kutalik
 
Z
,
Laan
 
M
,
Larson
 
M
,
Launer
 
LJ
,
Lehne
 
B
,
Lehtimäki
 
T
,
Liewald
 
DCM
,
Lin
 
L
,
Lind
 
L
,
Lindgren
 
CM
,
Liu
 
YMei
,
Loos
 
RJF
,
Lopez
 
LM
,
Lu
 
Y
,
Lyytikäinen
 
L-P
,
Mahajan
 
A
,
Mamasoula
 
C
,
Marrugat
 
J
,
Marten
 
J
,
Milaneschi
 
Y
,
Morgan
 
A
,
Morris
 
AP
,
Morrison
 
AC
,
Munson
 
PJ
,
Nalls
 
MA
,
Nandakumar
 
P
,
Nelson
 
CP
,
Niiranen
 
T
,
Nolte
 
IM
,
Nutile
 
T
,
Oldehinkel
 
AJ
,
Oostra
 
BA
,
O'Reilly
 
PF
,
Org
 
E
,
Padmanabhan
 
S
,
Palmas
 
W
,
Palotie
 
A
,
Pattie
 
A
,
Penninx
 
BWJH
,
Perola
 
M
,
Peters
 
A
,
Polasek
 
O
,
Pramstaller
 
PP
,
Nguyen
 
QT
,
Raitakari
 
OT
,
Ren
 
M
,
Rettig
 
R
,
Rice
 
K
,
Ridker
 
PM
,
Ried
 
JS
,
Riese
 
H
,
Ripatti
 
S
,
Robino
 
A
,
Rose
 
LM
,
Rotter
 
JI
,
Rudan
 
I
,
Ruggiero
 
D
,
Saba
 
Y
,
Sala
 
CF
,
Salomaa
 
V
,
Samani
 
NJ
,
Sarin
 
A-P
,
Schmidt
 
R
,
Schmidt
 
H
,
Shrine
 
N
,
Siscovick
 
D
,
Smith
 
AV
,
Snieder
 
H
,
Sõber
 
S
,
Sorice
 
R
,
Starr
 
JM
,
Stott
 
DJ
,
Strachan
 
DP
,
Strawbridge
 
RJ
,
Sundström
 
J
,
Swertz
 
MA
,
Taylor
 
KD
,
Teumer
 
A
,
Tobin
 
MD
,
Tomaszewski
 
M
,
Toniolo
 
D
,
Traglia
 
M
,
Trompet
 
S
,
Tuomilehto
 
J
,
Tzourio
 
C
,
Uitterlinden
 
AG
,
Vaez
 
A
,
van der Most
 
PJ
,
van Duijn
 
CM
,
Vergnaud
 
A-C
,
Verwoert
 
GC
,
Vitart
 
V
,
Völker
 
U
,
Vollenweider
 
P
,
Vuckovic
 
D
,
Watkins
 
H
,
Wild
 
SH
,
Willemsen
 
G
,
Wilson
 
JF
,
Wright
 
AF
,
Yao
 
J
,
Zemunik
 
T
,
Zhang
 
W
,
Attia
 
JR
,
Butterworth
 
AS
,
Chasman
 
DI
,
Conen
 
D
,
Cucca
 
F
,
Danesh
 
J
,
Hayward
 
C
,
Howson
 
JMM
,
Laakso
 
M
,
Lakatta
 
EG
,
Langenberg
 
C
,
Melander
 
O
,
Mook-Kanamori
 
DO
,
Palmer
 
CNA
,
Risch
 
L
,
Scott
 
RA
,
Scott
 
RJ
,
Sever
 
P
,
Spector
 
TD
,
van der Harst
 
P
,
Wareham
 
NJ
,
Zeggini
 
E
,
Levy
 
D
,
Munroe
 
PB
,
Newton-Cheh
 
C
,
Brown
 
MJ
,
Metspalu
 
A
,
Hung
 
AM
,
O'Donnell
 
CJ
,
Edwards
 
TL
,
Psaty
 
BM
,
Tzoulaki
 
I
,
Barnes
 
MR
,
Wain
 
LV
,
Elliott
 
P
,
Caulfield
 
MJ
,
Million Veteran Program
.
Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits
.
Nat Genet
 
2018
;
50
:
1412
1425
.

113

Giri
 
A
,
Hellwege
 
JN
,
Keaton
 
JM
,
Park
 
J
,
Qiu
 
C
,
Warren
 
HR
,
Torstenson
 
ES
,
Kovesdy
 
CP
,
Sun
 
YV
,
Wilson
 
OD
,
Robinson-Cohen
 
C
,
Roumie
 
CL
,
Chung
 
CP
,
Birdwell
 
KA
,
Damrauer
 
SM
,
DuVall
 
SL
,
Klarin
 
D
,
Cho
 
K
,
Wang
 
Y
,
Evangelou
 
E
,
Cabrera
 
CP
,
Wain
 
LV
,
Shrestha
 
R
,
Mautz
 
BS
,
Akwo
 
EA
,
Sargurupremraj
 
M
,
Debette
 
S
,
Boehnke
 
M
,
Scott
 
LJ
,
Luan
 
J
,
Zhao
 
J-H
,
Willems
 
SM
,
Thériault
 
S
,
Shah
 
N
,
Oldmeadow
 
C
,
Almgren
 
P
,
Li-Gao
 
R
,
Verweij
 
N
,
Boutin
 
TS
,
Mangino
 
M
,
Ntalla
 
I
,
Feofanova
 
E
,
Surendran
 
P
,
Cook
 
JP
,
Karthikeyan
 
S
,
Lahrouchi
 
N
,
Liu
 
C
,
Sepúlveda
 
N
,
Richardson
 
TG
,
Kraja
 
A
,
Amouyel
 
P
,
Farrall
 
M
,
Poulter
 
NR
,
Laakso
 
M
,
Zeggini
 
E
,
Sever
 
P
,
Scott
 
RA
,
Langenberg
 
C
,
Wareham
 
NJ
,
Conen
 
D
,
Palmer
 
CNA
,
Attia
 
J
,
Chasman
 
DI
,
Ridker
 
PM
,
Melander
 
O
,
Mook-Kanamori
 
DO
,
Harst
 
P. V D
,
Cucca
 
F
,
Schlessinger
 
D
,
Hayward
 
C
,
Spector
 
TD
,
Jarvelin
 
M-R
,
Hennig
 
BJ
,
Timpson
 
NJ
,
Wei
 
W-Q
,
Smith
 
JC
,
Xu
 
Y
,
Matheny
 
ME
,
Siew
 
EE
,
Lindgren
 
C
,
Herzig
 
K-H
,
Dedoussis
 
G
,
Denny
 
JC
,
Psaty
 
BM
,
Howson
 
JMM
,
Munroe
 
PB
,
Newton-Cheh
 
C
,
Caulfield
 
MJ
,
Elliott
 
P
,
Gaziano
 
JM
,
Concato
 
J
,
Wilson
 
PWF
,
Tsao
 
PS
,
Velez Edwards
 
DR
,
Susztak
 
K
,
O'Donnell
 
CJ
,
Hung
 
AM
,
Edwards
 
TL
;
Million Veteran Program
.
Trans-ethnic association study of blood pressure determinants in over 750,000 individuals
.
Nat Genet
 
2019
;
51
:
51
62
.

114

Lynch
 
AI
,
Eckfeldt
 
JH
,
Davis
 
BR
,
Ford
 
CE
,
Boerwinkle
 
E
,
Leiendecker-Foster
 
C
,
Arnett
 
DK.
 
Gene panels to help identify subgroups at high and low risk of coronary heart disease among those randomized to antihypertensive treatment
.
Pharmacogenet Genomics
 
2012
;
1
.

115

Johnson
 
JA.
 
Ethnic differences in cardiovascular drug response
.
Circulation
 
2008
;
118
:
1383
1393
.

116

Schreieck
 
J
,
Dostal
 
S
,
von
 
BN
,
Wacker
 
A
,
Flory
 
M
,
Weyerbrock
 
S
,
Koch
 
W
,
Schömig
 
A
,
Schmitt
 
C.
 
C825T polymorphism of the G-protein β3 subunit gene and atrial fibrillation: Association of the TT genotype with a reduced risk for atrial fibrillation
.
Am Heart J
 
2004
;
148
:
545
550
.

117

Roden
 
DM.
 
Drug-induced prolongation of the QT interval
.
N Engl J Med
 
2004
;
350
:
1013
1022
.

118

Zipes
 
DP
,
Wellens
 
HJJ.
 
Sudden cardiac death
.
Circulation
 
1998
;
98
:
2334
2351
.

119

International Conference on Harmonisation; guidance on E14 clinical evaluation of QT/QTc interval prolongation and proarrhythmic potential for non-antiarrhythmic drugs; availability. Notice
.
Fed Regist
 
2005
;
70
:
61134
61135
.

120

Kannankeril
 
P
,
Roden
 
DM
,
Darbar
 
D.
 
Drug-induced long QT syndrome
.
Pharmacol Rev
 
2010
;
62
:
760
781
.

121

Kannankeril
 
PJ
,
Roden
 
DM.
 
Drug-induced long QT and torsade de pointes: recent advances
.
Curr Opin Cardiol
 
2007
;
22
:
39
43
.

122

Morita
 
H
,
Wu
 
J
,
Zipes
 
DP.
 
The QT syndromes: long and short
.
Lancet
 
2008
;
372
:
750
763
.

123

CredibleMeds
. CredibleMeds.org (19 October
2020
).

124

Mahida
 
S
,
Hogarth
 
AJ
,
Cowan
 
C
,
Tayebjee
 
MH
,
Graham
 
LN
,
Pepper
 
CB.
 
Genetics of congenital and drug-induced long QT syndromes: current evidence and future research perspectives
.
J Interv Card Electrophysiol
 
2013
;
37
:
9
19
.

125

Hong
 
Y
,
Rautaharju
 
P
,
Hopkins
 
P
,
Arnett
 
D
,
Djoussé
 
L
,
Pankow
 
J
,
Sholinsky
 
P
,
Rao
 
D
,
Province
 
M.
 
Familial aggregation of QT-interval variability in a general population: results from the NHLBI Family Heart Study
.
Clin Genet
 
2001
;
59
:
171
177
.

126

Newton-Cheh
 
C
,
Larson
 
MG
,
Corey
 
DC
,
Benjamin
 
EJ
,
Herbert
 
AG
,
Levy
 
D
,
D’Agostino
 
RB
,
O’Donnell
 
CJ.
 
QT interval is a heritable quantitative trait with evidence of linkage to chromosome 3 in a genome-wide linkage analysis: the Framingham Heart Study
.
Heart Rhythm
 
2005
;
2
:
277
284
.

127

Kannankeril
 
PJ
,
Roden
 
DM
,
Norris
 
KJ
,
Whalen
 
SP
,
George
 
ALJ
,
Murray
 
KT.
 
Genetic susceptibility to acquired long QT syndrome: pharmacologic challenge in first-degree relatives
.
Heart Rhythm
 
2005
;
2
:
134
140
.

128

Chakravarti
 
A
,
Pfeufer
 
A
,
Kashuk
 
C
,
Guo
 
C-Y
,
Gieger
 
C
,
Newton-Cheh
 
C
,
O’Donnell
 
CJ
,
Arking
 
DE
,
Marbán
 
E
,
Wichmann
 
HE
,
Hirschhorn
 
JN
,
West
 
K
,
Akyol
 
M
,
Larson
 
MG
,
Ikeda
 
M
,
Spooner
 
PM
,
Jalilzadeh
 
S
,
Perz
 
S
,
Kääb
 
S
,
Illig
 
T
,
Meitinger
 
T
,
Kao
 
WHL
,
Post
 
W.
 
A common genetic variant in the NOS1 regulator NOS1AP modulates cardiac repolarization
.
Nat Genet
 
2006
;
38
:
644
651
.

129

Pfeufer
 
A
,
Sanna
 
S
,
Arking
 
DE
,
Müller
 
M
,
Gateva
 
V
,
Fuchsberger
 
C
,
Ehret
 
GB
,
Orrú
 
M
,
Pattaro
 
C
,
Köttgen
 
A
,
Perz
 
S
,
Usala
 
G
,
Barbalic
 
M
,
Li
 
M
,
Pütz
 
B
,
Scuteri
 
A
,
Prineas
 
RJ
,
Sinner
 
MF
,
Gieger
 
C
,
Najjar
 
SS
,
Kao
 
WHL
,
Mühleisen
 
TW
,
Dei
 
M
,
Happle
 
C
,
Möhlenkamp
 
S
,
Crisponi
 
L
,
Erbel
 
R
,
Jöckel
 
K-H
,
Naitza
 
S
,
Steinbeck
 
G
,
Marroni
 
F
,
Hicks
 
AA
,
Lakatta
 
E
,
Müller-Myhsok
 
B
,
Pramstaller
 
PP
,
Wichmann
 
H-E
,
Schlessinger
 
D
,
Boerwinkle
 
E
,
Meitinger
 
T
,
Uda
 
M
,
Coresh
 
J
,
Kääb
 
S
,
Abecasis
 
GR
,
Chakravarti
 
A.
 
Common variants at ten loci modulate the QT interval duration in the QTSCD Study
.
Nat Genet
 
2009
;
41
:
407
414
.

130

Newton-Cheh
 
C
,
Eijgelsheim
 
M
,
Rice
 
KM
,
de Bakker
 
PIW
,
Yin
 
X
,
Estrada
 
K
,
Bis
 
JC
,
Marciante
 
K
,
Rivadeneira
 
F
,
Noseworthy
 
PA
,
Sotoodehnia
 
N
,
Smith
 
NL
,
Rotter
 
JI
,
Kors
 
JA
,
Witteman
 
JCM
,
Hofman
 
A
,
Heckbert
 
SR
,
O'Donnell
 
CJ
,
Uitterlinden
 
AG
,
Psaty
 
BM
,
Lumley
 
T
,
Larson
 
MG
,
Ch Stricker
 
BH.
 
Common variants at ten loci influence QT interval duration in the QTGEN Study
.
Nat Genet
 
2009
;
41
:
399
406
.

131

Post
 
W
,
Shen
 
H
,
Damcott
 
C
,
Arking
 
DE
,
Kao
 
WHL
,
Sack
 
PA
,
Ryan
 
KA
,
Chakravarti
 
A
,
Mitchell
 
BD
,
Shuldiner
 
AR.
 
Associations between genetic variants in the NOS1AP (CAPON) gene and cardiac repolarization in the Old Order Amish
.
Hum Hered
 
2007
;
64
:
214
219
.

132

Aarnoudse
 
A-JLHJ
,
Straus
 
SMJM
,
Hofman
 
A
,
Uitterlinden
 
AG
,
Witteman
 
JCM
,
Stricker
 
BHC
,
Newton-Cheh
 
C
,
Kors
 
JA
,
de Bakker
 
PIW.
 
Common NOS1AP variants are associated with a prolonged QTc interval in the Rotterdam study
.
Circulation
 
2007
;
116
.

133

Tobin
 
MD
,
Kähönen
 
M
,
Braund
 
P
,
Nieminen
 
T
,
Hajat
 
C
,
Tomaszewski
 
M
,
Viik
 
J
,
Lehtinen
 
R
,
Ng
 
GA
,
Macfarlane
 
PW
,
Burton
 
PR
,
Lehtimäki
 
T
,
Samani
 
NJ.
 
Gender and effects of a common genetic variant in the NOS1 regulator NOS1AP on cardiac repolarization in 3761 individuals from two independent populations
.
Int J Epidemiol
 
2008
;
37
:
1132
141
.

134

Lehtinen
 
AB
,
Newton-Cheh
 
C
,
Ziegler
 
JT
,
Langefeld
 
CD
,
Freedman
 
BI
,
Daniel
 
KR
,
Herrington
 
DM
,
Bowden
 
DW.
 
Association of NOS1AP genetic variants with QT interval duration in families from the diabetes Heart Study
.
Diabetes
 
2008
;
57
:
1108
1114
.

135

Kapoor
 
A
,
Sekar
 
RB
,
Hansen
 
NF
,
Fox-Talbot
 
K
,
Morley
 
M
,
Pihur
 
V
,
Chatterjee
 
S
,
Brandimarto
 
J
,
Moravec
 
CS
,
Pulit
 
SL
,
Pfeufer
 
A
,
Mullikin
 
J
,
Ross
 
M
,
Green
 
ED
,
Bentley
 
D
,
Newton-Cheh
 
C
,
Boerwinkle
 
E
,
Tomaselli
 
GF
,
Cappola
 
TP
,
Arking
 
DE
,
Halushka
 
MK
,
Chakravarti
 
A.
 
An enhancer polymorphism at the cardiomyocyte intercalated disc protein NOS1AP locus is a major regulator of the QT interval
.
Am J Hum Genet
 
2014
;
94
:
854
869
.

136

Behr
 
ER
,
Ritchie
 
MD
,
Tanaka
 
T
,
Kääb
 
S
,
Crawford
 
DC
,
Nicoletti
 
P
,
Floratos
 
A
,
Sinner
 
MF
,
Kannankeril
 
PJ
,
Wilde
 
AAM
,
Bezzina
 
CR
,
Schulze-Bahr
 
E
,
Zumhagen
 
S
,
Guicheney
 
P
,
Bishopric
 
NH
,
Marshall
 
V
,
Shakir
 
S
,
Dalageorgou
 
C
,
Bevan
 
S
,
Jamshidi
 
Y
,
Bastiaenen
 
R
,
Myerburg
 
RJ
,
Schott
 
J-J
,
Camm
 
AJ
,
Steinbeck
 
G
,
Norris
 
K
,
Altman
 
RB
,
Tatonetti
 
NP
,
Jeffery
 
S
,
Kubo
 
M
,
Nakamura
 
Y
,
Shen
 
Y
,
George
 
AL
,
Roden
 
DM.
 
Genome wide analysis of drug-induced torsades de pointes: lack of common variants with large effect sizes
.
PLoS One
 
2013
;
8
:
e78511
.

137

Strauss
 
DG
,
Vicente
 
J
,
Johannesen
 
L
,
Blinova
 
K
,
Mason
 
JW
,
Weeke
 
P
,
Behr
 
ER
,
Roden
 
DM
,
Woosley
 
R
,
Kosova
 
G
,
Rosenberg
 
MA
,
Newton-Cheh
 
C.
 
Common genetic variant risk score is associated with drug-induced QT prolongation and torsade de pointes risk
.
Circulation
 
2017
;
135
:
1300
1310
.

138

Kääb
 
S
,
Crawford
 
DC
,
Sinner
 
MF
,
Behr
 
ER
,
Kannankeril
 
PJ
,
Wilde
 
AAM
,
Bezzina
 
CR
,
Schulze-Bahr
 
E
,
Guicheney
 
P
,
Bishopric
 
NH
,
Myerburg
 
RJ
,
Schott
 
J-J
,
Pfeufer
 
A
,
Beckmann
 
B-M
,
Martens
 
E
,
Zhang
 
T
,
Stallmeyer
 
B
,
Zumhagen
 
S
,
Denjoy
 
I
,
Bardai
 
A
,
Van Gelder
 
IC
,
Jamshidi
 
Y
,
Dalageorgou
 
C
,
Marshall
 
V
,
Jeffery
 
S
,
Shakir
 
S
,
Camm
 
AJ
,
Steinbeck
 
G
,
Perz
 
S
,
Lichtner
 
P
,
Meitinger
 
T
,
Peters
 
A
,
Wichmann
 
H-E
,
Ingram
 
C
,
Bradford
 
Y
,
Carter
 
S
,
Norris
 
K
,
Ritchie
 
MD
,
George
 
AL
,
Roden
 
DM.
 
A large candidate gene survey identifies the KCNE1D85N polymorphism as a possible modulator of drug-induced torsades de pointes
.
Circ Cardiovasc Genet
 
2012
;
5
:
91
99
.

139

Paulussen
 
ADC
,
Gilissen
 
RAHJ
,
Armstrong
 
M
,
Doevendans
 
PA
,
Verhasselt
 
P
,
Smeets
 
HJM
,
Schulze-Bahr
 
E
,
Haverkamp
 
W
,
Breithardt
 
G
,
Cohen
 
N
,
Aerssens
 
J.
 
Genetic variations of HCNQ1, KCNH2, SCN5A, KCNE1, and KCNE2 in drug-induced long QT syndrome patients
.
J Mol Med
 
2004
;
82
:
182
188
.

140

Weeke
 
P
,
Mosley
 
JD
,
Hanna
 
D
,
Delaney
 
JT
,
Shaffer
 
C
,
Wells
 
QS
,
Driest
 
S
,
Van Karnes
 
JH
,
Ingram
 
C
,
Guo
 
Y
,
Shyr
 
Y
,
Norris
 
K
,
Kannankeril
 
PJ
,
Ramirez
 
AH
,
Smith
 
JD
,
Mardis
 
ER
,
Nickerson
 
D
,
George
 
AL
,
Roden
 
DM.
 
Exome sequencing implicates an increased burden of rare potassium channel variants in the risk of drug-induced long QT interval syndrome
.
J Am Coll Cardiol
 
2014
; 63:
1430
37

141

Roden
 
DM
,
Viswanathan
 
PC.
 
Genetics of acquired long QT syndrome
.
J Clin Invest
 
2005
; 115:
2025
32

142

Magdy
 
T
,
Burmeister
 
BT
,
Burridge
 
PW.
 
Validating the pharmacogenomics of chemotherapy-induced cardiotoxicity: what is missing?
 
Pharmacol Ther
 
2016
;
168
:
113
125
.

143

Aminkeng
 
F
,
Ross
 
CJD
,
Rassekh
 
SR
,
Hwang
 
S
,
Rieder
 
MJ
,
Bhavsar
 
AP
,
Smith
 
A
,
Sanatani
 
S
,
Gelmon
 
KA
,
Bernstein
 
D
,
Hayden
 
MR
,
Amstutz
 
U
,
Carleton
 
BC
;
CPNDS Clinical Practice Recommendations Group
.
Recommendations for genetic testing to reduce the incidence of anthracycline‐induced cardiotoxicity
.
Br J Clin Pharmacol
 
2016
;
82
:
683
695
.

144

Craig
 
LA
,
Ekert
 
PG
,
Conyers
 
R
,
Elliott
 
DA.
 
Genetic determinants of anthracycline cardiotoxicity—ready for the clinic?
 
Br J Clin Pharmacol
 
2017
;
83
:
1141
1142
.

145

Sapkota
 
Y
,
Qin
 
N
,
Ehrhardt
 
MJ
,
Wang
 
Z
,
Chen
 
Y
,
Wilson
 
CL
,
Estepp
 
J
,
Rai
 
P
,
Hankins
 
JS
,
Burridge
 
PW
,
Jefferies
 
JL
,
Zhang
 
J
,
Hudson
 
MM
,
Robison
 
LL
,
Armstrong
 
GT
,
Mulrooney
 
DA
,
Yasui
 
Y.
 
Genetic variants associated with therapy-related cardiomyopathy among childhood cancer survivors of African ancestry
.
Cancer Res
 
2020
;DOI: 10.1158/0008-5472.CAN-20-2675.

146

Garcia-Pavia
 
P
,
Kim
 
Y
,
Restrepo-Cordoba
 
MA
,
Lunde
 
IG
,
Wakimoto
 
H
,
Smith
 
AM
,
Toepfer
 
CN
,
Getz
 
K
,
Gorham
 
J
,
Patel
 
P
,
Ito
 
K
,
Willcox
 
JA
,
Arany
 
Z
,
Li
 
J
,
Owens
 
AT
,
Govind
 
R
,
Nuñez
 
B
,
Mazaika
 
E
,
Bayes-Genis
 
A
,
Walsh
 
R
,
Finkelman
 
B
,
Lupon
 
J
,
Whiffin
 
N
,
Serrano
 
I
,
Midwinter
 
W
,
Wilk
 
A
,
Bardaji
 
A
,
Ingold
 
N
,
Buchan
 
R
,
Tayal
 
U
,
Pascual-Figal
 
DA
,
de Marvao
 
A
,
Ahmad
 
M
,
Garcia-Pinilla
 
JM
,
Pantazis
 
A
,
Dominguez
 
F
,
John Baksi
 
A
,
O’Regan
 
DP
,
Rosen
 
SD
,
Prasad
 
SK
,
Lara-Pezzi
 
E
,
Provencio
 
M
,
Lyon
 
AR
,
Alonso-Pulpon
 
L
,
Cook
 
SA
,
DePalma
 
SR
,
Barton
 
PJR
,
Aplenc
 
R
,
Seidman
 
JG
,
Ky
 
B
,
Ware
 
JS
,
Seidman
 
CE.
 
Genetic variants associated with cancer therapy–induced cardiomyopathy
.
Circulation
 
2019
;
140
:
31
41
.

147

Linschoten
 
M
,
Teske
 
AJ
,
Cramer
 
MJ
,
E van der
 
W
,
Asselbergs
 
FW.
 
Chemotherapy-related cardiac dysfunction
.
Circ Genomic Precis Med
 
2018
;
11
.

148

Jackson
 
N
,
Atar
 
D
,
Borentain
 
M
,
Breithardt
 
G
,
Eickels
 
M
,
van Endres
 
M
,
Fraass
 
U
,
Friede
 
T
,
Hannachi
 
H
,
Janmohamed
 
S
,
Kreuzer
 
J
,
Landray
 
M
,
Lautsch
 
D
,
Floch
 
C
,
Le Mol
 
P
,
Naci
 
H
,
Samani
 
NJ
,
Svensson
 
A
,
Thorstensen
 
C
,
Tijssen
 
J
,
Vandzhura
 
V
,
Zalewski
 
A
,
Kirchhof
 
P.
 
Improving clinical trials for cardiovascular diseases: a position paper from the Cardiovascular Round Table of the European Society of Cardiology
.
Eur Heart J
 
2016
;
37
:
747
754
.

149

Turner
 
RM
,
Pirmohamed
 
M.
 
Cardiovascular pharmacogenomics: expectations and practical benefits
.
Clin Pharmacol Ther
 
2014
;
95
:
281
293
.

150

Pereira
 
NL
,
Sargent
 
DJ
,
Farkouh
 
ME
,
Rihal
 
CS.
 
Genotype-based clinical trials in cardiovascular disease
.
Nat Rev Cardiol
 
2015
;
12
:
475
487
.

151

Piccini
 
JP
,
Abraham
 
WT
,
Dufton
 
C
,
Carroll
 
IA
,
Healey
 
JS
,
Veldhuisen
 
DJ
,
van Sauer
 
WH
,
Anand
 
IS
,
White
 
M
,
Wilton
 
SB
,
Aleong
 
R
,
Rienstra
 
M
,
Krueger
 
SK
,
Ayala-Paredes
 
F
,
Khaykin
 
Y
,
Merkely
 
B
,
Miloradović
 
V
,
Wranicz
 
JK
,
Ilkhanoff
 
L
,
Ziegler
 
PD
,
Davis
 
G
,
Emery
 
LL
,
Marshall
 
D
,
Kao
 
DP
,
Bristow
 
MR
,
Connolly
 
SJ.
 
Bucindolol for the maintenance of sinus rhythm in a genotype-defined HF population
.
JACC Heart Fail
 
2019
;
7
:
586
598
.

152

Tall
 
AR
,
Rader
 
DJ.
 
Trials and Tribulations of CETP Inhibitors
.
Circ Res
 
2018
;
122
:
106
112
.

153

Effects of anacetrapib in patients with atherosclerotic vascular disease
.
N Engl J Med
 
2017
;
377
:
1217
1227
.

154

Tardif
 
J-C
,
Dubé
 
M-P
,
Pfeffer
 
MA
,
Waters
 
DD
,
Koenig
 
W
,
Maggioni
 
AP
,
McMurray
 
JJV
,
Mooser
 
V
,
White
 
HD
,
Heinonen
 
T
,
Black
 
DM
,
Guertin
 
M-C
;
Dal-GenE Investigators
.
Study design of Dal-GenE, a pharmacogenetic trial targeting reduction of cardiovascular events with dalcetrapib
.
Am Heart J
 
2020
;
222
:
157
165
.

155

Cavallari
 
LH
,
Lee
 
CR
,
Beitelshees
 
AL
,
Cooper-DeHoff
 
RM
,
Duarte
 
JD
,
Voora
 
D
,
Kimmel
 
SE
,
McDonough
 
CW
,
Gong
 
Y
,
Dave
 
CV
,
Pratt
 
VM
,
Alestock
 
TD
,
Anderson
 
RD
,
Alsip
 
J
,
Ardati
 
AK
,
Brott
 
BC
,
Brown
 
L
,
Chumnumwat
 
S
,
Clare-Salzler
 
MJ
,
Coons
 
JC
,
Denny
 
JC
,
Dillon
 
C
,
Elsey
 
AR
,
Hamadeh
 
IS
,
Harada
 
S
,
Hillegass
 
WB
,
Hines
 
L
,
Horenstein
 
RB
,
Howell
 
LA
,
Jeng
 
LJB
,
Kelemen
 
MD
,
Lee
 
YM
,
Magvanjav
 
O
,
Montasser
 
M
,
Nelson
 
DR
,
Nutescu
 
EA
,
Nwaba
 
DC
,
Pakyz
 
RE
,
Palmer
 
K
,
Peterson
 
JF
,
Pollin
 
TI
,
Quinn
 
AH
,
Robinson
 
SW
,
Schub
 
J
,
Skaar
 
TC
,
Smith
 
DM
,
Sriramoju
 
VB
,
Starostik
 
P
,
Stys
 
TP
,
Stevenson
 
JM
,
Varunok
 
N
,
Vesely
 
MR
,
Wake
 
DT
,
Weck
 
KE
,
Weitzel
 
KW
,
Wilke
 
RA
,
Willig
 
J
,
Zhao
 
RY
,
Kreutz
 
RP
,
Stouffer
 
GA
,
Empey
 
PE
,
Limdi
 
NA
,
Shuldiner
 
AR
,
Winterstein
 
AG
,
Johnson
 
JA
;
IGNITE Network
.
Multisite investigation of outcomes with implementation of CYP2C19 genotype-guided antiplatelet therapy after percutaneous coronary intervention
.
JACC Cardiovasc Interv
 
2018
;
11
:
181
191
.

Author notes

Emma Forton Magavern, Juan Carlos Kaski shared co-first authorship.

Munir Pirmohamed, Mark J. Caulfield shared co-last authorship.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)