16
REVIEW Controversies in cardiovascular medicine A systematic review on pharmacogenetics in cardiovascular disease: is it ready for clinical application? Jeffrey J.W. Verschuren 1 , Stella Trompet 1,2 , Judith A.M. Wessels 3 , Henk-Jan Guchelaar 3 , Moniek P.M. de Maat 4 , Maarten L. Simoons 5 , and J. Wouter Jukema 1,6,7 * 1 Department of Cardiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands; 2 Department of Geriatrics and Gerontology, Leiden University Medical Center, Leiden, The Netherlands; 3 Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands; 4 Department of Hematology, Erasmus Medical Center, Rotterdam, The Netherlands; 5 Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands; 6 Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands; and 7 The Interuniversity Cardiology Institute (ICIN), Utrecht, The Netherlands Received 16 March 2011; revised 20 June 2011; accepted 27 June 2011; online publish-ahead-of-print 30 July 2011 Pharmacogenetics is the search for heritable genetic polymorphisms that influence responses to drug therapy. The most important appli- cation of pharmacogenetics is to guide choosing agents with the greatest potential of efficacy and smallest risk of adverse drug reactions. Many studies focusing on drug–gene interactions have been published in recent years, some of which led to adaptation of FDA recommen- dations, indicating that we are on the verge of the clinical application of genetic information in drug therapy. This systematic review provides a comprehensive overview of the current knowledge on pharmacogenetics of all major drug classes currently used in the treatment of car- diovascular diseases. ----------------------------------------------------------------------------------------------------------------------------------------------------------- Keywords Pharmacogenetics Single-nucleotide polymorphism Drug therapy Drug response Cardiovascular disease Systematic review Introduction Pharmacogenetic studies search for heritable genetic polymorph- isms that influence responses to drug therapy. Pharmacogenetics has many possible applications in cardiovascular pharmacotherapy including screening for polymorphisms to choose agents with the greatest potential for efficacy and least risk of toxicity. Pharmaco- genetics also informs dose adaptations for specific drugs in patients with aberrant metabolism. Each year, an estimated 785 000 Americans suffer a myocardial infarction (MI) and 610 000 people experience a new stroke. Despite advances in management, 470 000 recurrent MIs and 185 000 recurrent strokes occur annually. 1 The numbers from Europe are not much better, indicating the importance of opti- mizing treatment strategies. 2 Although several reviews on pharmacogenetics in cardiology have been published, most focused on specific drug subgroups. This systematic review offers a comprehensive summary of the current knowledge of pharmacogenetics in cardiology, elaborates on progress towards individualized drug therapy, and incorporates conclusions made during the Colloquium ‘Pharmacogenetics of cardiovascular drugs: implications for a safer and more efficient drug therapy’ held by the Royal Netherlands Academy of Arts and Sciences in October 2010. In particular, the review considers the most relevant and well-studied polygenic markers for some pressing clinical issues: (i) statins: focusing on variability in efficacy and risk of myopathy; (ii) resistance to antiplatelet medication; (iii) dosing issues with oral anticoagulants; (iv) suboptimal responses to b-blockers; and (v) the variable response in blood pressure and * Corresponding author. Tel: +31 71 526 20 20, Fax: +31 71 526 68 85, Email: [email protected] Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2011. For permissions please email: [email protected] European Heart Journal (2012) 3, 165–175 doi:10.1093/eurheartj/ehr239 Downloaded from https://academic.oup.com/eurheartj/article-abstract/33/2/165/437170 by guest on 16 November 2018

A systematic review on pharmacogenetics in cardiovascular disease

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A systematic review on pharmacogenetics in cardiovascular disease

REVIEW

Controversies in cardiovascular medicine

A systematic review on pharmacogeneticsin cardiovascular disease: is it ready forclinical application?Jeffrey J.W. Verschuren1, Stella Trompet1,2, Judith A.M. Wessels3,Henk-Jan Guchelaar3, Moniek P.M. de Maat4, Maarten L. Simoons5,and J. Wouter Jukema1,6,7*

1Department of Cardiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands; 2Department of Geriatrics and Gerontology, Leiden UniversityMedical Center, Leiden, The Netherlands; 3Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands; 4Department ofHematology, Erasmus Medical Center, Rotterdam, The Netherlands; 5Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands; 6Durrer Center forCardiogenetic Research, Amsterdam, The Netherlands; and 7The Interuniversity Cardiology Institute (ICIN), Utrecht, The Netherlands

Received 16 March 2011; revised 20 June 2011; accepted 27 June 2011; online publish-ahead-of-print 30 July 2011

Pharmacogenetics is the search for heritable genetic polymorphisms that influence responses to drug therapy. The most important appli-cation of pharmacogenetics is to guide choosing agents with the greatest potential of efficacy and smallest risk of adverse drug reactions.Many studies focusing on drug–gene interactions have been published in recent years, some of which led to adaptation of FDA recommen-dations, indicating that we are on the verge of the clinical application of genetic information in drug therapy. This systematic review provides acomprehensive overview of the current knowledge on pharmacogenetics of all major drug classes currently used in the treatment of car-diovascular diseases.- - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -Keywords Pharmacogenetics † Single-nucleotide polymorphism † Drug therapy † Drug response † Cardiovascular

disease † Systematic review

IntroductionPharmacogenetic studies search for heritable genetic polymorph-isms that influence responses to drug therapy. Pharmacogeneticshas many possible applications in cardiovascular pharmacotherapyincluding screening for polymorphisms to choose agents with thegreatest potential for efficacy and least risk of toxicity. Pharmaco-genetics also informs dose adaptations for specific drugs inpatients with aberrant metabolism.

Each year, an estimated 785 000 Americans suffer a myocardialinfarction (MI) and 610 000 people experience a new stroke.Despite advances in management, 470 000 recurrent MIs and185 000 recurrent strokes occur annually.1 The numbers fromEurope are not much better, indicating the importance of opti-mizing treatment strategies.2

Although several reviews on pharmacogenetics in cardiologyhave been published, most focused on specific drug subgroups.This systematic review offers a comprehensive summary of thecurrent knowledge of pharmacogenetics in cardiology, elaborateson progress towards individualized drug therapy, and incorporatesconclusions made during the Colloquium ‘Pharmacogenetics ofcardiovascular drugs: implications for a safer and more efficientdrug therapy’ held by the Royal Netherlands Academy of Artsand Sciences in October 2010. In particular, the review considersthe most relevant and well-studied polygenic markers for somepressing clinical issues: (i) statins: focusing on variability in efficacyand risk of myopathy; (ii) resistance to antiplatelet medication; (iii)dosing issues with oral anticoagulants; (iv) suboptimal responses tob-blockers; and (v) the variable response in blood pressure and

* Corresponding author. Tel: +31 71 526 20 20, Fax: +31 71 526 68 85, Email: [email protected]

Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2011. For permissions please email: [email protected]

European Heart Journal (2012) 3, 165–175doi:10.1093/eurheartj/ehr239

Dow

nloaded from https://academ

ic.oup.com/eurheartj/article-abstract/33/2/165/437170 by guest on 16 N

ovember 2018

Page 2: A systematic review on pharmacogenetics in cardiovascular disease

clinical events in patients taking angiotensin-converting enzyme(ACE)-inhibitors (Table 1).

MethodsRelevant articles were identified by searching MEDLINE using the fol-lowing keywords and Medical Subject Headings (MeSH) terms: phar-macogenetics, genetic heterogeneity, genetic polymorphism, singlenucleotide polymorphism (SNP), haplotypes, treatment outcome,adverse effects, drug therapy, cardiovascular diseases (CVDs), and/orcoronary disease. We screened the title and abstract of possibly rel-evant citations and retrieved, if potentially interesting, full reports orabstracts. We checked the references of retrieved publications foradditional relevant papers and included all available literature untilMay 2011. We excluded reviews, editorials, and articles in languagesother than English.

Statins: variability in efficacy andrisk of myopathyStatins, or 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibi-tors, are widely prescribed to lower plasma cholesterol levels,thereby reducing cardiovascular risk. Despite the clinical effective-ness of statins, proven in large randomized controlled trials,3,4 inter-individual variation in response means that many patients fail toachieve adequate reduction in cholesterol levels even after multiple-dose adjustments.5 Genetic factors are thought to be partly respon-sible for this inter-individual variation. Indeed, more than 40 candi-date genes have been described with respect to the differentialeffect of statins in decreasing the risk of clinical endpoints includingcardiovascular death, MI, and lipid-lowering abilities (see Supplemen-tary material online, Table S1).

Variability in lipid loweringSeveral pharmacogenetic studies examined the gene encoding thecholesterol ester transfer protein (CETP) involved in cholesterolmetabolism and, in particular, the TaqIB variant (rs708272). Initialstudies associated the B2B2 genotype with lower CETP levels,6

higher high density lipoprotein cholesterol (HDL-C) levels, andlower risk of progression of coronary artery disease (CAD),

compared with the B1B1 genotype.7– 11 However, on statin treat-ment, B1B1 patients showed lower progression of CAD than B2B2carriers. A meta-analysis confirmed the firm association betweenTaqIB and HDL-C levels and CAD risk, while no significant inter-action between this polymorphism and pravastatin treatmentcould be demonstrated.12 This meta-analysis included studies thatenrolled patients with and without a history of CAD. In contrast,the original study included only patients with significant CAD.7 Fur-thermore, long-term results from the Regression Growth EvaluationStatin Study (REGRESS), the first study to report the possible phar-macogenetic interactions between the CETP polymorphism andstatin treatment,13 also contrast with the meta-analysis. REGRESSdemonstrated significantly higher 10-year mortality in statin-treatedmale patients carrying the B2 allele, compared with the B1B1 geno-type.14 Therefore, although untreated B2B2 patients have a lowerrisk of CAD progression, statin treatment is more beneficial inpatients with the B1B1 genotype, negating the initial advantage ofthe B2 allele in CAD.

The extensive studies of apolipoprotein E (APOE), and in particu-lar the 12/13/14 variants defined by combinations of two poly-morphisms (Cys112Arg, rs429358 and Arg158Cys, rs7412) areequivocal.15 Several studies report an association between 12 car-riers and an increased lipid-lowering response to statins orreduced cardiovascular outcomes, such as non-fatal MI andcardiovascular-related mortality, whereas other studies did notfind significant associations.16 Although the first genome-wideassociation study (GWAS) on statin effects showed a significantassociation between the three APOE polymorphisms and the low-density lipoprotein cholesterol (LDL-C) response,17 a recentmeta-analysis did not confirm this association.18 Therefore, itseems unlikely that a true association exists between APOE poly-morphisms and the lipid-lowering response to statin treatment.

Since most studies discussed so far had insufficient power todetect the small effect produced by individual genetic loci,Barber et al.19 combined three statin GWASs encompassing3936 patients. Carriers of a polymorphism (rs8014194) in thecalmin (CLMN) gene on chromosome 14 had a significantlygreater reduction of 3% in total cholesterol compared with non-carriers. The exact function of calmin is unknown, but it containsa calponin domain expected to have actin-binding activity20 andis strongly expressed by the liver and adipose tissue.21 Neverthe-less, this polymorphism explained only 1% of the variability instatin response.19 A SNP near APOE, in the APOC1 gene(rs4420638), showed moderate evidence for being associatedwith statin-induced LDL-C changes. Most other putative associ-ations appeared to be independent of statin treatment and werein loci previously associated with lipid or lipoprotein traits.19

Kinesin-like protein 6 and variability inclinical outcomeNext, carriers of the arginine-coding allele (rs20455) of kinesin-likeprotein 6 (KIF6) showed a substantially higher benefit with pravas-tatin treatment compared with non-carriers in three large clinicaltrials, which appeared to be independent from lipid- and C-reactiveprotein-lowering effects.22– 24 Kinesin-like protein 6 is a member ofthe molecular motor superfamily involved in the intracellular

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Table 1 Major issues in cardiovascular drugpharmacogenetics and the strength of the geneticevidence

Drug class Problem Evidence

Statins Variable lipid lowering MediocreRisk of myopathy Strong

Antiplateletdrugs

Resistance Strong

Anticoagulants Dosing problems Strong

b-Blockers Variable response Poor

ACE-inhibitors Variable blood pressure reduction MediocreVariable benefit to decrease risk of

eventsMediocre

J.J.W. Verschuren et al.166D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 3: A systematic review on pharmacogenetics in cardiovascular disease

transport of several important molecules, including mRNA.25 Arecent meta-analysis of 19 studies (in total 17 000 CAD casesand 39 369 controls) concluded that the SNP in question wasnot associated with the risk of CAD.26 This study was, however,unable to explore the effect of statins, since this information wasnot available for all included studies, but the probability of astrong influence on the effect of statin decreases with this publi-cation. Nevertheless, further (functional) studies are needed toexplore the role of KIF6.

Risk of myopathyGenetic variations are also associated with an altered risk ofadverse drug reactions (ADRs), especially statin-induced myopa-thy. The incidences of myopathy and rhabdomyolysis are estimatedat 11.0 and 3.4 per 100 000 person-years, respectively. The casemortality rate with rhabdomyolysis is 10%.27

The polypeptide organic anion transporter P1B1 (OATP1B1)encoded by the SLCO1B1 gene is involved in the hepatic uptakeof statins.28,29 Two common polymorphisms (521T . C,rs4149056 and 388A . G, rs2306283) influence the transporterfunction of OATP1B1,30 markedly altering statin pharmacoki-netics.31 A GWAS showed a strong association between a non-coding polymorphism (rs4363657) and statin-induced myopathyin patients treated with high-dose simvastatin.32 This polymorph-ism is in almost complete linkage disequilibrium (R2¼ 0.97) withthe 521T . C polymorphism and was expressed by more than60% of patients who developed myopathy with an odds ratio of4.7 per copy of the C allele.32 A subsequent study confirmedthis pharmacogenetic effect in simvastatin-induced myopathy,33

but not with either atorvastatin or pravastatin,33 probably indicat-ing a simvastatin specific effect.34

In conclusion, the reports on genetic variability and response tostatin treatment are difficult to interpret. Variability in studydesigns and endpoint definitions probably contribute to thesemixed results. Since statins’ full pharmacodynamic spectrum,besides their lipid-lowering properties, remains largely unknown, itis not surprising that clear cut pharmacogenetic results are lacking.It is likely that future research will further elucidate statins’ exactmechanisms of action and help explain the variability in response.Newly identified loci influencing lipid concentration identified inrecent GWAS might also prove to have pharmacogenetic associ-ations with statin treatment.35,36 Pharmacogenetic data on ADRsof statin is limited and more research dedicated to this problem isneeded before concrete advises can be given on this notion.

Resistance to antiplateletmedicationBlood platelets maintain appropriate haemostasis upon vascularinjury, but also contribute to the pathophysiology of thrombosis.Thrombosis of atherosclerotic plaques is the most importantunderlying mechanism of CAD and embolic stroke, and conse-quently antiplatelet therapy is the cornerstone of secondary pre-vention in modern CVD treatment.37– 40 Inter-patient variationsin platelet reactivity and responses to antithrombotic treatmentresult in marked differences in the benefits derived from

antiplatelet treatment. Indeed, up to 25% of patients continue toexperience new thrombotic events despite guideline therapy.41,42

The term ‘resistance’ is widely used in such cases, although a clear-cut definition is lacking.43 The current clinical definition regardsresistance as recurrent cardiovascular events despite supposedadequate therapy. Biochemical definitions which are based onresidual platelet activity during treatment measured ex vivo byseveral laboratory tests are however also clinically relevant.Trenk et al.44 for example, found a three-fold increase [95% confi-dence interval (CI) 1.4–6.8, P ¼ 0.004] in the 1-year incidence ofdeath and MI among patients with high residual platelet activityat discharge after percutaneous coronary intervention (PCI)despite clopidogrel. However, although it is likely that biochemi-cally non-responsive patients are also clinically non-responsive,biochemical ‘resistance’ is neither constant in an individual norover time.42 Furthermore, there is an evident lack of concordancebetween laboratory tests used to express resistance.45– 47

Against this background, recent research suggests that geneticvariation makes an important contribution to variation inresponses to the three main groups of anti-platelet therapy:aspirin (see Supplementary material online, Table S2); thienopyri-dine derivates [clopidogrel (see Supplementary material online,Table S3), prasugrel (see Supplementary material online,Table S4), and the P2Y12 receptor antagonist ticagrelor]; and GPIIb/IIIa receptor inhibitors (orbofiban and abciximab) (see Sup-plementary material online, Table S5).

AspirinThe antiplatelet effects of aspirin (acetylsalicylic acid) arise fromirreversible acetylation of cyclooxygenase (COX)-1, which cata-lyses the formation of thromboxane A2, prostaglandin, andrelated metabolites from arachidonic acid.48,49 Blocking COX-1decreases the formation of thromboxane A2, thereby reducingplatelet aggregation.50 Aspirin resistance is poorly defined: apatient can be a non-responder in one test and a normal respon-der in another. Because of this lack of uniformity, the reportedprevalence of aspirin resistance differs widely between studies. Arecent systematic review reported a mean prevalence of aspirinresistance of 24% with a range between 0 and 57%.51

Conflicting results on aspirin efficacyOf the many pharmacogenetic studies on aspirin, Halushka et al.49

were the first to describe an effect of two polymorphisms in theCOX-1 gene, 2842A . G (rs10306114) and 50C . T(rs3842787), that are in complete linkage disequilibrium. Healthyindividuals heterozygous for these polymorphisms showedgreater inhibition of platelet aggregation compared with homozy-gotes of the wild-type allele. Two later studies confirmed thisrelationship in CAD patients.52,53 However, many other studiesfailed to find significant associations between variation of theCOX-1 gene and residual platelet activity54,55 or clinical events,including non-fatal MI or cardiac death.56

Platelet glycoprotein (GP) IIb/IIIa receptors bind fibrinogen,thereby cross-linking platelets and von Willebrand factor(vWF).57 Studies that focused on the 1565T . C polymorphism(rs5918; PlA1/A2) of the GP IIIa (integrin b3) gene producedhighly variable conclusions (see Supplementary material online,

Overview of pharmacogenetics in CVD 167D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 4: A systematic review on pharmacogenetics in cardiovascular disease

Table S2). A meta-analysis that included most of these studies con-cluded that the PlA1/A2 variant was significantly associated withaspirin resistance, but only in healthy individuals.45 Co-medicationsused by CVD patients potentially obscured aspirin resistance, whiledifferences in laboratory techniques may have also influencedoutcome.

The meta-analysis also revealed that variations of four othergenes [GP Ia (807C . T, rs1126643), COX-1 (2842A . G/50C. T), P2Y12 (H1/H2, constituted by rs10935838, rs2046934,rs5853517, and rs6809699), and P2Y1 (A1622G, rs701265)] werenot associated with aspirin resistance.45 However, a later relativelylarge study, including 200 high-risk atherosclerosis patients, found astrong association between aspirin resistance and the T allele ofthe 807C . T polymorphism of the GP Ia gene.58 A furtherstudy ruled out any influence of the 2842A . G/50C . T poly-morphism of COX-1 and PlA1/A2 of GP IIIa on platelet responsein a healthy Chinese population, in which these sites were non-polymorphic. However, the study showed attenuated antiplateleteffect during aspirin treatment in the presence of the P2Y1

893CC genotype (rs1065776).55 This last association contrastswith results from 469 Caucasian patients with a history of MI.Here, carriers of the 893T allele had an almost three-fold increasedrisk of aspirin resistance compared with the CC genotype.59

Complications of aspirin therapyA small number of studies focused on genes involved inaspirin-related ADRs. Park et al.60,61 reported that the promoterpolymorphisms 2863C . A (rs1800630) and 21031T . C (rs1799964) of TNF-a and 2509C . T (rs1800469) of TGF-b1 may con-tribute to aspirin-induced urticaria. Piazuelo et al.62 related the car-riage of the A allele of the 27 bp variable number tandem repeats ofeNOS with a decreased risk of upper GI bleeding. Further pharmaco-genetic reports on ADRs of aspirin are necessary given the wide-spread prescription of aspirin.

In conclusion, the precise role of genetic variation in the varia-bility of response to aspirin treatment has yet to be defined. Theonly consistent association is that of the PlA2 variant of the GPIIIa gene and aspirin resistance in healthy individuals. Standardiz-ation of techniques to measure and define response is a prerequi-site for future studies and to establish genetic variation’scontribution to aspirin resistance.

Thienopyridine derivates/P2Y12 receptorinhibitorsClopidogrel resistance is common: the incidence was 21% (95% CI17–25%) in a meta-analysis of patients after PCI.42 Estimates of theincidence vary widely between studies, mainly due to differences inthe time of measurement and dose of clopidogrel. However, thetechnique used to assess clopidogrel responses did not result ina different prevalence of clopidogrel resistance between studies.42

Clopidogrel is an orally administered prodrug. Intestinal absorp-tion is limited by P-GP, an efflux pump encoded by the ABCB1gene. Most of the bioavailable fraction undergoes hydrolysis toan inactive metabolite. Approximately 15% is oxidized by thehepatic cytochrome (CYP) 450 system in two sequential steps togenerate an active metabolite,63 which binds to, and irreversiblyinhibits, the platelet ADP receptor P2Y12. By blocking this

receptor, clopidogrel prevents platelet degranulation and inhibitsconversion of the GP IIb/IIIa receptor to the form that binds fibro-gen and links platelets, thereby inhibiting platelet aggregation.64 –67

Metabolism and resistanceSeveral studies examined the influence of genetic variation in oneor more enzymes of the CYP450 system on clopidogrel resistance.The most consistent finding is that the *2 allele of the CYP2C19gene (rs4244285) (involved in both hepatic oxidative steps) leadsto the formation of an inactive enzyme and, therefore, decreasedplatelet responsiveness to clopidogrel68 (Figure 1). After the firstreport,68 which enrolled healthy male volunteers, studies repli-cated the reduced platelet responsiveness in vitro and establishedan increased risk of cardiovascular events and death during clopi-dogrel treatment in different patient populations (see Supplemen-tary material online, Table S3). Other loss-of-function alleles(*3, rs4986893; *4, rs28399504; and *5, rs56337013) have beenassociated with clopidogrel resistance although not as stronglyand as consistently as the *2 allele, probably reflecting theirlower allele frequencies.69 –72 Furthermore, recent studies associ-ated a gain-of-function allele (*17, rs1248560) and a higher plateletinhibition by clopidogrel and, as a consequence, increased bleedingrisk.73,74

The CYP2C9 loss-of-function allele *3 (rs1057910) has beenassociated with increased residual platelet activity and poorresponder status after a 300 mg clopidogrel loading dosage inhealthy subjects69 and CVD patients.75 Surprisingly, the latterstudy could not detect a similar effect of the risk allele during clo-pidogrel maintenance therapy. The authors hypothesized that sincethis enzyme is involved in the second metabolization step, thereduced enzyme capacity of the *3 allele becomes critical onlywith higher clopidogrel plasma concentrations.63

Studies on genetic variation in other involved CYP450 enzymes,especially CYP3A4 and CYP3A5, did not show a significant influ-ence on platelet responsiveness after clopidogrel treatment (seeSupplementary material online, Table S3). From a clinical point ofview, an important finding came from Gladding et al.,76 whodemonstrated that increasing the clopidogrel dose in patientswith non-responder CYP2C19 polymorphisms can increase theantiplatelet response, indicating that personalizing doses usingpharmacogenetics could potentially optimize treatment.

Drug transport and receptorsAlthough CYP2C19 polymorphisms probably account for most ofthe variability in clopidogrel response, variations in the functionof intestinal efflux transporters might also influence clopidogrelbioavailability. For instance, the 3435C . T SNP of ABCB1(rs1045642) was shown to influence P-glycoprotein transporterexpression, resulting in two- to four-fold lower peak concen-trations of clopidogrel and its active metabolite in TT carriersafter oral administration.77 Two recent studies confirmed thesefindings.70,78 Mega et al.78 also reported that the increased risk ofevents of 3435TT homozygotes increased further in combinationwith a CYP2C19 loss-of-function allele. However, not all publishedpapers show consistent results75 and a recent GWAS could notreproduce the association between this polymorphism and clopi-dogrel pharmacodynamics in healthy volunteers.79

J.J.W. Verschuren et al.168D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 5: A systematic review on pharmacogenetics in cardiovascular disease

Another candidate gene encodes the P2Y12 receptor, which isessential for platelet inhibition exerted by clopidogrel, prasugrel,and ticagrelor.80–82 Ziegler et al.83 reported a four-fold increasedrisk for cerebrovascular events in patients who carried the34C . T (rs6809699) variant of the P2Y12 receptor receiving clo-pidogrel for peripheral artery disease. Moreover, Staritz et al.84

associated another polymorphism (52G . T, rs6785930) with clo-pidogrel resistance in patients receiving dual antiplatelet therapy(clopidogrel and aspirin) after PCI. However, other studies couldnot confirm these findings with respect to platelet response orcardiovascular events.70,85,86

Finally, T allele carriers of the GP Ia gene 807C . T (rs1126643)polymorphism show increased platelet aggregation during clopido-grel treatment than non-carriers, increasing their thromboticrisk.87 The same group confirmed these results in patients

receiving dual antiplatelet therapy.88 However, another studycould not confirm these results in 600 acute coronary syndrome(ACS) patients on dual antiplatelet therapy.89 The PlA polymorph-ism of the platelet GP IIIa gene (rs5918) probably does not influ-ence the effect of thienopyridine derivates, although smallstudies report positive as well as negative effects.90,91 Simonet al.70 did not find a significant association of the PlA polymorph-ism in a population consisting of 2208 patients with acute MItreated with clopidogrel and the rate of cardiovascular events(225 deaths and 94 non-fatal MIs or strokes).

In conclusion, resistance to clopidogrel is a major problem, andprediction of the problem could be of great value. The CYP2C19loss-of-function allele *2 shows the most promise in predicting clo-pidogrel resistance and subsequent treatment failure, although thiswas not demonstrated by all studies.92 Many other polymorphisms

Figure 1 Clopidogrel metabolism. From Sangkuhl et al.179 Used with permission of PharmGKB and Stanford University.

Overview of pharmacogenetics in CVD 169D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 6: A systematic review on pharmacogenetics in cardiovascular disease

in theoretically promising target genes do not produce consistentresults. Furthermore, proton pump inhibitors, especially omepra-zol, show high affinity for CYP2C19 and might inhibit clopidogrelmetabolism, raising concerns about concomitant use.93 Clinical evi-dence of this adverse drug–drug interaction, however, derivesmainly from observational studies. A recent review concludedthat although definite evidence is lacking and further studies areneeded, clinicians should keep in mind this possibly clinically rel-evant interaction when considering prescribing this combination.93

PrasugrelThe pharmacogenetics of prasugrel, a newer thienopyridine, hasnot been as comprehensively investigated as clopidogrel. Never-theless, several studies indicate that the main contributor to clopi-dogrel resistance, CYP2C19*2, does not affect prasugrel’sefficacy.69,94 In a large study, no significant attenuation of the pra-sugrel response was found in carriers of at least one of the knownloss-of-function alleles of the tested CYP450 genes. Furthermore,in ACS patients, none of the genotypes was associated with clinicalcardiovascular events.94 Prasugrel’s metabolic pathway explainsthis lack of association; CYP2C19 makes only a minor contributionto prasugrel’s metabolism. CYP3A4 and CYP2B6 are largelyresponsible for converting prasugrel to its active form.94,95

CYP3A4 appears not to be very polymorphic and no clear associ-ation emerged between CYP2B6 variants and prasugrel function.94

Prasugrel’s strong antiplatelet effect leads to a highly reducedresidual platelet activity, making any effect of genetic variation dif-ficult to determine. Prasugrel inhibits platelets sufficiently toreduce clinical endpoints despite decreased efficacy arising fromthe polymorphisms.

TicagrelorThe newest P2Y12 receptor antagonist, ticagrelor, appears to besuperior to clopidogrel80,82 and has similar potency as prasugrel.81

A genetic substudy of the largest ticagrelor trial to date, thePLATelet inhibition and patient Outcomes (PLATO) trial, geno-typed 10 285 patients randomized to ticagrelor or clopidogreltreatment. No interaction between ticagrelor and anyloss-of-function CYP2C19 allele (*2 till *8), gain-of-functionCYP2C19*17 allele, or ABCB1 3435C . T genotype emergedwith regard to the either primary efficacy endpoint (compositeof cardiovascular death, MI, or stroke) or any type of major bleed-ing after up to 12 months treatment.74

Glycoprotein IIb/IIIa antagonistsGlycoprotein IIb/IIIa antagonists are mechanistically different fromaspirin, thienopyridine derivates, and other P2Y12 inhibitors. Glyco-protein IIb/IIIa antagonists act primarily outside the platelet bycompetitively inhibiting the receptor essential for platelet bridgingand aggregation.96 Several studies have focused on polymorphismsin the GP IIIa gene and antiplatelet effect, but the results appearinconclusive. One study on orbofiban97 and two on abciximab98,99

found a relationship between the PlA2 allele with reduced plateletinhibition and increased cardiovascular event rate. However, otherstudies did not replicate this association.100 –102 In addition, Shieldset al. developed a model to predict recurrent events during orbo-fiban treatment that included 10 candidate genes: platelet

receptors GP IIIa, PlA1/A2 (rs5918), GP Ia 807C . T(rs1126643), GP Iba, 25C . T (rs2243093); platelet ligandsb-fibrinogen, 2455G . A (rs1800790) and vWF, 21051G . A;interleukins IL-1RN*2, and IL-6, 2174G . C (rs1800795);adhesion proteins E-selectin, 128A . C (rs5361) and P-selectin,715A . C; and metalloproteinase MMP-9, 21562C . T. In 924ACS patients, an association emerged between a combination ofgenetic variants and both recurrent MIs and bleeding outcomesduring orbofiban treatment.103 Since the influence of individualgenetic polymorphisms is usually small, combining multiple variantsin a model might be useful. However, interpreting the results maybe more difficult.

Dosing issues with oralanticoagulantsCoumarin anticoagulants (vitamin K antagonists) are widely used totreat and prevent venous and arterial thrombo-embolisms.However, anticoagulants’ narrow therapeutic range necessitatesfrequent monitoring of the international normalized ratio (INR)coagulation status, which is expensive for health-care systemsand inconvenient for patients. The large individual variability indrug response with all three of the most frequently prescribedanticoagulants (warfarin, acenocoumarol, and phenprocoumon)complicates therapy and makes defining a standard fixed doseunachievable. Besides several patient-related (amongst othersage, weight, body surface area, and diet) and clinical factors(hepatic or renal disease, anaemia, co-medication), pharmacoge-netics explains a large part of the variability in dose require-ments104 – 106 (see Supplementary material online, Table S6).

Increased metabolismFuruya et al.107 first reported the influence of polymorphisms inCYP2C9, the primary enzyme involved in warfarin metabolism,on dose requirement in vivo. Carriers of the *2 allele(rs1799853) required a 20% lower warfarin dose to maintain atarget INR between 2 and 4. Later studies associated the *3(rs1057910) allele with an increased sensitivity to warfarin.108 Invitro, the *2 and *3 genotypes decrease CYP2C9 activity by 30and 80%, respectively.109,110 A large meta-analysis of 39 studiesencompassing 7907 patients111 concluded that compared withthe wild-type CYP2C9*1/*1 genotype, all variant genotypes(*1/*2, *1/*3, *2/*2, *2/*3, and *3/*3) required lower maintenancedoses of warfarin. For instance, maintenance doses in the *2 and *3homozygotes were 36 and 78.1% lower, respectively.

However, using this pharmacogenetic knowledge in clinical prac-tice is difficult. A meta-analysis investigating the safety and efficacyof genotype-guided dosing of warfarin in reducing the occurrenceof major bleeding events and over-anticoagulation found a non-significant trend towards more rapid achievement of a stabledose in the genotype-guided dosing arm.112 Several ongoing ran-domized controlled trials, which are expected to encompassmore than 2500 patients, will help elucidate the role of genetictesting in warfarin management.112

J.J.W. Verschuren et al.170D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 7: A systematic review on pharmacogenetics in cardiovascular disease

A second contributorAnother extensively studied gene is that encoding VKORC1(vitamin K epoxide reductase complex subunit 1), which convertsthe oxidized, inactive form of vitamin K to the reduced, activeform. Coumarins inhibit VKORC1, thereby inhibiting activation ofthe vitamin K-dependent clotting factors II, VII, IX, and X.113

A recent meta-analysis analysed the relationship between themean maintenance dose of warfarin and three VKORC1 polymorph-isms (1173T . C, rs9934438; 3730G . A, rs7294; and21639A . G, rs9923231).114 Carriers of 1173T . C and21639A . G needed higher maintenance warfarin doses.

Genetic variation in several other genes—including APOE, gluta-myl carboxylase (GGCX), calumenin (CALU), CYP4F2, epoxidehydrolase 1 (EPHX1), and factor VII (F7)—may also influence war-farin dosage requirements. However, although some studies foundsignificant associations, results were inconsistent.105,115 –119 Finally,recent GWASs identified only CYP2C9 and VKORC1 as major hot-spots, indicating that polymorphisms in other genes probably onlyhave minor influence on warfarin dosing.116,120

Although warfarin is usually the anticoagulant of choice, aceno-coumarol and phenprocoumon are used in most Europeancountries.121 All three drugs are metabolized by CYP2C9.However, phenprocoumon appears to be less influenced byCYP2C9 polymorphisms122 due to significant metabolism by thenon-polymorphic CYP3A4.121 The effect of the VKORC1 poly-morphism is similar for all three drugs.121,123,124

Dosing advice with genetic guidanceIn conclusion, the association between warfarin dose and poly-morphisms of the CYP2C9 and VKORC1 genes seems unquestion-able, as demonstrated by numerous studies and severalmeta-analyses. Polymorphisms in CYP2C9 and VKORC1 togetherwith additional clinical factors including co-medication accountfor 50–60% of the inter-individual warfarin variability.105,125 – 127

Based on this evidence, a free web site is available to help cliniciansestimate the therapeutic dose in patients new to warfarin (http://www.warfarindosing.org) (Figure 2). Furthermore, the US Foodand Drug Administration (FDA) now recommends consideringlower initiation doses of warfarin for patients with certain poly-morphisms in CYP2C9 and VKORC1.128

Suboptimal responses tob-blockersb-Blockers competitively antagonize b-adrenergic receptors(b-AR). Chronic b1-AR activation contributes to the pathogenesisof heart failure and b-AR blockade improves survival, remodelling,and left ventricular ejection fraction (LVEF).129 –131 Other majorindications for b-blocker therapy are hypertension, angina pec-toris, and MI. As with most other drugs, not all patients benefitfrom b-blockers to the same extent, possibly due to genetic vari-ation (see Supplementary material online, Table S7).

Receptors and treatment efficacyThe primary focus of studies examining pharmacogenetic determi-nants of b-blocker response has been the b-AR genes and two

polymorphisms—Ser49Gly (rs1801252) and Arg389Gly (rs1801253)—in the b1-adrenergic receptor (ADRB1) gene in particular.Ser49Gly and Arg389Gly are in linkage disequilibrium and 65% ofthe population carry the Ser49/Arg389 haplotype.132 Not allstudies reported consistent results, but a meta-analysis combiningthree studies (encompassing 504 heart failure patients) concludedthat Arg389 homozygote individuals showed significantly greaterimprovement of LVEF when treated with b-blockers, comparedwith Gly389 carriers.132 Other more recent studies drew thesame conclusion.133,134 However, two large studies reported incon-sistent results on mortality and heart failure-related hospitalization.In the BEST study, the combination of Arg389 homozygote genotypeand bucindolol treatment was associated with decreased mortalityand hospitalization.135 However, no association emerged inMERIT-HT.136 Furthermore, several,137– 139 but not all,140 – 142

studies associated Arg389 homozygotes with an increased effectof b-blockers in decreasing systolic and diastolic blood pressure orheart rate. Furthermore, the Gly allele of Ser49Gly was more com-monly associated with a favourable outcome or improved responseto b-blockers.143 –145 However, many studies did not find anyassociation.142,146,147

Most studies assessing two common polymorphisms in ARDB2(Gly16Arg, rs104213 and Gln27Glu, rs1042714) did not observeany association.146,147 However, two small studies associated theGlu27 allele with increased LVEF after b-blocker treatment com-pared with the Gln27 allele.148,149 A third polymorphism inARDB2, Thr164Ile (rs1800888), occurs in only 1% of Cauca-sians.134,146 Although the Ile164 variant did not have a majorimpact on outcome in a population of 443 heart failure patients,multivariate analysis suggested a possible negative impact ofb-blocker therapy on survival in heterozygous subjects.150

Dosing problems of b-blockersMany b-blockers are metabolized predominately by hepaticCYP2D6,151 which is highly polymorphic with many alleles having adecreased or absent function.152 Several studies report thatCYP2D6 genotype profoundly affects b-blocker concentrations andthe effect seems especially pronounced with metoprolol.153–156

Poor metabolizing genotypes increase metoprolol plasma concen-trations, which results in greater reductions in heart rate and bloodpressure,157–159 and an increased risk of ADRs.159 Although the evi-dence is derived from a few studies, dose adjustments should be con-sidered in patients vulnerable to complications, such as heart failurepatients.160

The use of different b-blockers between, and even within,studies complicates the evaluation of the evidence on the pharma-cogenetics of b-blocker therapy. Differences in receptor specificityof the individual b-blockers could explain this heterogeneity. Forinstance, metoprolol and bisoprolol are b1-AR selective, while car-vedilol and bucindolol are non-selective.161 In summary, the mostconvincing evidence suggests that the variable benefit fromb-blocker treatment is influenced by the Arg389Gly polymorphismof the ADRB1 gene. Furthermore, indisputable evidence shows thatpolymorphisms of the CYP2D6 gene influence b-blocker metab-olism and several reports indicate that poor metabolizers haveincreased efficacy and a greater risk of side effects. The data onother possible related polymorphisms is too thin to draw definite

Overview of pharmacogenetics in CVD 171D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 8: A systematic review on pharmacogenetics in cardiovascular disease

conclusions. Combining different risk alleles could provide moreanswers.162

The variable response in patientstaking angiotensin-convertingenzyme-inhibitors

The angiotensin-converting enzyme geneinsertion/deletion polymorphismMost studies examining genetic factors influencing responses toACE-inhibitor treatment evaluated the ACE gene insertion/deletion(I/D) polymorphism (rs4646994) (see Supplementary materialonline, Table S8), which correlates strongly with ACE plasma con-centrations.163,164 Nevertheless, initial results were inconsistent,partly due to limited sample size. Recently, however, the Geneticsof Hypertension-Associated Treatment (GenHAT) study did notfind an association between the I/D ACE polymorphism and the6-year non-fatal cardiac events.165 Similarly, the Perindopril Protec-tion Against Recurrent Stroke Study (PROGRESS) did not find anassociation between this polymorphism and 4-year risk of cardiacevents, mortality, neurological events, or decline or with bloodpressure reduction during perindopril treatment.166 In contrast,in the population-based Rotterdam Study, hypertensive patientson ACE-inhibitors had an increased 10-year mortality risk when

carrying the DD genotype compared with the II genotype, withthe ID genotype in an intermediate position.167 However, the Rot-terdam Study’s observational nature represents a limitation.

In conclusion, although the ACE gene I/D polymorphism was aprime candidate, an association with therapeutic response seemsunlikely. Indeed, since Cambien et al.168 classified this poly-morphism as a potent risk factor for coronary heart disease,many predominantly small positive trials reported similar obser-vations. But a meta-analysis, examining the ACE I/D polymorph-ism with regard to restenosis, showed that small positive studieswere much more likely to be published than negative ones. Thispublication bias distorted the actual association.169

Other targets in therenin–angiotensin–aldosterone systemAngiotensinogen (AGT) is another candidate gene in pharmaco-genetic research on ACE-inhibitors. In another cohort of theRotterdam study, 4097 subjects were analysed for theMet235Thr (rs699) polymorphism of AGT.170 The Thr alleleincreased the risk of MI and stroke in ACE-inhibitor users. Nosignificant association was found with b-blockers. The sameauthors published a paper on a different cohort of 2216 subjectsfrom the Rotterdam study investigating whether the Met235ThrAGT polymorphism modified the risk of atherosclerosis associ-ated with b-blocker or ACE-inhibitor therapy.171 They also

Figure 2 Dosing algorithm warfarin. The flow chart illustrates the publically available warfarin dosing algorithm (http://www.warfarindosing.org). This algorithm incorporates patient factors, clinical parameters, and genetic information to provide an estimation of the loading dose aswell as the maintenance dose of warfarin in patients new to this drug. Clinical testing of the algorithm is currently ongoing (GIFT trial,NCT01006733).�, lower warfarin dose; �, higher warfarin dose. aHigher dose with higher body surface area. bSevere liver disease may pre-dispose to an elevated international normalized ratio. cConcomitant statin use (except pravastatin) decreases warfarin maintenance dose.

J.J.W. Verschuren et al.172D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 9: A systematic review on pharmacogenetics in cardiovascular disease

examined the possible relation with the I/D ACE polymorphismand the angiotensin II receptor type 1 (AGTR1) 573C . T(rs5182) polymorphism. None of the candidate polymorphismsstrongly modified the risk of atherosclerosis, measured by arter-ial disease, carotid atherosclerosis, and aortic atherosclerosis inhypertensives taking a b-blocker or ACE-inhibitor.

Furthermore, Su et al.172 genotyped a Chinese population of1447 hypertensive patients from a benazepril trial for several poly-morphisms in AGT, AGTR1, and AGTR2. No association with bloodpressure response emerged for the Met235Thr AGT polymorph-ism. However, subjects with the AA genotype of the rs7079 poly-morphism of the AGT gene showed an enhanced antihypertensiveeffect. Although mortality risk seems to be higher among 235Thrallele carriers of the AGT gene receiving an ACE-inhibitor-basedregimen,170 the risk of atherosclerosis171 or blood pressurereduction172 does not seem to be increased. Further studies exam-ining the influence of this AGT polymorphism might better charac-terize this relationship. Moreover, common haplotypes composedof seven polymorphisms in the AGTR1 gene were also associatedwith a greater antihypertensive effect on ACE-inhibitor therapy.

Prediction of benefitFinally, the first results of the Perindopril Genetic Associationstudy (PERGENE) were recently published.173 PERGENE included8907 subjects and was designed to assess the feasibility of pharma-cogenetic profiling of treatment benefit of ACE-inhibitors inpatients with stable CAD. PERGENE investigated 52 haplotype-tagging polymorphisms in 12 candidate genes within the pharmaco-dynamic pathway of ACE-inhibitors and their relation withcardiovascular mortality, non-fatal MI, and resuscitated cardiacarrest during 4.2 years of follow-up. Two polymorphisms locatedin the AGTR1 gene (rs275651 and rs5182) and one in the bradyki-nin type I receptor (rs12050217) gene were significantly associatedwith perindopril’s treatment benefit. Combining these three poly-morphisms in a pharmacogenetic score identified patients who didnot benefit from perindopril (26.5%) (Figure 3). The event rateincreased significantly in these patients allocated to perindopril(6.3–10.4%, Pinteraction , 0.0001). The authors replicated this phar-macogenetic score in a subgroup of the PROGRESS trial.173 Fivepolymorphisms of the ACE gene (one in complete linkage disequi-librium with the I/D polymorphism) were shown not to be

Figure 3 Pharmacogenetic score proposed by the PERGENE study for prediction outcome of treatment with angiotensin-convertingenzyme-inhibitors. Results of the PERGENE study indicated that carriage of three or more minor alleles (risk alleles) of rs275651 A . T,rs5182 C . T, and rs12050217 A . G corresponded with an increased risk of cardiovascular mortality, myocardial infarction, or resuscitatedcardiac arrest during treatment with perindopril compared with placebo. Clinical consequences of these findings, whether indeed patients withthis specific risk profile should not receive angiotensin-converting enzyme-inhibitor treatment, remain to be prospectively studied. Modifiedfrom Brugts et al.173

Overview of pharmacogenetics in CVD 173D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 10: A systematic review on pharmacogenetics in cardiovascular disease

associated with the endpoints, again stressing that the ACE I/D genevariation does not seems to carry clinical value. The PERGENE trialtakes an important step closer to realizing individualized therapyfor ACE-inhibitors, but requires replication.

Angiotensin receptor blockers (ARB) inhibit therenin–angiotensin–aldosterone system through a different mech-anism than ACE-inhibitors. Although some small studies investi-gated pharmacogenetics of ARBs, replication in largerpopulations is necessary before drawing conclusions (see Sup-plementary material online, Table S8).

Future perspectives andconclusionDuring the Colloquium, mentioned in the introduction, all abovediscussed subjects were covered and the following conclusionswere made. Elucidating genetic variation in patients with CVDmay enable tailored therapy, thereby optimizing efficacy whileminimizing costs and adverse effects. Nevertheless, despite thevast amount of available publications, pharmacogenetics of CVDis not yet established in clinical practice. Indeed, studies of mostpolymorphisms produced inconsistent results due to thecomplex systems underlying CVD, which means variation in asingle gene usually produces only a small effect on clinical pheno-types. Very large patient cohorts are needed to convincingly detectthese subtle differences. Large studies designed to detect pharma-cogenetic association (e.g. large GWAS) are ongoing and will prob-ably be of great value in unravelling the current inconsistencies aswell as elucidating associations of genetic variation in other targetgenes. Moreover, new approaches like sequencing might result innew targets for subsequent research.

The relevance and usefulness of GWAS have recently beenreviewed by Daly174 (Table 2) and commercially available arrays,like the Drug Metabolizing Enzymes and Transporters (DMET)

platform, offer systematic exploration of potentially relevant path-ways.175 However, pharmacogenetics needs evidence other thanGWAS to convince cardiologists of its clinical utility. In particular,large prospective studies should investigate the effect of genetictesting on clinical outcomes and stratify patients to determinegroups for which genetic testing is relevant. For example, theJUPITER trial showed that statin treatment is more beneficial insubjects with high C-reactive protein levels.176 Development ofrisk models combining clinical characteristics (age, gender, andhistory), patient characteristics (for instance blood pressure,weight, and echocardiographic parameters), biomarkers, and gen-etics will be most useful and informative.

Besides the genetic variation depicted in Table 3, the evidence formost other pharmacogenetic associations is not strong enough forclinical application. Nevertheless, currently several guidelines ondrug prescription already contain pharmacogenetic information.Very recently, Swen et al.177 published an update of the currentDutch guidelines implementing pharmacogenetics in several thera-peutic recommendations. Also, over 60 FDA-approved drug labelscontain pharmacogenetic information.178 For instance, in 2007, theFDA recommended initiation warfarin doses for carriers of certainCYP2C9 and VKORC1 polymorphisms. Moreover, in 2009, the FDAadded a warning to the clopidogrel prescribing information to high-light the impact of the poor metabolizer genotypes of CYP2C19.Finally, the pharmacogenetic score developed in the PERGENEtrial seems to be able to identify patients who will not benefit fromACE-inhibitor treatment and the clinical consequences of thisknowledge, whether indeed patients with this specific risk profileshould not receive ACE-inhibitor treatment, should be prospectivelytested.

Ongoing large clinical trials, primarily designed as pharmacoge-netic studies, and GWAS with large study populations promiseto provide more convincing evidence. Overall, there are enoughreasons to remain optimistic that, even though we are taking

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Table 2 GWAS on pharmacogenetics of cardiovascular drugs

Study name Drug Outcome measure Population size (disc./repl.)

LowestP-value

Associated gene(s) Ref.

TNT Atorvastatin Lipid-loweringresponse

1984/3761 5.5E230 APOE, PCSK9, HMGCR 17

TNT, CAP, andPRINCE

Statin Lipid-loweringresponse

3936a 1.9E208 CLMN, APOC1 19

SEARCH and HPS Simvastatin Myopathy 85|90b/21|16 643b 4.0E209 SLCO1B1 32

PAPI Clopidogrel Platelet aggregation 429/227 1.5E213 CYP2C19 79

— Warfarin Maintenance dose 181/374 2.6E213 VKORC1, CYP2C9 120

WARG Warfarin Maintenance dose 1053/588 5.4E278 VKORC1, CYP2C9,CYP4F2

116

Rotterdam Study Acenocoumarol Maintenance dose 1451/287 2.0E2123 STX4A (VKORC1),CYP2C9

124

TNT, Treating to New Targets study; CAP, Cholesterol And Pharmacogenetics study; PRINCE, PRavastatin/Inflammation CRP Evaluation study; SEARCH, Study of theEffectiveness of Additional Reductions in Cholesterol and Homocysteine; HPS, Heart Protection Study; PAPI, the Amish Pharmacogenomics of Anti-Platelet Intervention study;WARG, WARfarin Genetics study; disc., discovery cohort; repl., replication cohort.aCombined number of patients included in the meta-analysis.bCase|control.

J.J.W. Verschuren et al.174D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 11: A systematic review on pharmacogenetics in cardiovascular disease

small steps at a time, we are heading to an era where we can finallyuse pharmacogenetics in clinical practice to optimize treatment forthe individual patient.

Supplementary materialSupplementary material is available at European Heart Journalonline.

AcknowledgmentsOur thanks goes out to the speakers during the ‘Pharmacogeneticsof cardiovascular drugs Academy Colloquium’ of the RoyalNetherlands Academy of Arts and Sciences for their contributionto the symposium: A. Isaacs, Professor A. Uitterlinden, ProfessorA. Zwinderman, Professor M.L. Simoons, Professor H.-J.Guchelaar, B.R. Winkelmann, A. Briggs, R. van Schaik,M. Ingelman-Sundberg, Professor J.W. Jukema, A.-H. Maitland v/d.Zee, Professor Schaefer, D. Voora, G. Rudez, J. Oldenburg,P. Sharma, G. Pare, P. v/d. Harst, J.J. Brugts, Professor W. vanGilst, C.R. Bezzina, and Professor J. Danser.

FundingThe research leading to these results has received funding from theEuropean Union’s Seventh Framework Programme (FP7/2007-2013)under grant agreement (no. HEALTH-F2-2009-223004). Supportedby grants from the Interuniversity Cardiology Institute of theNetherlands (ICIN) and the Durrer Center for CardiogeneticResearch both Institutes of the Netherlands Royal Academy of Artsand Sciences (KNAW), the Netherlands Heart Foundation, theCenter for Medical Systems Biology (CMSB), a centre of excellenceapproved by the Netherlands Genomics Initiative/Netherlands Organ-ization for Scientific Research (NWO), and the Netherlands Consor-tium for Healthy Ageing (NCHA). J.W. Jukema is an established clinicalinvestigator of the Netherlands Heart Foundation (2001D032). Thefunders had no role in study design, data collection and analysis,decision to publish or the preparation of the manuscript.

Conflict of interest: none declared.

References1. Lloyd-Jones D, Adams RJ, Brown TM, Carnethon M, Dai S, De Simone G,

Ferguson TB, Ford E, Furie K, Gillespie C, Go A, Greenlund K, Haase N,Hailpern S, Ho PM, Howard V, Kissela B, Kittner S, Lackland D, Lisabeth L,Marelli A, McDermott MM, Meigs J, Mozaffarian D, Mussolino M, Nichol G,Roger VL, Rosamond W, Sacco R, Sorlie P, Roger VL, Thom T,Wasserthiel-Smoller S, Wong ND, Wylie-Rosett J. Heart disease and strokestatistics—2010 update: a report from the American Heart Association. Circula-tion 2010;121:e46–e215.

2. Vaartjes I, van Dis I, Visseren FLJ, Bots ML. Hart- en vaatziekten in Nederland2010, cijfers over leefstijl- en risicofactoren, ziekte en sterfte. The Netherlands:Mouthaan Grafisch Bedrijf; 2010.

3. Baigent C, Keech A, Kearney PM, Blackwell L, Buck G, Pollicino C, Kirby A,Sourjina T, Peto R, Collins R, Simes R. Efficacy and safety of cholesterol-loweringtreatment: prospective meta-analysis of data from 90,056 participants in 14 ran-domised trials of statins. Lancet 2005;366:1267–1278.

4. LaRosa JC, He J, Vupputuri S. Effect of statins on risk of coronary disease: ameta-analysis of randomized controlled trials. JAMA 1999;282:2340–2346.

5. Mangravite LM, Thorn CF, Krauss RM. Clinical implications of pharmacoge-nomics of statin treatment. Pharmacogenomics J 2006;6:360–374.

6. Kuivenhoven JA, De Knijff P, Boer JM, Smalheer HA, Botma GJ, Seidell JC,Kastelein JJ, Pritchard PH. Heterogeneity at the CETP gene locus. Influence onplasma CETP concentrations and HDL cholesterol levels. Arterioscler ThrombVasc Biol 1997;17:560–568.

7. Kuivenhoven JA, Jukema JW, Zwinderman AH, De Knijff P, McPherson R,Bruschke AV, Lie KI, Kastelein JJ. The role of a common variant of the cholesterylester transfer protein gene in the progression of coronary atherosclerosis. TheRegression Growth Evaluation Statin Study Group. N Engl J Med 1998;338:86–93.

8. Freeman DJ, Samani NJ, Wilson V, McMahon AD, Braund PS, Cheng S,Caslake MJ, Packard CJ, Gaffney D. A polymorphism of the cholesteryl estertransfer protein gene predicts cardiovascular events in non-smokers in theWest of Scotland Coronary Prevention Study. Eur Heart J 2003;24:1833–1842.

9. Ordovas JM, Cupples LA, Corella D, Otvos JD, Osgood D, Martinez A, Lahoz C,Coltell O, Wilson PWF, Schaefer EJ. Association of cholesteryl ester transferprotein-TaqIB polymorphism with variations in lipoprotein subclasses and cor-onary heart disease risk: the Framingham study. Arterioscler Thromb Vasc Biol2000;20:1323–1329.

10. Liu S, Schmitz C, Stampfer MJ, Sacks F, Hennekens CH, Lindpaintner K,Ridker PM. A prospective study of TaqIB polymorphism in the gene codingfor cholesteryl ester transfer protein and risk of myocardial infarction inmiddle-aged men. Atherosclerosis 2002;161:469–474.

11. Carlquist JF, Muhlestein JB, Horne BD, Hart NI, Bair TL, Molhuizen HO,Anderson JL. The cholesteryl ester transfer protein Taq1B gene polymorphismpredicts clinical benefit of statin therapy in patients with significant coronaryartery disease. Am Heart J 2003;146:1007–1014.

12. Boekholdt SM, Sacks FM, Jukema JW, Shepherd J, Freeman DJ, McMahon AD,Cambien F, Nicaud V, de Grooth GJ, Talmud PJ, Humphries SE, Miller GJ,Eiriksdottir G, Gudnason V, Kauma H, Kakko S, Savolainen MJ, Arca M,Montali A, Liu S, Lanz HJ, Zwinderman AH, Kuivenhoven JA, Kastelein JJ. Cho-lesteryl ester transfer protein TaqIB variant, high-density lipoprotein cholesterollevels, cardiovascular risk, and efficacy of pravastatin treatment: individual patientmeta-analysis of 13,677 subjects. Circulation 2005;111:278–287.

13. Jukema JW, Bruschke AV, van Boven AJ, Reiber JH, Bal ET, Zwinderman AH,Jansen H, Boerma GJ, van Rappard FM, Lie KI. Effects of lipid lowering by pravas-tatin on progression and regression of coronary artery disease in symptomaticmen with normal to moderately elevated serum cholesterol levels. TheRegression Growth Evaluation Statin Study (REGRESS). Circulation 1995;91:2528–2540.

14. Regieli JJ, Jukema JW, Grobbee DE, Kastelein JJ, Kuivenhoven JA,Zwinderman AH, van der Graaf Y, Bots ML, Doevendans PA. CETP genotypepredicts increased mortality in statin-treated men with proven cardiovasculardisease: an adverse pharmacogenetic interaction. Eur Heart J 2008;29:2792–2799.

15. Utermann G. Apolipoprotein E polymorphism in health and disease. Am Heart J1987;113:433–440.

16. Nieminen T, Kahonen M, Viiri LE, Gronroos P, Lehtimaki T. Pharmacogenetics ofapolipoprotein E gene during lipid-lowering therapy: lipid levels and preventionof coronary heart disease. Pharmacogenomics 2008;9:1475–1486.

17. Thompson JF, Hyde CL, Wood LS, Paciga SA, Hinds DA, Cox DR, Hovingh GK,Kastelein JJ. Comprehensive whole-genome and candidate gene analysis forresponse to statin therapy in the Treating to New Targets (TNT) cohort. CircCardiovasc Genet 2009;2:173–181.

18. Zintzaras E, Kitsios GD, Triposkiadis F, Lau J, Raman G. APOE gene polymorph-isms and response to statin therapy. Pharmacogenomics J 2009;9:248–257.

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Table 3 Best explored single nucleotidepolymorphisms in the field of cardiovascularpharmacogenetics

Gene SNP Drug Effect ofvariant

CYP2C19 *2 (rs4244285) Clopidogrel Decreasedeffect

CYP2C9 *2 (rs1799853) Warfarin Increasedeffect*3 (rs1057910)

VKORC1 21639A . G(rs9923231)

Warfarin Decreasedeffect

1173T . C(rs9934438)

ACE I/D (rs4646994) ACE-inhibitors No effect

Overview of pharmacogenetics in CVD 175D

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 12: A systematic review on pharmacogenetics in cardiovascular disease

19. Barber MJ, Mangravite LM, Hyde CL, Chasman DI, Smith JD, McCarty CA, Li X,Wilke RA, Rieder MJ, Williams PT, Ridker PM, Chatterjee A, Rotter JI,Nickerson DA, Stephens M, Krauss RM. Genome-wide association oflipid-lowering response to statins in combined study populations. PLoS One2010;5:e9763.

20. Ishisaki Z, Takaishi M, Furuta I, Huh N. Calmin, a protein with calponin homologyand transmembrane domains expressed in maturing spermatogenic cells. Geno-mics 2001;74:172–179.

21. Hirosawa M, Nagase T, Ishikawa K, Kikuno R, Nomura N, Ohara O. Character-ization of cDNA clones selected by the GeneMark analysis from size-fractionated cDNA libraries from human brain. DNA Res 1999;6:329–336.

22. Iakoubova OA, Tong CH, Rowland CM, Kirchgessner TG, Young BA,Arellano AR, Shiffman D, Sabatine MS, Campos H, Packard CJ, Pfeffer MA,White TJ, Braunwald E, Shepherd J, Devlin JJ, Sacks FM. Association of theTrp719Arg polymorphism in kinesin-like protein 6 with myocardial infarctionand coronary heart disease in 2 prospective trials: the CARE and WOSCOPStrials. J Am Coll Cardiol 2008;51:435–443.

23. Iakoubova OA, Sabatine MS, Rowland CM, Tong CH, Catanese JJ, Ranade K,Simonsen KL, Kirchgessner TG, Cannon CP, Devlin JJ, Braunwald E. Polymorph-ism in KIF6 gene and benefit from statins after acute coronary syndromes: resultsfrom the PROVE IT-TIMI 22 study. J Am Coll Cardiol 2008;51:449–455.

24. Iakoubova OA, Robertson M, Tong CH, Rowland CM, Catanese JJ, Blauw GJ,Jukema JW, Murphy MB, Devlin JJ, Ford I, Shepherd J. KIF6 Trp719Arg poly-morphism and the effect of statin therapy in elderly patients: results from thePROSPER study. Eur J Cardiovasc Prev Rehabil 2010;17:455–461.

25. Miki H, Setou M, Kaneshiro K, Hirokawa N. All kinesin superfamily protein, KIF,genes in mouse and human. Proc Natl Acad Sci U S A 2001;98:7004–7011.

26. Assimes TL, Holm H, Kathiresan S, Reilly MP, Thorleifsson G, Voight BF,Erdmann J, Willenborg C, Vaidya D, Xie C, Patterson CC, Morgan TM,Burnett MS, Li M, Hlatky MA, Knowles JW, Thompson JR, Absher D,Iribarren C, Go A, Fortmann SP, Sidney S, Risch N, Tang H, Myers RM,Berger K, Stoll M, Shah SH, Thorgeirsson G, Andersen K, Havulinna AS,Herrera JE, Faraday N, Kim Y, Kral BG, Mathias RA, Ruczinski I, Suktitipat B,Wilson AF, Yanek LR, Becker LC, Linsel-Nitschke P, Lieb W, Konig IR,Hengstenberg C, Fischer M, Stark K, Reinhard W, Winogradow J, Grassl M,Grosshennig A, Preuss M, Schreiber S, Wichmann HE, Meisinger C, Yee J,Friedlander Y, Do R, Meigs JB, Williams G, Nathan DM, MacRae CA, Qu L,Wilensky RL, Matthai WH Jr, Qasim AN, Hakonarson H, Pichard AD,Kent KM, Satler L, Lindsay JM, Waksman R, Knouff CW, Waterworth DM,Walker MC, Mooser VE, Marrugat J, Lucas G, Subirana I, Sala J, Ramos R,Martinelli N, Olivieri O, Trabetti E, Malerba G, Pignatti PF, Guiducci C,Mirel D, Parkin M, Hirschhorn JN, Asselta R, Duga S, Musunuru K, Daly MJ,Purcell S, Eifert S, Braund PS, Wright BJ, Balmforth AJ, Ball SG,Ouwehand WH, Deloukas P, Scholz M, Cambien F, Huge A, Scheffold T,Salomaa V, Girelli D, Granger CB, Peltonen L, McKeown PP, Altshuler D,Melander O, Devaney JM, Epstein SE, Rader DJ, Elosua R, Engert JC,Anand SS, Hall AS, Ziegler A, O’Donnell CJ, Spertus JA, Siscovick D,Schwartz SM, Becker D, Thorsteinsdottir U, Stefansson K, Schunkert H,Samani NJ, Quertermous T. Lack of association between the Trp719Arg poly-morphism in kinesin-like protein-6 and coronary artery disease in 19 case-control studies. J Am Coll Cardiol 2010;56:1552–1563.

27. Law M, Rudnicka AR. Statin safety: a systematic review. Am J Cardiol 2006;97:S52–S60.

28. Pasanen MK, Fredrikson H, Neuvonen PJ, Niemi M. Different effects ofSLCO1B1 polymorphism on the pharmacokinetics of atorvastatin and rosuvas-tatin. Clin Pharmacol Ther 2007;82:726–733.

29. Niemi M, Pasanen MK, Neuvonen PJ. SLCO1B1 polymorphism and sex affect thepharmacokinetics of pravastatin but not fluvastatin. Clin Pharmacol Ther 2006;80:356–366.

30. Tirona RG, Leake BF, Merino G, Kim RB. Polymorphisms in OATP-C: identifi-cation of multiple allelic variants associated with altered transport activityamong European- and African-Americans. J Biol Chem 2001;276:35669–35675.

31. Pasanen MK, Neuvonen M, Neuvonen PJ, Niemi M. SLCO1B1 polymorphismmarkedly affects the pharmacokinetics of simvastatin acid. Pharmacogenet Geno-mics 2006;16:873–879.

32. Link E, Parish S, Armitage J, Bowman L, Heath S, Matsuda F, Gut I, Lathrop M,Collins R. SLCO1B1 variants and statin-induced myopathy—a genomewidestudy. N Engl J Med 2008;359:789–799.

33. Voora D, Shah SH, Spasojevic I, Ali S, Reed CR, Salisbury BA, Ginsburg GS. TheSLCO1B1*5 genetic variant is associated with statin-induced side effects. J AmColl Cardiol 2009;54:1609–1616.****

34. Brunham LR, Lansberg PJ, Zhang L, Miao F, Carter C, Hovingh GK, Visscher H,Jukema JW, Stalenhoef AF, Ross CJ, Carleton BC, Kastelein JJ, Hayden MR.Differential effect of the rs4149056 variant in SLCO1B1 on myopathy associated

with simvastatin and atorvastatin. Pharmacogenomics J 2011; doi:10.1038/tpj.2010.92..

35. Willer CJ, Sanna S, Jackson AU, Scuteri A, Bonnycastle LL, Clarke R, Heath SC,Timpson NJ, Najjar SS, Stringham HM, Strait J, Duren WL, Maschio A,Busonero F, Mulas A, Albai G, Swift AJ, Morken MA, Narisu N, Bennett D,Parish S, Shen H, Galan P, Meneton P, Hercberg S, Zelenika D, Chen WM,Li Y, Scott LJ, Scheet PA, Sundvall J, Watanabe RM, Nagaraja R, Ebrahim S,Lawlor DA, Ben-Shlomo Y, Davey-Smith G, Shuldiner AR, Collins R,Bergman RN, Uda M, Tuomilehto J, Cao A, Collins FS, Lakatta E,Lathrop GM, Boehnke M, Schlessinger D, Mohlke KL, Abecasis GR. Newly ident-ified loci that influence lipid concentrations and risk of coronary artery disease.Nat Genet 2008;40:161–169.

36. Kathiresan S, Willer CJ, Peloso GM, Demissie S, Musunuru K, Schadt EE,Kaplan L, Bennett D, Li Y, Tanaka T, Voight BF, Bonnycastle LL, Jackson AU,Crawford G, Surti A, Guiducci C, Burtt NP, Parish S, Clarke R, Zelenika D,Kubalanza KA, Morken MA, Scott LJ, Stringham HM, Galan P, Swift AJ,Kuusisto J, Bergman RN, Sundvall J, Laakso M, Ferrucci L, Scheet P, Sanna S,Uda M, Yang Q, Lunetta KL, Dupuis J, de Bakker PI, O’Donnell CJ,Chambers JC, Kooner JS, Hercberg S, Meneton P, Lakatta EG, Scuteri A,Schlessinger D, Tuomilehto J, Collins FS, Groop L, Altshuler D, Collins R,Lathrop GM, Melander O, Salomaa V, Peltonen L, Orho-Melander M,Ordovas JM, Boehnke M, Abecasis GR, Mohlke KL, Cupples LA. Common var-iants at 30 loci contribute to polygenic dyslipidemia. Nat Genet 2009;41:56–65.

37. Kushner FG, Hand M, Smith SC Jr, King SB III, Anderson JL, Antman EM,Bailey SR, Bates ER, Blankenship JC, Casey DE Jr, Green LA, Hochman JS,Jacobs AK, Krumholz HM, Morrison DA, Ornato JP, Pearle DL, Peterson ED,Sloan MA, Whitlow PL, Williams DO. 2009 focused updates: ACC/AHA guide-lines for the management of patients with ST-elevation myocardial infarction(updating the 2004 guideline and 2007 focused update) and ACC/AHA/SCAIguidelines on percutaneous coronary intervention (updating the 2005 guidelineand 2007 focused update): a report of the American College of CardiologyFoundation/American Heart Association Task Force on Practice Guidelines. Cir-culation 2009;120:2271–2306.

38. Van de Werf F, Bax J, Betriu A, Blomstrom-Lundqvist C, Crea F, Falk V,Filippatos G, Fox K, Huber K, Kastrati A, Rosengren A, Steg PG, Tubaro M,Verheugt F, Weidinger F, Weis M, Vahanian A, Camm J, De Caterina R,Dean V, Dickstein K, Filippatos G, Funck-Brentano C, Hellemans I,Kristensen SD, McGregor K, Sechtem U, Silber S, Tendera M, Widimsky P,Zamorano JL, Silber S, Aguirre FV, Al-Attar N, Alegria E, Andreotti F,Benzer W, Breithardt O, Danchin N, Di Mario C, Dudek D, Gulba D,Halvorsen S, Kaufmann P, Kornowski R, Lip GY, Rutten F. Management ofacute myocardial infarction in patients presenting with persistent ST-segmentelevation: the Task Force on the Management of ST-Segment Elevation AcuteMyocardial Infarction of the European Society of Cardiology. Eur Heart J 2008;29:2909–2945.

39. Pollack CV Jr, Braunwald E. 2007 update to the ACC/AHA guidelines for themanagement of patients with unstable angina and non-ST-segment elevationmyocardial infarction: implications for emergency department practice. AnnEmerg Med 2008;51:591–606.

40. Rapezzi C, Biagini E, Branzi A. Guidelines for the diagnosis and treatment ofnon-ST-segment elevation acute coronary syndromes: the task force for thediagnosis and treatment of non-ST-segment elevation acute coronary syn-dromes of the European Society of Cardiology. Eur Heart J 2008;29:277–278.

41. Matetzky S, Shenkman B, Guetta V, Shechter M, Bienart R, Goldenberg I,Novikov I, Pres H, Savion N, Varon D, Hod H. Clopidogrel resistance is associ-ated with increased risk of recurrent atherothrombotic events in patients withacute myocardial infarction. Circulation 2004;109:3171–3175.

42. Snoep JD, Hovens MM, Eikenboom JC, van der Bom JG, Jukema JW,Huisman MV. Clopidogrel nonresponsiveness in patients undergoing percuta-neous coronary intervention with stenting: a systematic review andmeta-analysis. Am Heart J 2007;154:221–231.

43. Szczeklik A, Musial J, Undas A, Sanak M. Aspirin resistance. J Thromb Haemost2005;3:1655–1662.

44. Trenk D, Hochholzer W, Fromm MF, Chialda LE, Pahl A, Valina CM, Stratz C,Schmiebusch P, Bestehorn HP, Buttner HJ, Neumann FJ. Cytochrome P4502C19 681G . A polymorphism and high on-clopidogrel platelet reactivityassociated with adverse 1-year clinical outcome of elective percutaneous coron-ary intervention with drug-eluting or bare-metal stents. J Am Coll Cardiol 2008;51:1925–1934.

45. Goodman T, Ferro A, Sharma P. Pharmacogenetics of aspirin resistance: a com-prehensive systematic review. Br J Clin Pharmacol 2008;66:222–232.

46. Fontana P, Nolli S, Reber G, de Moerloose P. Biological effects of aspirin andclopidogrel in a randomized cross-over study in 96 healthy volunteers.J Thromb Haemost 2006;4:813–819.

J.J.W. Verschuren et al.175aD

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 13: A systematic review on pharmacogenetics in cardiovascular disease

47. Lordkipanidze M, Pharand C, Schampaert E, Turgeon J, Palisaitis DA, Diodati JG.A comparison of six major platelet function tests to determine the prevalence ofaspirin resistance in patients with stable coronary artery disease. Eur Heart J2007;28:1702–1708.

48. Shimokawa T, Smith WL. Prostaglandin endoperoxide synthase. The aspirinacetylation region. J Biol Chem 1992;267:12387–12392.

49. Halushka MK, Walker LP, Halushka PV. Genetic variation in cyclooxygenase 1:effects on response to aspirin. Clin Pharmacol Ther 2003;73:122–130.

50. Preston FE, Whipps S, Jackson CA, French AJ, Wyld PJ, Stoddard CJ. Inhibition ofprostacyclin and platelet thromboxane A2 after low-dose aspirin. N Engl J Med1981;304:76–79.

51. Hovens MMC, Snoep JD, Eikenboom JCJ, van der Bom JG, Mertens BJA,Huisman MV. Prevalence of persistent platelet reactivity despite use of aspirin:a systematic review. Am Heart J 2007;153:175–181.

52. Maree AO, Curtin RJ, Chubb A, Dolan C, Cox D, O’Brien J, Crean P,Shields DC, Fitzgerald DJ. Cyclooxygenase-1 haplotype modulates plateletresponse to aspirin. J Thromb Haemost 2005;3:2340–2345.

53. Lepantalo A, Mikkelsson J, Resendiz JC, Viiri L, Backman JT, Kankuri E,Karhunen PJ, Lassila R. Polymorphisms of COX-1 and GPVI associate with theantiplatelet effect of aspirin in coronary artery disease patients. ThrombHaemost 2006;95:253–259.

54. Kunicki TJ, Williams SA, Nugent DJ, Harrison P, Segal HC, Syed A, Rothwell PM.Lack of association between aspirin responsiveness and seven candidate genehaplotypes in patients with symptomatic vascular disease. Thromb Haemost2009;101:123–133.

55. Li Q, Chen BL, Ozdemir V, Ji W, Mao YM, Wang LC, Lei HP, Fan L, Zhang W,Liu J, Zhou HH. Frequency of genetic polymorphisms of COX1, GPIIIa and P2Y1in a Chinese population and association with attenuated response to aspirin.Pharmacogenomics 2007;8:577–586.

56. Clappers N, van Oijen MG, Sundaresan S, Brouwer MA, Te Morsche RH,Keuper W, Peters WH, Drenth JP, Verheugt FW. The C50T polymorphism ofthe cyclooxygenase-1 gene and the risk of thrombotic events during low-dosetherapy with acetyl salicylic acid. Thromb Haemost 2008;100:70–75.

57. Feher G, Feher A, Pusch G, Lupkovics G, Szapary L, Papp E. The genetics of anti-platelet drug resistance. Clin Genet 2009;75:1–18.

58. Su G, Wang Z, Ding Y. Association of the platelet membrane glycoprotein I aC807T gene polymorphism with aspirin resistance. J Huazhong Univ Sci TechnologMed Sci 2007;27:664–667.

59. Jefferson BK, Foster JH, McCarthy JJ, Ginsburg G, Parker A, Kottke-Marchant K,Topol EJ. Aspirin resistance and a single gene. Am J Cardiol 2005;95:805–808.

60. Choi JH, Kim SH, Cho BY, Lee SK, Kim SH, Suh CH, Park HS. Association ofTNF-alpha promoter polymorphisms with aspirin-induced urticaria. J ClinPharm Ther 2009;34:231–238.

61. Park HJ, Ye YM, Hur GY, Kim SH, Park HS. Association between a TGFbeta1promoter polymorphism and the phenotype of aspirin-intolerant chronic urti-caria in a Korean population. J Clin Pharm Ther 2008;33:691–697.

62. Piazuelo E, Fuentes J, Garcia-Gonzalez MA, Jimenez P, Lanas A. A case–controlstudy of the association between polymorphisms of the endothelial nitric oxidesynthase and glycoprotein IIIa genes and upper gastrointestinal bleeding in usersof low-dose aspirin. Clin Ther 2008;30:121–130.

63. Mega JL, Close SL, Wiviott SD, Shen L, Hockett RD, Brandt JT, Walker JR,Antman EM, Macias W, Braunwald E, Sabatine MS. Cytochrome p-450 poly-morphisms and response to clopidogrel. N Engl J Med 2009;360:354–362.

64. Ellis KJ, Stouffer GA, McLeod HL, Lee CR. Clopidogrel pharmacogenomics andrisk of inadequate platelet inhibition: US FDA recommendations. Pharmacoge-nomics 2009;10:1799–1817.

65. Sugunaraj JP, Palaniswamy C, Selvaraj DR, Chaitanya Arudra SK, Sukhija R. Clo-pidogrel resistance. Am J Ther 2010;17:210–215.

66. Ford NF. Clopidogrel resistance: pharmacokinetic or pharmacogenetic? J ClinPharmacol 2009;49:506–512.

67. Marin F, Gonzalez-Conejero R, Capranzano P, Bass TA, Roldan V, Angiolillo DJ.Pharmacogenetics in cardiovascular antithrombotic therapy. J Am Coll Cardiol2009;54:1041–1057.

68. Hulot JS, Bura A, Villard E, Azizi M, Remones V, Goyenvalle C, Aiach M, Lechat P,Gaussem P. Cytochrome P450 2C19 loss-of-function polymorphism is a majordeterminant of clopidogrel responsiveness in healthy subjects. Blood 2006;108:2244–2247.

69. Brandt JT, Close SL, Iturria SJ, Payne CD, Farid NA, Ernest CS, Lachno DR,Salazar D, Winters KJ. Common polymorphisms of CYP2C19 and CYP2C9affect the pharmacokinetic and pharmacodynamic response to clopidogrel butnot prasugrel. J Thromb Haemost 2007;5:2429–2436.

70. Simon T, Verstuyft C, Mary-Krause M, Quteineh L, Drouet E, Meneveau N,Steg PG, Ferrieres J, Danchin N, Becquemont L. Genetic determinants ofresponse to clopidogrel and cardiovascular events. N Engl J Med 2009;360:363–375.

71. Geisler T, Schaeffeler E, Dippon J, Winter S, Buse V, Bischofs C, Zuern C,Moerike K, Gawaz M, Schwab M. CYP2C19 and nongenetic factors predictpoor responsiveness to clopidogrel loading dose after coronary stent implan-tation. Pharmacogenomics 2008;9:1251–1259.

72. Gladding P, Webster M, Zeng I, Farrell H, Stewart J, Ruygrok P, Ormiston J,El-Jack S, Armstrong G, Kay P, Scott D, Gunes A, Dahl ML. The pharmacoge-netics and pharmacodynamics of clopidogrel response: an analysis from thePRINC (Plavix Response in Coronary Intervention) trial. JACC Cardiovasc Interv2008;1:620–627.

73. Sibbing D, Koch W, Gebhard D, Schuster T, Braun S, Stegherr J, Morath T,Schomig A, von BN, Kastrati A. Cytochrome 2C19*17 allelic variant, plateletaggregation, bleeding events, and stent thrombosis in clopidogrel-treatedpatients with coronary stent placement. Circulation 2010;121:512–518.

74. Wallentin L, James S, Storey RF, Armstrong M, Barratt BJ, Horrow J, Husted S,Katus H, Steg PG, Shah SH, Becker RC. Effect of CYP2C19 and ABCB1 singlenucleotide polymorphisms on outcomes of treatment with ticagrelor versus clo-pidogrel for acute coronary syndromes: a genetic substudy of the PLATO trial.Lancet 2010;376:1320–1328.

75. Harmsze A, van Werkum JW, Bouman HJ, Ruven HJ, Breet NJ, Ten Berg JM,Hackeng CM, Tjoeng MM, Klungel OH, de Boer A, Deneer VH. BesidesCYP2C19*2, the variant allele CYP2C9*3 is associated with higheron-clopidogrel platelet reactivity in patients on dual antiplatelet therapy under-going elective coronary stent implantation. Pharmacogenet Genomics 2010;20:18–25.

76. Gladding P, White H, Voss J, Ormiston J, Stewart J, Ruygrok P, Bvaldivia B,Baak R, White C, Webster M. Pharmacogenetic testing for clopidogrel usingthe rapid INFINITI analyzer: a dose-escalation study. JACC Cardiovasc Interv2009;2:1095–1101.

77. Taubert D, von BN, Grimberg G, Lazar A, Jung N, Goeser T, Kastrati A,Schomig A, Schomig E. Impact of P-glycoprotein on clopidogrel absorption.Clin Pharmacol Ther 2006;80:486–501.

78. Mega JL, Close SL, Wiviott SD, Shen L, Walker JR, Simon T, Antman EM,Braunwald E, Sabatine MS. Genetic variants in ABCB1 and CYP2C19 and cardi-ovascular outcomes after treatment with clopidogrel and prasugrel in theTRITON-TIMI 38 trial: a pharmacogenetic analysis. Lancet 2010;376:1312–1319.

79. Shuldiner AR, O’Connell JR, Bliden KP, Gandhi A, Ryan K, Horenstein RB,Damcott CM, Pakyz R, Tantry US, Gibson Q, Pollin TI, Post W, Parsa A,Mitchell BD, Faraday N, Herzog W, Gurbel PA. Association of cytochromeP450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopido-grel therapy. JAMA 2009;302:849–857.

80. 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, Freij A, Thorsen M. Ticagrelor versus clopi-dogrel in patients with acute coronary syndromes. N Engl J Med 2009;361:1045–1057.

81. Biondi-Zoccai G, Lotrionte M, Agostoni P, Abbate A, Romagnoli E, Sangiorgi G,Angiolillo DJ, Valgimigli M, Testa L, Gaita F, Sheiban I. Adjusted indirect compari-son meta-analysis of prasugrel versus ticagrelor for patients with acute coronarysyndromes. Int J Cardiol 2010, doi:10.1016/j.ijcard.2010.08.035.

82. Storey RF, Angiolillo DJ, Patil SB, Desai B, Ecob R, Husted S, Emanuelsson H,Cannon CP, Becker RC, Wallentin L. Inhibitory effects of ticagrelor comparedwith clopidogrel on platelet function in patients with acute coronary syndromes:the PLATO (PLATelet inhibition and patient Outcomes) PLATELET substudy.J Am Coll Cardiol 2010;56:1456–1462.

83. Ziegler S, Schillinger M, Funk M, Felber K, Exner M, Mlekusch W, Sabeti S,Amighi J, Minar E, Brunner M, Muller M, Mannhalter C. Association of a func-tional polymorphism in the clopidogrel target receptor gene, P2Y12, and therisk for ischemic cerebrovascular events in patients with peripheral arterydisease. Stroke 2005;36:1394–1399.

84. Staritz P, Kurz K, Stoll M, Giannitsis E, Katus HA, Ivandic BT. Platelet reactivityand clopidogrel resistance are associated with the H2 haplotype of theP2Y12-ADP receptor gene. Int J Cardiol 2009;133:341–345.

85. Cuisset T, Frere C, Quilici J, Morange PE, Saut N, Lambert M, Camoin L, Vague IJ,Bonnet JL, Alessi MC. Role of the T744C polymorphism of the P2Y12 gene onplatelet response to a 600-mg loading dose of clopidogrel in 597 patients withnon-ST-segment elevation acute coronary syndrome. Thromb Res 2007;120:893–899.

86. Giusti B, Gori AM, Marcucci R, Saracini C, Sestini I, Paniccia R, Valente S,Antoniucci D, Abbate R, Gensini GF. Cytochrome P450 2C19 loss-of-functionpolymorphism, but not CYP3A4 IVS10 + 12G/A and P2Y12 T744C polymorph-isms, is associated with response variability to dual antiplatelet treatment in high-risk vascular patients. Pharmacogenet Genomics 2007;17:1057–1064.

87. Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, Ramirez C, Escaned J, Moreno R,Hernandez-Antolin R, Sabate M, Trabetti E, Pignatti PF, Macaya C. 807 C/T poly-morphism of the glycoprotein Ia gene and pharmacogenetic modulation of

Overview of pharmacogenetics in CVD 175bD

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 14: A systematic review on pharmacogenetics in cardiovascular disease

platelet response to dual antiplatelet treatment. Blood Coagul Fibrinolysis 2004;15:427–433.

88. Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, Ramirez C, Cavallari U, Trabetti E,Sabate M, Jimenez-Quevedo P, Hernandez R, Moreno R, Escaned J, Alfonso F,Banuelos C, Costa MA, Bass TA, Pignatti PF, Macaya C. Variability in plateletaggregation following sustained aspirin and clopidogrel treatment in patientswith coronary heart disease and influence of the 807 C/T polymorphism ofthe glycoprotein Ia gene. Am J Cardiol 2005;96:1095–1099.

89. Cuisset T, Frere C, Quilici J, Morange PE, Saut N, Romero-Barra M, Camoin L,Lambert M, Juhan-Vague I, Bonnet JL, Alessi MC. Lack of association betweenthe 807 C/T polymorphism of glycoprotein Ia gene and post-treatment plateletreactivity after aspirin and clopidogrel in patients with acute coronary syndrome.Thromb Haemost 2007;97:212–217.

90. Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, Alfonso F, Sabate M, Fernandez C,Stranieri C, Trabetti E, Pignatti PF, Macaya C. PlA polymorphism and plateletreactivity following clopidogrel loading dose in patients undergoing coronarystent implantation. Blood Coagul Fibrinolysis 2004;15:89–93.

91. Dropinski J, Musial J, Jakiela B, Wegrzyn W, Sanak M, Szczeklik A.Anti-thrombotic action of clopidogrel and P1(A1/A2) polymorphism of beta3integrin in patients with coronary artery disease not being treated withaspirin. Thromb Haemost 2005;94:1300–1305.

92. Pare G, Mehta SR, Yusuf S, Anand SS, Connolly SJ, Hirsh J, Simonsen K, Bhatt DL,Fox KA, Eikelboom JW. Effects of CYP2C19 genotype on outcomes of clopido-grel treatment. N Engl J Med 2010;363:1704–1714.

93. Tran M, Tafreshi J, Pai RG. Combination of clopidogrel and proton pump inhibi-tors: implications for clinicians. J Cardiovasc Pharmacol Ther 2010;15:326–337.

94. Mega JL, Close SL, Wiviott SD, Shen L, Hockett RD, Brandt JT, Walker JR,Antman EM, Macias WL, Braunwald E, Sabatine MS. Cytochrome P450genetic polymorphisms and the response to prasugrel: relationship to pharma-cokinetic, pharmacodynamic, and clinical outcomes. Circulation 2009;119:2553–2560.

95. Rehmel JLF, Eckstein JA, Farid NA, Heim JB, Kasper SC, Kurihara A,Wrighton SA, Ring BJ. Interactions of two major metabolites of prasugrel, a thie-nopyridine antiplatelet agent, with the cytochromes P450. Drug Metab Dispos2006;34:600–607.

96. Schror K, Weber AA. Comparative pharmacology of GP IIb/IIIa antagonists.J Thromb Thrombolysis 2003;15:71–80.

97. O’Connor FF, Shields DC, Fitzgerald A, Cannon CP, Braunwald E, Fitzgerald DJ.Genetic variation in glycoprotein IIb/IIIa (GPIIb/IIIa) as a determinant of theresponses to an oral GPIIb/IIIa antagonist in patients with unstable coronary syn-dromes. Blood 2001;98:3256–3260.

98. Wheeler GL, Braden GA, Bray PF, Marciniak SJ, Mascelli MA, Sane DC. Reducedinhibition by abciximab in platelets with the PlA2 polymorphism. Am Heart J2002;143:76–82.

99. Michelson AD, Furman MI, Goldschmidt-Clermont P, Mascelli MA, Hendrix C,Coleman L, Hamlington J, Barnard MR, Kickler T, Christie DJ, Kundu S,Bray PF. Platelet GP IIIa Pl(A) polymorphisms display different sensitivities toagonists. Circulation 2000;101:1013–1018.

100. Aalto-Setala K, Karhunen PJ, Mikkelsson J, Niemela K. The effect of glycoproteinIIIa PIA 1/A2 polymorphism on the PFA-100 response to GP IIb IIa receptorinhibitors-the importance of anticoagulants used. J Thromb Thrombolysis 2005;20:57–63.

101. Weber AA, Jacobs C, Meila D, Weber S, Zotz RB, Scharf RE, Kelm M,Strauer BE, Schror K. No evidence for an influence of the human plateletantigen-1 polymorphism on the antiplatelet effects of glycoprotein IIb/IIIa inhibi-tors. Pharmacogenetics 2002;12:581–583.

102. Weber AA, Meila D, Jacobs C, Weber S, Kelm M, Strauer BE, Zotz RB,Scharf RE, Schror K. Low incidence of paradoxical platelet activation by glyco-protein IIb/IIIa inhibitors. Thromb Res 2002;106:25–29.

103. Shields DC, Fitzgerald AP, O’Neill PA, Muckian C, Kenny D, Moran B,Cannon CP, Byrne CE, Fitzgerald DJ. The contribution of genetic factors tothrombotic and bleeding outcomes in coronary patients randomised to IIb/IIIaantagonists. Pharmacogenomics J 2002;2:182–190.

104. Hillman MA, Wilke RA, Caldwell MD, Berg RL, Glurich I, Burmester JK. Relativeimpact of covariates in prescribing warfarin according to CYP2C9 genotype.Pharmacogenetics 2004;14:539–547.

105. Caldwell MD, Berg RL, Zhang KQ, Glurich I, Schmelzer JR, Yale SH, Vidaillet HJ,Burmester JK. Evaluation of genetic factors for warfarin dose prediction. Clin MedRes 2007;5:8–16.

106. Klein TE, Altman RB, Eriksson N, Gage BF, Kimmel SE, Lee MT, Limdi NA,Page D, Roden DM, Wagner MJ, Caldwell MD, Johnson JA. Estimation of thewarfarin dose with clinical and pharmacogenetic data. N Engl J Med 2009;360:753–764.

107. Furuya H, Fernandez-Salguero P, Gregory W, Taber H, Steward A, Gonzalez FJ,Idle JR. Genetic polymorphism of CYP2C9 and its effect on warfarin

maintenance dose requirement in patients undergoing anticoagulation therapy.Pharmacogenetics 1995;5:389–392.

108. Aithal GP, Day CP, Kesteven PJ, Daly AK. Association of polymorphisms in thecytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleedingcomplications. Lancet 1999;353:717–719.

109. Rettie AE, Wienkers LC, Gonzalez FJ, Trager WF, Korzekwa KR. Impaired(S)-warfarin metabolism catalysed by the R144C allelic variant of CYP2C9. Phar-macogenetics 1994;4:39–42.

110. Haining RL, Hunter AP, Veronese ME, Trager WF, Rettie AE. Allelic variants ofhuman cytochrome P450 2C9: baculovirus-mediated expression, purification,structural characterization, substrate stereoselectivity, and prochiral selectivityof the wild-type and I359L mutant forms. Arch Biochem Biophys 1996;333:447–458.

111. Lindh JD, Holm L, Andersson ML, Rane A. Influence of CYP2C9 genotype onwarfarin dose requirements—a systematic review and meta-analysis. Eur J ClinPharmacol 2009;65:365–375.

112. Kangelaris KN, Bent S, Nussbaum RL, Garcia DA, Tice JA. Genetic testing beforeanticoagulation? A systematic review of pharmacogenetic dosing of warfarin.J Gen Intern Med 2009;24:656–664.

113. Schalekamp T, de Boer A. Pharmacogenetics of oral anticoagulant therapy. CurrPharm Des 2010;16:187–203.

114. Yang L, Ge W, Yu F, Zhu H. Impact of VKORC1 gene polymorphism on inter-individual and interethnic warfarin dosage requirement—a systematic review andmeta analysis. Thromb Res 2010;125:e159–e166.

115. Voora D, Koboldt DC, King CR, Lenzini PA, Eby CS, Porche-Sorbet R, Deych E,Crankshaw M, Milligan PE, McLeod HL, Patel SR, Cavallari LH, Ridker PM,Grice GR, Miller RD, Gage BF. A polymorphism in the VKORC1 regulatorcalumenin predicts higher warfarin dose requirements in African Americans.Clin Pharmacol Ther 2010;87:445–451.

116. Takeuchi F, McGinnis R, Bourgeois S, Barnes C, Eriksson N, Soranzo N,Whittaker P, Ranganath V, Kumanduri V, McLaren W, Holm L, Lindh J,Rane A, Wadelius M, Deloukas P. A genome-wide association study confirmsVKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarindose. PLoS Genet 2009;5:e1000433.

117. D’Ambrosio RL, D’Andrea G, Cappucci F, Chetta M, Di Perna P, Brancaccio V,Grandone E, Margaglione M. Polymorphisms in factor II and factor VII genesmodulate oral anticoagulation with warfarin. Haematologica 2004;89:1510–1516.

118. Wadelius M, Chen LY, Downes K, Ghori J, Hunt S, Eriksson N, Wallerman O,Melhus H, Wadelius C, Bentley D, Deloukas P. Common VKORC1 andGGCX polymorphisms associated with warfarin dose. Pharmacogenomics J2005;5:262–270.

119. Pautas E, Moreau C, Gouin-Thibault I, Golmard JL, Mahe I, Legendre C,Taillandier-Heriche E, Durand-Gasselin B, Houllier AM, Verrier P, Beaune P,Loriot MA, Siguret V. Genetic factors (VKORC1, CYP2C9, EPHX1, andCYP4F2) are predictor variables for warfarin response in very elderly, frail inpa-tients. Clin Pharmacol Ther 2010;87:57–64.

120. Cooper GM, Johnson JA, Langaee TY, Feng H, Stanaway IB, Schwarz UI,Ritchie MD, Stein CM, Roden DM, Smith JD, Veenstra DL, Rettie AE,Rieder MJ. A genome-wide scan for common genetic variants with a large influ-ence on warfarin maintenance dose. Blood 2008;112:1022–1027.

121. Beinema M, Brouwers JR, Schalekamp T, Wilffert B. Pharmacogenetic differencesbetween warfarin, acenocoumarol and phenprocoumon. Thromb Haemost 2008;100:1052–1057.

122. Ufer M, Kammerer B, Kahlich R, Kirchheiner J, Yasar U, Brockmoller J, Rane A.Genetic polymorphisms of cytochrome P450 2C9 causing reduced phenprocou-mon (S)-7-hydroxylation in vitro and in vivo. Xenobiotica 2004;34:847–859.

123. Puehringer H, Loreth RM, Klose G, Schreyer B, Krugluger W, Schneider B,Oberkanins C. VKORC1 21639G . A and CYP2C9*3 are the major geneticpredictors of phenprocoumon dose requirement. Eur J Clin Pharmacol 2010;66:591–598.

124. Teichert M, Eijgelsheim M, Rivadeneira F, Uitterlinden AG, van Schaik RH,Hofman A, De Smet PA, van Gelder T, Visser LE, Stricker BH. A genome-wideassociation study of acenocoumarol maintenance dosage. Hum Mol Genet 2009;18:3758–3768.

125. Gage BF, Eby C, Johnson JA, Deych E, Rieder MJ, Ridker PM, Milligan PE, Grice G,Lenzini P, Rettie AE, Aquilante CL, Grosso L, Marsh S, Langaee T, Farnett LE,Voora D, Veenstra DL, Glynn RJ, Barrett A, McLeod HL. Use of pharmacoge-netic and clinical factors to predict the therapeutic dose of warfarin. Clin Pharma-col Ther 2008;84:326–331.

126. Tham LS, Goh BC, Nafziger A, Guo JY, Wang LZ, Soong R, Lee SC. A warfarin-dosing model in Asians that uses single-nucleotide polymorphisms in vitamin Kepoxide reductase complex and cytochrome P450 2C9. Clin Pharmacol Ther2006;80:346–355.

J.J.W. Verschuren et al.175cD

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 15: A systematic review on pharmacogenetics in cardiovascular disease

127. Wadelius M, Chen LY, Lindh JD, Eriksson N, Ghori MJ, Bumpstead S, Holm L,McGinnis R, Rane A, Deloukas P. The largest prospective warfarin-treatedcohort supports genetic forecasting. Blood 2009;113:784–792.

128. Kuehn BM. Warfarin label update. JAMA 2007;298:1389.129. Bristow MR, Gilbert EM, Abraham WT, Adams KF, Fowler MB, Hershberger RE,

Kubo SH, Narahara KA, Ingersoll H, Krueger S, Young S, Shusterman N.Carvedilol produces dose-related improvements in left ventricular functionand survival in subjects with chronic heart failure. MOCHA Investigators. Circula-tion 1996;94:2807–2816.

130. Hjalmarson A, Goldstein S, Fagerberg B, Wedel H, Waagstein F, Kjekshus J,Wikstrand J, El AD, Vitovec J, Aldershvile J, Halinen M, Dietz R, Neuhaus KL,Janosi A, Thorgeirsson G, Dunselman PH, Gullestad L, Kuch J, Herlitz J,Rickenbacher P, Ball S, Gottlieb S, Deedwania P. Effects of controlled-releasemetoprolol on total mortality, hospitalizations, and well-being in patients withheart failure: the Metoprolol CR/XL Randomized Intervention Trial in conges-tive heart failure (MERIT-HF). MERIT-HF Study Group. JAMA 2000;283:1295–1302.

131. Effect of metoprolol CR/XL in chronic heart failure: Metoprolol CR/XL Ran-domised Intervention Trial in Congestive Heart Failure (MERIT-HF). Lancet1999;353:2001–2007.

132. Muthumala A, Drenos F, Elliott PM, Humphries SE. Role of beta adrenergicreceptor polymorphisms in heart failure: systematic review and meta-analysis.Eur J Heart Fail 2008;10:3–13.

133. Lobmeyer MT, Gong Y, Terra SG, Beitelshees AL, Langaee TY, Pauly DF,Schofield RS, Hamilton KK, Herbert PJ, Adams KF Jr, Hill JA, Aranda JM Jr,Johnson JA. Synergistic polymorphisms of beta1 and alpha2C-adrenergic recep-tors and the influence on left ventricular ejection fraction response to beta-blocker therapy in heart failure. Pharmacogenet Genomics 2007;17:277–282.

134. Chen L, Meyers D, Javorsky G, Burstow D, Lolekha P, Lucas M, Semmler AB,Savarimuthu SM, Fong KM, Yang IA, Atherton J, Galbraith AJ, Parsonage WA,Molenaar P. Arg389Gly-beta1-adrenergic receptors determine improvement inleft ventricular systolic function in nonischemic cardiomyopathy patients withheart failure after chronic treatment with carvedilol. Pharmacogenet Genomics2007;17:941–949.

135. Liggett SB, Mialet-Perez J, Thaneemit-Chen S, Weber SA, Greene SM, Hodne D,Nelson B, Morrison J, Domanski MJ, Wagoner LE, Abraham WT, Anderson JL,Carlquist JF, Krause-Steinrauf HJ, Lazzeroni LC, Port JD, Lavori PW,Bristow MR. A polymorphism within a conserved beta(1)-adrenergic receptormotif alters cardiac function and beta-blocker response in human heart failure.Proc Natl Acad Sci USA 2006;103:11288–11293.

136. White HL, de Boer RA, Maqbool A, Greenwood D, van Veldhuisen DJ,Cuthbert R, Ball SG, Hall AS, Balmforth AJ. An evaluation of the beta-1 adrener-gic receptor Arg389Gly polymorphism in individuals with heart failure: aMERIT-HF sub-study. Eur J Heart Fail 2003;5:463–468.

137. Liu J, Liu ZQ, Yu BN, Xu FH, Mo W, Zhou G, Liu YZ, Li Q, Zhou HH.Beta1-adrenergic receptor polymorphisms influence the response to metopro-lol monotherapy in patients with essential hypertension. Clin Pharmacol Ther2006;80:23–32.

138. Johnson JA, Zineh I, Puckett BJ, McGorray SP, Yarandi HN, Pauly DF. Beta1-adrenergic receptor polymorphisms and antihypertensive response to meto-prolol. Clin Pharmacol Ther 2003;74:44–52.

139. Sofowora GG, Dishy V, Muszkat M, Xie HG, Kim RB, Harris PA, Prasad HC,Byrne DW, Nair UB, Wood AJ, Stein CM. A common beta1-adrenergic recep-tor polymorphism (Arg389Gly) affects blood pressure response to beta-blockade. Clin Pharmacol Ther 2003;73:366–371.

140. O’Shaughnessy KM, Fu B, Dickerson C, Thurston D, Brown MJ. Thegain-of-function G389R variant of the beta1-adrenoceptor does not influenceblood pressure or heart rate response to beta-blockade in hypertensive sub-jects. Clin Sci (Lond) 2000;99:233–238.

141. Karlsson J, Lind L, Hallberg P, Michaelsson K, Kurland L, Kahan T, Malmqvist K,Ohman KP, Nystrom F, Melhus H. Beta1-adrenergic receptor gene polymorph-isms and response to beta1-adrenergic receptor blockade in patients with essen-tial hypertension. Clin Cardiol 2004;27:347–350.

142. Filigheddu F, Argiolas G, Degortes S, Zaninello R, Frau F, Pitzoi S, Bulla E, Bulla P,Troffa C, Glorioso N. Haplotypes of the adrenergic system predict the bloodpressure response to beta-blockers in women with essential hypertension. Phar-macogenomics 2010;11:319–325.

143. Terra SG, Hamilton KK, Pauly DF, Lee CR, Patterson JH, Adams KF,Schofield RS, Belgado BS, Hill JA, Aranda JM, Yarandi HN, Johnson JA.Beta1-adrenergic receptor polymorphisms and left ventricular remodelingchanges in response to beta-blocker therapy. Pharmacogenet Genomics 2005;15:227–234.

144. Terra SG, Pauly DF, Lee CR, Patterson JH, Adams KF, Schofield RS, Belgado BS,Hamilton KK, Aranda JM, Hill JA, Yarandi HN, Walker JR, Phillips MS,Gelfand CA, Johnson JA. Beta-adrenergic receptor polymorphisms and

responses during titration of metoprolol controlled release/extended releasein heart failure. Clin Pharmacol Ther 2005;77:127–137.

145. Magnusson Y, Levin MC, Eggertsen R, Nystrom E, Mobini R, Schaufelberger M,Andersson B. Ser49Gly of beta1-adrenergic receptor is associated with effectivebeta-blocker dose in dilated cardiomyopathy. Clin Pharmacol Ther 2005;78:221–231.

146. de Groote P, Helbecque N, Lamblin N, Hermant X, Mc FE, Foucher-Hossein C,Amouyel P, Dallongeville J, Bauters C. Association between beta-1 and beta-2adrenergic receptor gene polymorphisms and the response to beta-blockadein patients with stable congestive heart failure. Pharmacogenet Genomics 2005;15:137–142.

147. Pacanowski MA, Gong Y, Cooper-Dehoff RM, Schork NJ, Shriver MD,Langaee TY, Pepine CJ, Johnson JA. Beta-adrenergic receptor gene polymorph-isms and beta-blocker treatment outcomes in hypertension. Clin Pharmacol Ther2008;84:715–721.

148. Troncoso R, Moraga F, Chiong M, Roldan J, Bravo R, Valenzuela R, Diaz-Araya G,del Campo A, Sanhueza C, Rodriguez A, Vukasovic JL, Mellado R, Greig D,Castro PF, Lavandero S. Gln(27) � Glubeta(2)-adrenergic receptor polymorph-ism in heart failure patients: differential clinical and oxidative response to carve-dilol. Basic Clin Pharmacol Toxicol 2009;104:374–378.

149. Kaye DM, Smirk B, Williams C, Jennings G, Esler M, Holst D. Beta-adrenoceptorgenotype influences the response to carvedilol in patients with congestive heartfailure. Pharmacogenetics 2003;13:379–382.

150. Littlejohn MD, Palmer BR, Richards AM, Frampton CM, Pilbrow AP,Troughton RW, Cameron VA, Kennedy MA. Ile164 variant ofbeta2-adrenoceptor does not influence outcome in heart failure but may inter-act with beta blocker treatment. Eur J Heart Fail 2008;10:55–59.

151. Brodde OE, Kroemer HK. Drug–drug interactions of beta-adrenoceptor block-ers. Arzneimittelforschung 2003;53:814–822.

152. Zanger UM, Raimundo S, Eichelbaum M. Cytochrome P450 2D6: overview andupdate on pharmacology, genetics, biochemistry. Naunyn Schmiedebergs ArchPharmacol 2004;369:23–37.

153. Ismail R, Teh LK. The relevance of CYP2D6 genetic polymorphism on chronicmetoprolol therapy in cardiovascular patients. J Clin Pharm Ther 2006;31:99–109.

154. Lefebvre J, Poirier L, Poirier P, Turgeon J, Lacourciere Y. The influence ofCYP2D6 phenotype on the clinical response of nebivolol in patients with essen-tial hypertension. Br J Clin Pharmacol 2007;63:575–582.

155. Fux R, Morike K, Prohmer AM, Delabar U, Schwab M, Schaeffeler E, Lorenz G,Gleiter CH, Eichelbaum M, Kivisto KT. Impact of CYP2D6 genotype on adverseeffects during treatment with metoprolol: a prospective clinical study. Clin Phar-macol Ther 2005;78:378–387.

156. Rau T, Heide R, Bergmann K, Wuttke H, Werner U, Feifel N, Eschenhagen T.Effect of the CYP2D6 genotype on metoprolol metabolism persists during long-term treatment. Pharmacogenetics 2002;12:465–472.

157. 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 meto-prolol: a prospective longitudinal study. Clin Pharmacol Ther 2009;85:269–272.

158. Yuan H, Huang Z, Yang G, Lv H, Sang H, Yao Y. Effects of polymorphism of thebeta(1) adrenoreceptor and CYP2D6 on the therapeutic effects of metoprolol.J Int Med Res 2008;36:1354–1362.

159. Bijl MJ, Visser LE, van Schaik RH, Kors JA, Witteman JC, Hofman A, Vulto AG,van Gelder T, Stricker BH. Genetic variation in the CYP2D6 gene is associatedwith a lower heart rate and blood pressure in beta-blocker users. Clin PharmacolTher 2009;85:45–50.

160. Swen JJ, Wilting I, de Goede AL, Grandia L, Mulder H, Touw DJ, de Boer A,Conemans JM, Egberts TC, Klungel OH, Koopmans R, van der Weide J,Wilffert B, Guchelaar HJ, Deneer VH. Pharmacogenetics: from bench to byte.Clin Pharmacol Ther 2008;83:781–787.

161. Reiter MJ. Cardiovascular drug class specificity: beta-blockers. Prog Cardiovasc Dis2004;47:11–33.

162. Petersen M, Andersen JT, Hjelvang BR, Broedbaek K, Afzal S, Nyegaard M,Borglum AD, Stender S, Kober L, Torp-Pedersen C, Poulsen HE. Associationof beta-adrenergic receptor polymorphisms and mortality in carvedilol-treatedchronic heart-failure patients. Br J Clin Pharmacol 2011;71:556–565.

163. Rigat B, Hubert C, Alhenc-Gelas F, Cambien F, Corvol P, Soubrier F. An inser-tion/deletion polymorphism in the angiotensin I-converting enzyme geneaccounting for half the variance of serum enzyme levels. J Clin Invest 1990;86:1343–1346.

164. Tiret L, Rigat B, Visvikis S, Breda C, Corvol P, Cambien F, Soubrier F. Evidence,from combined segregation and linkage analysis, that a variant of the angiotensinI-converting enzyme (ACE) gene controls plasma ACE levels. Am J Hum Genet1992;51:197–205.

165. Arnett DK, Boerwinkle E, Davis BR, Eckfeldt J, Ford CE, Black H. Pharmacoge-netic approaches to hypertension therapy: design and rationale for the Genetics

Overview of pharmacogenetics in CVD 175dD

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018

Page 16: A systematic review on pharmacogenetics in cardiovascular disease

of Hypertension Associated Treatment (GenHAT) study. Pharmacogenomics J2002;2:309–317.

166. Harrap SB, Tzourio C, Cambien F, Poirier O, Raoux S, Chalmers J, Chapman N,Colman S, Leguennec S, MacMahon S, Neal B, Ohkubo T, Woodward M. TheACE gene I/D polymorphism is not associated with the blood pressure and car-diovascular benefits of ACE inhibition. Hypertension 2003;42:297–303.

167. Bleumink GS, Schut AF, Sturkenboom MC, van Duijn CM, Deckers JW,Hofman A, Kingma JH, Witteman JC, Stricker BH. Mortality in patients withhypertension on angiotensin-I converting enzyme (ACE)-inhibitor treatment isinfluenced by the ACE insertion/deletion polymorphism. Pharmacogenet Geno-mics 2005;15:75–81.

168. Cambien F, Poirier O, Lecerf L, Evans A, Cambou JP, Arveiler D, Luc G, Bard JM,Bara L, Ricard S. Deletion polymorphism in the gene for angiotensin-convertingenzyme is a potent risk factor for myocardial infarction. Nature 1992;359:641–644.

169. Agema WR, Jukema JW, Zwinderman AH, van der Wall EE. A meta-analysis ofthe angiotensin-converting enzyme gene polymorphism and restenosis after per-cutaneous transluminal coronary revascularization: evidence for publication bias.Am Heart J 2002;144:760–768.

170. Schelleman H, Klungel OH, Witteman JC, Breteler MM, Yazdanpanah M,Danser AH, Hofman A, van Duijn CM, de Boer A, Stricker BH. AngiotensinogenM235T polymorphism and the risk of myocardial infarction and stroke amonghypertensive patients on ACE-inhibitors or beta-blockers. Eur J Hum Genet2007;15:478–484.

171. Schelleman H, Klungel OH, Witteman JC, Hofman A, van Duijn CM, de Boer A,Stricker BH. Pharmacogenetic interactions of three candidate gene polymorph-isms with ACE-inhibitors or beta-blockers and the risk of atherosclerosis. Br JClin Pharmacol 2007;64:57–66.

172. Su X, Lee L, Li X, Lv J, Hu Y, Zhan S, Cao W, Mei L, Tang YM, Wang D,Krauss RM, Taylor KD, Rotter JI, Yang H. Association between angiotensinogen,angiotensin II receptor genes, and blood pressure response to an angiotensin-converting enzyme inhibitor. Circulation 2007;115:725–732.

173. Brugts JJ, Isaacs A, Boersma E, van Duijn CM, Uitterlinden AG, Remme W,Bertrand M, Ninomiya T, Ceconi C, Chalmers J, MacMahon S, Fox K,Ferrari R, Witteman JC, Danser AH, Simoons ML, de Maat MP. Genetic deter-minants of treatment benefit of the angiotensin-converting enzyme-inhibitorperindopril in patients with stable coronary artery disease. Eur Heart J 2010;31:1854–1864.

174. Daly AK. Genome-wide association studies in pharmacogenomics. Nat Rev Genet2010;11:241–246.

175. Sissung TM, English BC, Venzon D, Figg WD, Deeken JF. Clinical pharmacologyand pharmacogenetics in a genomics era: the DMET platform. Pharmacogenomics2010;11:89–103.

176. Ridker PM, MacFadyen J, Libby P, Glynn RJ. Relation of baseline high-sensitivityC-reactive protein level to cardiovascular outcomes with rosuvastatin in the Jus-tification for Use of statins in Prevention: an Intervention Trial Evaluating Rosu-vastatin (JUPITER). Am J Cardiol 2010;106:204–209.

177. Swen JJ, Nijenhuis M, de Boer A, Grandia L, Maitland-van der Zee AH, Mulder H,Rongen GA, van Schaik RH, Schalekamp T, Touw DJ, van der Weide J, Wilffert B,Deneer VH, Guchelaar HJ. Pharmacogenetics: from bench to byte- an update ofguidelines. Clin Pharmacol Ther 2011;89:662–673.

178. Frueh FW, Amur S, Mummaneni P, Epstein RS, Aubert RE, DeLuca TM,Verbrugge RR, Burckart GJ, Lesko LJ. Pharmacogenomic biomarker informationin drug labels approved by the United States food and drug administration:prevalence of related drug use. Pharmacotherapy 2008;28:992–998.

179. Sangkuhl K, Klein TE, Altman RB. Clopidogrel pathway. Pharmacogenet Genomics2010;20:463–465.

J.J.W. Verschuren et al.175eD

ownloaded from

https://academic.oup.com

/eurheartj/article-abstract/33/2/165/437170 by guest on 16 Novem

ber 2018