12
STATE-OF-THE-ART PAPER Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina Pharmacogenetics primarily uses genetic variation to identify subgroups of patients who may respond differently to a certain medication. Since its first description, the field of pharmacogenetics has expanded to study a broad range of cardiovascular drugs and has become a mainstream research discipline. Three principle classes of pharmacogenetic markers have emerged: 1) pharmacokinetic; 2) pharmacodynamic; and 3) underlying disease mechanism. In the realm of cardiovascular pharmacogenetics, significant advances have identified markers in each class for a variety of therapeutics, some with a potential for improving patient outcomes. While ongoing clinical trials will determine if routine use of pharmacogenetic testing may be beneficial, the data today support pharmacogenetic testing for certain variants on an individualized, case-by-case basis. Our primary goal is to re- view the association data for the major pharmacogenetic variants associated with commonly used cardiovascu- lar medications: antiplatelet agents, warfarin, statins, beta-blockers, diuretics, and antiarrhythmic drugs. In addi- tion, we highlight which variants and in which contexts pharmacogenetic testing can be implemented by practicing clinicians. The pace of genetic discovery has outstripped the generation of the evidence justifying its clinical adoption. Until the evidentiary gaps are filled, however, clinicians may choose to target therapeutics to individual patients whose genetic background indicates that they stand to benefit the most from pharmaco- genetic testing. (J Am Coll Cardiol 2012;60:9–20) © 2012 by the American College of Cardiology Foundation “The right dose of the right drug to the right person” is one of the goals of pharmacogenomics and personalized medicine. The need for pharmacogenomics in clinical prac- tice is underscored, for example, by the improved ischemic outcomes with newer platelet P2RY12 receptor inhibitors which also have higher risk of adverse events compared to clopidogrel (1,2). Therefore, there is a critical need to target therapeutics to individual patients who stand to benefit the most and suffer the least. Principles of Pharmacogenetics A prerequisite for pharmacogenetics is heterogeneity in drug response. The definitions of drug response are varied and can include surrogate measurements measured in the laboratory (e.g., international normalized ratio [INR] for warfarin) or clinical endpoints (e.g., stent thrombosis for clopidogrel). A genetic basis for drug response is suggested when responses are similar within family members (and therefore are heritable) or significantly different in across ethnic backgrounds. Underlying genetic variation can be deter- mined either using a targeted approach where known variants in candidate genes are hypothesized to influence drug response or genome-wide association—an “unbiased” screen for common variants across the entire genome. Rare genetic variants missed by genome-wide association studies (GWAS) can be identified through sequencing candidate genes, exomes, or the entire genome in the case of rare drug-induced adverse events. Three broad classes of genetic variants influence drug response: 1) pharmacokinetic; 2) pharmacodynamic; and 3) those associated with the underlying disease mechanism (Table 1, Fig. 1). This review will discuss specific drugs, and in each case, we will apply these pharmacogenetic principles followed by potential clinical implications. Statins Statins (3-hydroxy-3methylglutaryl-coenzyme A reduc- tase [HMGCR] inhibitors) can elicit 4 types (at least) of “responses”: 1) low-density lipoprotein cholesterol (LDLc) lowering; 2) protection from cardiovascular events; 3) mus- culoskeletal side effects; and 4) statin adherence. LDLc lowering. The majority of patients respond with 30% to 50% LDLc reduction; however, there is wide (10% to 70%) variation (3). Two loci appear to affect LDLc lowering: HMGCR and APOE (Table 2). HMGCR. Carriers of a minor haplotype (defined by 3 single nucleotide polymorphisms [SNPs]: rs17244841, rs17238540, and rs3846662) of HMGCR experience a 5% From the Duke Institute for Genome Science & Policy and Center for Personalized Medicine, Duke University, Durham, North Carolina. Dr. Voora has reported he has no relationships relevant to the contents of this paper to disclose. Dr. Ginsburg is a scientific advisor to CardioDx. Manuscript received May 12, 2011; revised manuscript received January 5, 2012, accepted January 18, 2012. Journal of the American College of Cardiology Vol. 60, No. 1, 2012 © 2012 by the American College of Cardiology Foundation ISSN 0735-1097/$36.00 Published by Elsevier Inc. http://dx.doi.org/10.1016/j.jacc.2012.01.067

Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

Journal of the American College of Cardiology Vol. 60, No. 1, 2012© 2012 by the American College of Cardiology Foundation ISSN 0735-1097/$36.00

STATE-OF-THE-ART PAPER

Clinical Application ofCardiovascular PharmacogeneticsDeepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD

Durham, North Carolina

Pharmacogenetics primarily uses genetic variation to identify subgroups of patients who may respond differentlyto a certain medication. Since its first description, the field of pharmacogenetics has expanded to study a broadrange of cardiovascular drugs and has become a mainstream research discipline. Three principle classes ofpharmacogenetic markers have emerged: 1) pharmacokinetic; 2) pharmacodynamic; and 3) underlying diseasemechanism. In the realm of cardiovascular pharmacogenetics, significant advances have identified markers ineach class for a variety of therapeutics, some with a potential for improving patient outcomes. While ongoingclinical trials will determine if routine use of pharmacogenetic testing may be beneficial, the data today supportpharmacogenetic testing for certain variants on an individualized, case-by-case basis. Our primary goal is to re-view the association data for the major pharmacogenetic variants associated with commonly used cardiovascu-lar medications: antiplatelet agents, warfarin, statins, beta-blockers, diuretics, and antiarrhythmic drugs. In addi-tion, we highlight which variants and in which contexts pharmacogenetic testing can be implemented bypracticing clinicians. The pace of genetic discovery has outstripped the generation of the evidence justifying itsclinical adoption. Until the evidentiary gaps are filled, however, clinicians may choose to target therapeutics toindividual patients whose genetic background indicates that they stand to benefit the most from pharmaco-genetic testing. (J Am Coll Cardiol 2012;60:9–20) © 2012 by the American College of Cardiology Foundation

Published by Elsevier Inc. http://dx.doi.org/10.1016/j.jacc.2012.01.067

“The right dose of the right drug to the right person” isone of the goals of pharmacogenomics and personalizedmedicine. The need for pharmacogenomics in clinical prac-tice is underscored, for example, by the improved ischemicoutcomes with newer platelet P2RY12 receptor inhibitorswhich also have higher risk of adverse events compared toclopidogrel (1,2). Therefore, there is a critical need to targettherapeutics to individual patients who stand to benefit themost and suffer the least.

Principles of Pharmacogenetics

A prerequisite for pharmacogenetics is heterogeneity in drugresponse. The definitions of drug response are varied and caninclude surrogate measurements measured in the laboratory(e.g., international normalized ratio [INR] for warfarin) orclinical endpoints (e.g., stent thrombosis for clopidogrel).

A genetic basis for drug response is suggested whenresponses are similar within family members (and thereforeare heritable) or significantly different in across ethnicbackgrounds. Underlying genetic variation can be deter-mined either using a targeted approach where known

From the Duke Institute for Genome Science & Policy and Center for PersonalizedMedicine, Duke University, Durham, North Carolina. Dr. Voora has reported he hasno relationships relevant to the contents of this paper to disclose. Dr. Ginsburg is ascientific advisor to CardioDx.

Manuscript received May 12, 2011; revised manuscript received January 5, 2012,accepted January 18, 2012.

variants in candidate genes are hypothesized to influencedrug response or genome-wide association—an “unbiased”screen for common variants across the entire genome. Raregenetic variants missed by genome-wide association studies(GWAS) can be identified through sequencing candidategenes, exomes, or the entire genome in the case of raredrug-induced adverse events.

Three broad classes of genetic variants influence drugresponse: 1) pharmacokinetic; 2) pharmacodynamic; and3) those associated with the underlying disease mechanism(Table 1, Fig. 1). This review will discuss specific drugs, andin each case, we will apply these pharmacogenetic principlesfollowed by potential clinical implications.

Statins

Statins (3-hydroxy-3methylglutaryl-coenzyme A reduc-tase [HMGCR] inhibitors) can elicit 4 types (at least) of“responses”: 1) low-density lipoprotein cholesterol (LDLc)lowering; 2) protection from cardiovascular events; 3) mus-culoskeletal side effects; and 4) statin adherence.LDLc lowering. The majority of patients respond with30% to 50% LDLc reduction; however, there is wide (10%to 70%) variation (3). Two loci appear to affect LDLclowering: HMGCR and APOE (Table 2).

HMGCR. Carriers of a minor haplotype (defined by 3single nucleotide polymorphisms [SNPs]: rs17244841,

rs17238540, and rs3846662) of HMGCR experience a 5%
Page 2: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

taAbRdtpkmgdrafdtSamh

e(

S

10 Voora and Ginsburg JACC Vol. 60, No. 1, 2012Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20

to 20% reduced LDLc loweringwith pravastatin (4) or simvasta-tin (5) due to an alternativelyspliced HMGCR transcript thatproduces a version of HMGCRthat is less sensitive to simvasta-tin inhibition (6). This haplotypewas not identified in a GWASwith atorvastatin (7), and there-fore, whether this associationholds for other statins is unclear.

APOE. Two variants, rs7412and rs429358, define 3 haplotypes,namely, �2, �3, and �4, that areassociated with lipid, cognitive,and thrombotic traits. In general,the magnitude of LDLc loweringis greatest for carriers of �2, fol-lowed by �3, and �4 haplotypes,respectively. The influence ofAPOE haplotypes appears to beconsistent across variety of statin

ypes (4,7,8) and doses (8), and results in mild (�15%)ttenuations of LDLc lowering.dditional loci. Information on ABCB1 and ABCG2 cane found in the Online Appendix.eduction in cardiovascular events. Statins prevent car-iovascular disease by lowering LDLc and potential pleio-ropic effects. There are few genetic predictors of theseleiotropic benefits, but the rs20455 polymorphism ininesin-like protein 6 (KIF6, an underlying disease gene)ay be a candidate. Carriers of the risk allele may receive a

reater benefit from statin therapy compared to noncarriersespite equal LDLc, triglycerides, or C-reactive proteineduction (9,10). However, subsequent genome-wide meta-nalysis of large randomized placebo-controlled statin trialsound no evidence for association with coronary arteryisease (11) or any differential treatment benefit with statinherapy (12,13).tatin-induced musculoskeletal side effects. Statins havewell-defined safety profile, but come with a small risk ofusculoskeletal side effects (14). Genetic variants in the

epatic transporter, SLCO1B1, influence the risk of adverse

Abbreviationsand Acronyms

ACS � acute coronarysyndrome(s)

BP � blood pressure

CYP � cytochrome P450

GWAS � genome-wideassociation study

HR � heart rate

INR � internationalnormalized ratio

LDLc � low-densitylipoprotein cholesterol

LVEF � left ventricularejection fraction

MI � myocardial infarction

PCI � percutaneouscoronary intervention

RF � reduced function

SNP � single nucleotidepolymorphism

Sources of Pharmacogenetic VariationTable 1 Sources of Pharmacogenetic Variation

Category Description

Pharmacokinetic Variability in concentration of drug at site ofdrug effect

Pharmacodynamic Variability in drug ability to influence its targe

Underlying disease mechanisms Variability in disease being treated

vents (Table 2); it does not appear that cytochrome P450CYP) SNPs influence statin-induced side effects.

LCO1B1. The solute carrier organic anion transporterfamily, member 1B1 gene (SLCO1B1, also referred to asSLC21A6, OATP-C, or OATP1B1) harbors many geneticvariants, and each variant is numerically and sequentiallylabeled beginning with the unmutated copy of the gene, *1(referred to as “star 1”) followed by *2, *3, *4, and so forth.The *5 variant (rs4149056, Val174Ala) interferes with thelocalization of this transporter to the hepatocyte plasmamembrane (15) and leads to higher plasma statin concen-trations (16–18). In candidate gene and GWAS, carriers of*5 are at 4- to 5-fold increased risk of severe, creatine kinase(CK)- positive simvastatin-induced myopathy and 2- to3-fold increased risk of CK- negative myopathy (19,20).

In trials of randomly assigned statins as well as inobservational studies, the risk for myopathy with *5 dependson the statin type: the risk is greatest for simvastatin �atorvastatin � pravastatin, rosuvastatin, or fluvastatin (20–23).These effects parallel the influence of the *5 allele on theclearance of these statins (16–18,24) and thus appear to bestatin-specific.Adherence to statin therapy. Often, statin therapy ishampered by nonadherence. Although the genetics of ad-herence to statins has not been studied in any great depth,2 studies (a clinical trial and observational cohort) observedthat carriers of the SLCO1B1*5 allele have a higher rate ofstatin nonadherence (20,25).Clinical implications for statin pharmacogenetics. It isunlikely that genetic testing for statin efficacy will enterclinical care since the magnitude of associations is small(�10% to 15% differences in LDLc lowering), andphysicians can reasonably forecast the magnitude ofLDLc lowering based on statin type, dose, and baselineLDLc.

In contrast, statin-induced side effects and nonadherenceare less predictable. While the current level of evidencesurrounding SLCO1B1*5 may not support prospectivegenotyping at this time, the test is currently offered onconsumer-directed whole genome genotyping platforms(e.g., 23andMe, deCODEME, and so on). A potential strat-egy for prospective SLCO1B1*5 testing might offer carrierseither pravastatin or rosuvastatin or fluvastatin as first-line

Types of Genes Examples

Drug-metabolizing enzymesDrug transporters

Warfarin: CYP2C9Clopidogrel: CYP2C19Simvastatin: SLCO1B1Metoprolol: CYP2D6

Transmembrane receptorsIntracellular enzymes

Clopidogrel: P2RY12Simvastatin: HMGCRMetoprolol: ADBR1

Often downstream or independent of drug target Hydrochlorothiazide: ADD1

t

Simvastatin: APOE

Page 3: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

n(aPCeiPmm[ba(ai

ltiml(udt

ao((u

A

11JACC Vol. 60, No. 1, 2012 Voora and GinsburgJuly 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics

agents, which seem to depend the least on SLCO1B1 for theirclearance.

Thienopyridines

Thienopyridines are effective in patients with acute coronarysyndrome (ACS) and percutaneous coronary intervention(PCI); however, some patients remain at risk for death,myocardial infarction (MI), and stent thrombosis. Plateletfunction in response to clopidogrel is variable (26), heritable(27), and reduced inhibition predicts future events (28);therefore, clopidogrel is a prime candidate for pharmacoge-netics. Clopidogrel pharmacogenetics centers around 2 loci,CYP2C19 (pharmacokinetic) and ABCB1 (pharmacoki-

etic); other loci, CYP2C9 (pharmacokinetic), PON1pharmacokinetic), and P2RY12 (pharmacodynamic) maylso influence response (Table 3).latelet function in response to clopidogrel. CYP2C19.lopidogrel is an inactive prodrug activated by several

nzymes, including CYP2C19, to an active metabolite thatnhibits the platelet adenosine diphosphate receptor,2RY12 (29). CYP2C19 *2 (rs4244285) is the most com-on reduced-function (RF) variant; additional, rare variantsimic the *2 allele: *3 [rs4986893], *4 [rs28399504], *5

rs56337013]) (30). An individual person’s genotype cane characterized in 3 ways: 1) presence of at least 1 RFllele (carrier vs. noncarrier); 2) the number of RF allelesi.e., 0, 1, 2); or 3) the predicted, total CYP2C19 enzymaticctivity (Table 4). Carriers of *2—also referred to as

Figure 1 Categories of Major Cardiovascular Pharmacogenetic

Genetic variants that influence the response to cardiovascular agents fall into 3 b3) underlying disease mechanism.

ntermediate metabolizers or poor metabolizers—produce

ower active metabolite and have attenuated platelet inhibi-ion (31). In general, there is a gene-dose effect wherencreasing number of RF alleles (i.e., 0, 1, 2, or extensive

etabolizers, intermediate metabolizers, and poor metabo-izers) predicts a decreasing amount of platelet inhibition27,31,32). Carriers of another variant *17 (rs3758581,ltrametabolizers) exhibit increased CYP2C19 activity, pro-uce more active metabolite, and improved platelet inhibi-ion, in most reports (33,34).

Higher loading and maintenance doses (e.g., 1,200 mgnd 150 mg/day) appear, in part, to overcome the effectsf the *2 allele (30,32,35,36), although not completely37), and can require up to 300 mg/day (38). Ticlopidine39), prasugrel (31,40,41), and ticagrelor (42) all produceniform platelet inhibition in *2 carriers and noncarriers.

BCB1. The adenosine triphosphate-binding cassette, sub-family B (MDR/TAP), member 1 (ABCB1) gene encodesan apical membrane protein in enterocytes and hepatocytesand serves to reduce bioavailability. Clopidogrel appears to behandled by this transporter, and 3 SNPs—C1236T (rs1128503),G2677T (rs2032582), and C3435T (rs1045642)—capture thecommon genetic variation at this locus. Persons who carry aT allele at each SNPs (i.e., the T-T-T haplotype) producedreduced active metabolite (43) in an initial report. Despitethis initial observation, the link to reduced platelet inhibi-tion has been difficult to establish (27,30), although may bepresent for persons who carry 2 copies of the T-T-T

nts

ategories: 1) pharmacokinetic; 2) pharmacodynamic; and

Varia

road c

haplotype (44). As with CYP2C19 variants, prasugrel-

Page 4: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

acodifr(ds

A

inG

enet

icA

ssoc

iati

ons

Wit

hth

eR

espo

nse

toS

tati

nsab

le2

Mai

nG

enet

icA

ssoc

iati

ons

Wit

hth

eR

espo

nse

toSta

tins

Gen

eVar

iant

(s)

Ris

kA

llele

/H

aplo

type

Freq

uenc

ies

Sta

tin

Res

pons

e/Ty

peof

Stu

dyEf

fect

ofVar

iant

Sta

tin

Type

Ref

.#

Cau

casi

ans

Afr

ican

sA

sian

s

PO

E�2

,�3

,and

�4

hapl

otyp

esde

fined

byal

lele

sat

rs7

41

2an

drs

42

93

58

�2

:15

%–2

0%

*�3

:90

%–9

5%

�4

:20

%–3

0%

LDLc

low

erin

g/G

WA

S,C

G2

LDL

redu

ctio

nw

ith�3

vs.�

22

LDL

redu

ctio

nw

ith�4

vs.�

2Cla

ssef

fect

7,8

,14

5–1

47

MG

CR

H7

hapl

otyp

ede

fined

byal

lele

sat

rs1

72

44

84

1,r

s17

23

85

40

,an

drs

38

46

66

2

3%

6%

LDLc

low

erin

g/CG

2LD

Lre

duct

ion

inH

7ca

rrie

rsSim

vast

atin

4,5

LCO

1B

1*5

:rs4

14

90

56

15

%�

1%

13

%M

uscu

losk

elet

alsi

deef

fect

s/G

WA

S,C

G1

InCK

posi

tive

sym

ptom

s1

InCK

nega

tive

sym

ptom

sw

ithrisk

alle

le

Sim

vast

atin

,ato

rvas

tatin

19

,20

LCO

1B

1*5

:rs4

14

90

56

15

%�

1%

13

%N

onad

here

nce/

CG

1R

isk

ofno

nadh

eren

cew

ithrisk

alle

leSim

vast

atin

,ato

rvas

tatin

20

,25

pres

ents

Cau

casi

anpo

pula

tion.

G�

cand

idat

ege

neas

soci

atio

nst

udy;

CK

�cr

eatin

eki

nase

;GW

AS

�ge

nom

e-w

ide

asso

ciat

ion

stud

y;LD

Lc�

low

-den

sity

lipop

rote

inch

oles

tero

l.

12 Voora and Ginsburg JACC Vol. 60, No. 1, 2012Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20

induced platelet inhibition is not affected by T-T-T haplo-type (44).Additional loci. Using GWAS, investigators have unsuc-cessfully searched for additional variants for clopidogrel(27). An additional discussion of genetic variation atABCB1, P2RY12, CYP2C9, and PON1 can be found in theOnline Appendix.Clinical response to clopidogrel. CYP2C19. In parallelwith the platelet function data, the CYP2C19*2 allele isassociated with a graded risk of death, MI, or stroke.Carriers of 1 allele (intermediate metabolizers) have a�1.5-fold increased risk, and carriers of 2 alleles (poormetabolizers) experience a �1.8-fold increase. This patternalso extends to stent thrombosis as well with a �2.6- and�4-fold increased risk in those with 1 and 2 *2 alleles,respectively (32,45–49). Therefore, the CYP2C19 geneticssociations with platelet function are mirrored in thelinical response to clopidogrel in the setting of PCI. Thesebservations formed the foundation for updating the clopi-ogrel label by the Food and Drug Administration tonclude pharmacogenetic information. Similarly, the gain ofunction variant, CYP2C19*17, is associated with increasedisk of bleeding (50), and protection from ischemic events51) CYP2C19*2 carriers treated with prasugrel or ticagreloro not show a heightened risk of cardiovascular death, MI,troke, or stent thrombosis (40,52).

BCB1. Carriers of 2 copies of the ABCB1 T-T-T haplo-type treated with clopidogrel are at an increased risk forsubsequent death, MI, or stroke compared to persons whocarried none, thus mirroring the platelet function data(44,45,52). Genetic variation at the ABCB1 locus does notappear to influence prasugrel- or ticagrelor-treated patients(40,52).Clinical implications. The current state of evidence reflectsa strong and consistent association with the CYP2C19*2 alleleand an increased risk for cardiovascular outcomes in ACS/PCI patients treated with clopidogrel, although not othersettings (51). Alternative antiplatelet agents (i.e., prasugreland ticagrelor) mitigate the adverse risk of CYP2C19*2-associated adverse events. Although CYP2C19 genotyping iscommercially available, whether genotype-guided therapywill improve outcomes or reduce costs is unknown. Con-sensus statements (53) currently do not recommend routinetesting. However, there is sufficient evidence to supportphysicians who may choose to pursue CYP2C19*2 testing inselected patients: 1) for diagnosis in patients with compli-cations of clopidogrel therapy such as stent thrombosis incompliant clopidogrel users; or 2) for the choice of dualantiplatelet therapy in the ACS/PCI setting where thephysician believes that additional information regardingthe risk/benefit profile for clopidogrel will influence thechoice of drug therapy (54). Outside of these scenarios,the ACS/PCI setting, or situations where there are ampledata to guide drug choice (e.g., ST-segment elevation MI

[55], diabetic patients [56], age �75 years, or priorM

a T A H S S

*Re C

Page 5: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

pascsilcacr

A

A(taoHacs(vf

ipgd

pmr(atcahp

u(yshvocCppf

W

TCaWs

eviatio

13JACC Vol. 60, No. 1, 2012 Voora and GinsburgJuly 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics

transient ischemic attack/stroke), there is minimal ratio-nale to support CYP2C19 testing.

Based on the available data, it is reasonable that if aerson is found to be a poor metabolizer (Table 4), then anlternative to clopidogrel, such as prasugrel or ticagrelor,hould be considered. This seems preferable over increasedlopidogrel doses, which have not shown benefit overtandard dose clopidogrel (57–59). The greatest uncertaintys in the intermediate metabolizer group (i.e., those with 1oss of function allele) and is where integration of otherlinical risk factors such as diabetes, body mass index, cost,nd other bleeding and thrombosis risk factors need to beonsidered in determining the therapy with the optimalisk:benefit profile.

spirin

spirin irreversibly inhibits prostaglandin G/H synthase 1PTGS1, or COX-1) and the conversion of arachidonic acido thromboxane. With adequate dosing and compliance,spirin is capable of completely inhibiting COX-1 in �99%f persons; thus, true “aspirin resistance” is rare (60).owever, alternate agonists such as adenosine diphosphate

nd collagen can produce robust aggregation in the face ofomplete COX-1 inhibition (61), a response that demon-trates wide interindividual variability (62), heritability63,64), and at high levels, association with future cardio-ascular events (65,66), therefore making aspirin a candidateor pharmacogenetic discovery. Because aspirin uniformly

Main Genetic Associations With the Response to ClopidogrelTable 3 Main Genetic Associations With the Response to Clop

Gene Variant(s)

Risk Allele/Haplotype Frequencies

Drug RCaucasians Africans Asians

CYP2C19 *2 (rs4244285) 16% 14% 27% Drug concrecurreGWAS

CYP2C19 *17 (rs3758581) 5% 10% 6% Drug concbleedin

ABCB1 T-T-T haplotype defined byT allele at C1236T(rs1128503), G2677T(rs2032582), andC3435T (rs1045642)

71% 12% 65% Drug concrecurre

MI � myocardial infarction; DMET � drug metabolizing enzyme and transporter panel; other abbr

CYP2C19 Diplotype Classification*Table 4 CYP2C19 Diplotype Classification*

Alleles Classification

*1 and *1 Extensive metabolizer

*2/*3/*4/*5/*6 and *1/*17 Intermediate metabolizer

*2/*3/*4/*5/*6 and *2/*3/*4/*5/*6 Poor metabolizer

*1/*17 or *17/*17 Ultrametabolizer

a*Adapted from Ingelman-Sundberg et al. (147) and Scott et al. (148).

nhibits its target, COX-1, there are no pharmacokinetic orharmacodynamic genetic considerations. Instead, investi-ators have focused on platelet function loci (underlyingisease) and LPA (underlying disease).Variants in several platelet genes, namely, PEAR1,

ITGB3, VAV3, GPVI, F2R, and GP1BA, are associated withlatelet function in response to aspirin (67–70). One withoderate evidence is rs5918 in ITGB3 where carriers of the

isk allele have heightened platelet function on aspirin71–73). Recent GWAS of platelet function in response tospirin have identified additional genetic loci that have yeto be replicated (74). Finally, the most robust associationomes from a large study that found carriers of the minorllele for an intronic SNP, rs12041331, in PEAR1 haveigher PEAR1 platelet protein content and heightenedlatelet function in response to aspirin (75).The link to an increased risk of clinical events in aspirin

sers has not been established despite several large studies76,77), although the PEAR1 variant rs12041331 has notet been tested. An uncommon variant in LPA (rs3798220)eems to modify the protective effects of aspirin: carriersad a more than 2-fold reduction in the risk for cardio-ascular disease with aspirin, whereas noncarriers (�95%f Caucasians) had none in a large placebo-controlledlinical trial (78).linical implications. Because of the lack of consistent orreliminary associations with the variants described in thereceding text, there is currently no role for genetic testingor aspirin.

arfarin

hree loci contribute to the heterogeneity in response:YP2C9 (pharmacokinetic), VKORC1 (pharmacodynamic),nd CYP4F2 (pharmacodynamic) (Table 5).

arfarin dose requirements. CYP2C9. CYP2C9 is re-ponsible for S-warfarin (the more active enantiomer) clear-

el

se/Type of StudyEffect of VariantWith Risk Allele Ref. #

ion, platelet function,stent thrombosis/, CG

2Active metabolite concentration2Inhibition of platelet function1Risk of recurrent MI, stroke,

stent thrombosisGraded risk with 0, 1, or 2 alleles

31,32,45–47,52

ion, platelet function, 1Active metabolite concentration1Inhibition of platelet function1Risk bleeding

32,33,50

ion, platelet function,stroke, death/CG

2Active metabolite concentration2No change in inhibition of

platelet function1Risk of recurrent MI, stroke,

death (in carriers of 2 T-T-Thaplotypes)

27,30,43,45,48,49,52

ns as in Table 2.

idogr

espon

entratnt MI,

, DMET

entratg/CG

entratnt MI,

nce. The 2 most common reduced-function variants are

Page 6: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

et�r

CwAaC

A

taatsctcd“3w

D

BgaGeuCweppsCag

14 Voora and Ginsburg JACC Vol. 60, No. 1, 2012Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20

*2 (rs1799853) and *3 (rs1057910). The CYP2C9*2 allelereduces warfarin clearance by 30% and the *3 allele by90% (79), which translate into 19% and 33% reductionsper allele in warfarin dose requirements compared tononcarriers (80).

VKORC1. Vitamin K epoxide reductase complex, subunit 1(VKORC1) is warfarin’s target (81,82), and resequencingthis gene identified 2 haplotypes (A and B defined by thealleles at 5 SNPS: rs7196161, rs9923231, rs9934438,rs8050894, and rs2359612) that influence VKORC1 genexpression and predict warfarin dose requirements (83). Ofhese 5 SNPs, an A allele at rs9923231 (also referred to as

1639 G�A and the 3673 SNP) in the promoter region isesponsible for reducing: VKORC1 gene expression (84).

Therefore, carriers of an A allele at rs9923231 (or the Ahaplotype) require a 30% per allele reduction in warfarindose requirements (85).

CYP4F2. Several GWAS have not only confirmed theobservations above but also have identified a novel associa-tion between rs2108622 in CYP4F2 and reduced hepatic

YP4F2, higher levels of hepatic vitamin K, and higherarfarin dose requirements (86–90).dditional loci. Additional information regarding CALU

nd GGCX can be found in the Online Appendix.linical response to warfarin. RETROSPECTIVE STUDIES.

Standard dosing algorithms (i.e., a 5 mg or 10 mg loadingdose followed by titration based on the INR) lead to anincreased risk of an out-of-range INR (�4.0) or a delay inthe time-to-therapeutic range INR in carriers of theCYP2C9*2 or CYP2C9*3 alleles (91,92), the A haplotype or

allele of the �1639 SNP in VKORC1 (93), or the T allelein CYP4F2 (94). Importantly, only variants in CYP2C9 havebeen linked to the risk of bleeding (91,92).

PROSPECTIVE STUDIES OF TAILORED WARFARIN THERAPY.

Small randomized pilot studies, parallel cohort studies, andsingle arm studies with or without historical controls haveprospectively explored genetically guided warfarin therapy.

Main Genetic Associations With the Response to WarfarinTable 5 Main Genetic Associations With the Response to Warf

Gene Variant(s)

Risk Allele Frequencies

Caucasians Africans Asians

CYP2C9 *2 (rs1799853)*3 (rs1057910)

*2: 10%*3: 6%

�1%�1%

�1%4%

Dr

VKORC1 �1639 (rs9923231)* 60% 98% 2% Wa

CYP4F2 rs2108622 23% 6% 25% Wa

*Carriers of the risk, A, allele are also carriers of the A haplotype.INR � international normalized ratio; other abbreviations as in Tables 2 and 3.

Whereas some studies suggest a benefit over standard i

therapy (95–97), others failed to show any significantadvantage (98) except for smaller and fewer dosing changesand INR measurements (99). Definitive clinical trials areongoing to assess the benefit of incorporating genetics intowarfarin therapy.Clinical implications. There are strong genetic associa-tions surrounding CYP2C9, VKORC1, and CYP4F2 variantshat influence the response to warfarin. Commercial testingnd algorithms that assist in the interpretation of genotypesre available. Therefore, there is evidence to justify and toolso enable genotype-guided warfarin therapy. Until largecale trials demonstrate a benefit for routine testing, physi-ians may choose to pursue testing in selected patients whohey feel may benefit by: 1) diagnosing those with compli-ations from warfarin therapy (e.g., hemorrhage); 2) pre-icting dose for those at high risk of bleeding (e.g.,triple-therapy” with aspirin, clopidogrel, and warfarin); or) weighing the costs of newer anticoagulants againstarfarin.

iuretics

lood pressure–lowering response. The adducin 1 (alpha)ene (ADD1, underlying disease) is implicated in animalnd human sodium handling (100), and persons with aly460Trp substitution are more salt sensitive and have

nhanced blood pressure (BP) lowering with thiazide di-retics (100).linical outcomes. In an observational study, patientsith Trp460 who were treated with thiazide diuretics

xperienced a greater protection from MI and stroke thanatients with Gly460 (101). However, this difference in therotective effects of diuretics could not be replicated ineveral larger studies (102–104).linical implications. Given the inconsistent associations

nd lack of a commercially available test, the use of thisenetic variant in guiding treatment options in hypertension

g Response/ype of Study

Effect of Variant inCarriers of Risk Allele Ref. #

centration, warfarinequirements,range INR values,rhage/GWAS, CG

2Clearance of S-warfarin2Dose requirements1Risk of out-of-range INR values1Time to stable, therapeutic INR1Risk of hemorrhage

79,80,88,89,91–93

dose requirements,range INR values/e (human/animal),, CG

2Dose requirements1Risk of out-of-range INR values1Time to stable, therapeutic INR

81–83,88,89

dose requirements/, DMET

1Dose requirements 87–89

arin

DruT

ug condose rout-of-hemor

rfarinout-of-linkagGWAS

rfarinGWAS

s limited.

Page 7: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

t

A

a

mcst

A

Ghrrld

G

inG

enet

icA

ssoc

iati

ons

Wit

hth

eR

espo

nse

toB

eta-

Adr

ener

gic

Rec

epto

rA

ntag

onis

tsab

le6

Mai

nG

enet

icA

ssoc

iati

ons

Wit

hth

eR

espo

nse

toB

eta-

Adr

ener

gic

Rec

epto

rA

ntag

onis

ts

Gen

eV

aria

nt(s

)

Ris

kA

llele

Freq

uenc

ies

Dru

gR

espo

nse/

Type

ofStu

dyEf

fect

ofVar

iant

inC

arrier

sof

Ris

kA

llele

Bet

a-B

lock

erTy

peR

ef.

#C

auca

sian

sA

fric

ans

Asi

ans

YP2

D6

*4(r

s38

92

09

7)

18

%2

3%

Dru

gco

ncen

trat

ion,

HR

and

BP

redu

ctio

n/CG

2Cle

aran

ce1

Red

uctio

nin

BP

and

HR

1R

isk

ofbr

adyc

ardi

a

Met

opro

lol

10

5,1

06

,12

7,1

28

DR

B1

Ser

49

Gly

(rs1

80

12

52

)2

0%

�1

%�

1%

HR

,BP

redu

ctio

n/CG

2R

educ

tion

inB

Pan

dH

RM

etop

rolo

l1

12

DR

B1

Arg

38

9G

ly(r

s18

01

25

3)

31

%4

1%

20

%H

R,i

mpr

ovem

ent

inLV

EF,

BP

redu

ctio

n,ho

spita

lizat

ion

orde

ath

inCH

F/CG

2R

educ

tion

inB

Pan

dH

R2

Impr

ovem

ent

inLV

EFM

etop

rolo

l(B

P/H

R/L

VEF)

Car

vedi

lol(

LVEF

)B

ucin

dolo

l(ho

spita

lizat

ion/

deat

h)

11

3,1

15

,11

6

bloo

dpr

essu

re;C

G�

cand

idat

ege

neas

soci

atio

nst

udy;

HR

�he

art

rate

;LVE

F�

left

vent

ricu

lar

ejec

tion

frac

tion.

15JACC Vol. 60, No. 1, 2012 Voora and GinsburgJuly 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics

Beta-Blockers

Beta-adrenergic receptor antagonists (or beta-blockers) area diverse class of agents that primarily antagonize the beta-1adrenergic receptor, encoded by ADBR1. Accordingly,changes in heart rate (HR), BP, and left ventricular ejectionfraction (LVEF) are considered the surrogate responses inplace of clinical responses such as protection from MI,death, or heart failure. Variation in CYP2D6 (pharmacoki-netic) and ADRB1, ADRB2, and GRK5 (all pharmacody-namic) have received the most attention (Table 6).Heart rate and blood pressure reduction. CYP2D6. Beta-blockers are substrates for CYP, and metoprolol is exten-sively metabolized by hepatic CYP2D6. The most commonRF variant CYP2D6*4 (rs3892097) results in the absence ofCYP2D6 activity. Carriers have a higher systemic exposureto metoprolol (105), which translates into a greater reduc-tion in HR and BP (106,107). Beta-blockers such asatenolol and carvedilol (108) do not require CYP2D6 forheir metabolism.

DRB1. Ethnic differences in the dose response for pro-pranolol (109) motivated pharmacodynamic genetic inves-tigations of beta-blockers. Two variants in ADBR1, theSer49Gly (rs1801252) and Arg389Gly (rs1801253), lead toimpaired down-regulation (110) and higher signal transduc-tion, respectively (111). Therefore, carriers of either varianthave enhanced, beta-1-receptor activity and more beta-blocker sensitivity. Healthy volunteers and patients withhypertension who carry 2 Arg389 variants have a greaterHR (112) or BP (113) reduction mainly with metoprolol,lthough not with all beta-blockers (114).

In patients with systolic heart failure treated with eitheretoprolol or carvedilol (115), but not bucindolol (116),

arriers of 2 copies of the ADBR1 Arg389 variant hadignificantly greater improvements in LVEF compared tohe Gly389 carriers.

DRB2. Genetic variation in the beta-2 adrenergic receptor(ADBR2) centers around 2 variants: Arg16Gly (rs1042713) and

lu27Gln (rs1042714). Receptors with Gly16 versus Arg16ave enhanced down-regulation of ADBR2. In contrast,eceptors with Glu27 appear to be resistant to down-egulation (117). Extension to variation in BP or HRowering in response to beta-blocker therapy has not beenemonstrated (114).

RK5. Downstream of the beta-1-adrenergic receptor areG-protein coupled receptor kinases responsible for desensi-tization of the beta-1-adrenergic receptor. A Glu41Leugenetic variant in 1 of these kinases, G protein-coupledreceptor kinase 5 (GRK5), is more prevalent in AfricanAmericans. The Leu41 more effectively uncoupledisoproterenol-stimulated responses than GRK5-Q41, thusproducing a pharmacological-like “beta-blockade” in mice(118), although with no differences in atenolol-induced HR

reduction in humans (119). M

a T C A A

BP

Page 8: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

16 Voora and Ginsburg JACC Vol. 60, No. 1, 2012Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20

Clinical benefit in patients with cardiovascular disease.ADRB1. In heart failure patients, when compared to pla-cebo, ADRB1 Arg389 homozygotes had a greater reductionin the time to first hospitalization or death when treatedwith bucindolol (116) but not carvedilol or metoprolol(120–125) compared to Gly389 carriers. Whether thesedifferences are due to drug specific effects (i.e., bucindolol vs.metoprolol) or the play of chance has not been adequatelytested. In patients with chronic coronary artery diseaserandomly assigned to verapamil or atenolol, carriers of atleast 1 copy of the Ser49/ Arg389 haplotype had a 9- versus2-fold worsened prognosis when assigned to verapamilversus atenolol (126) despite equivalent BP and HR controlin both groups.

ADRB2. One study examined the influence of geneticvariation at ADRB2 and its influence on ACS patients andfound those homozygous for both the Arg16 and Glu27alleles had a 20% rate of subsequent death versus 6% ofthose homozygous for both Gly16 and Gln27 (125). Thistrend has been observed in some studies of patients withheart failure (124), although not all (122,123,126).

GRK5. Extension of the associations of the Glu41Leuvariant to clinical outcomes of patients was demonstrated in1 study where Leu41 carriers exhibited improvement in sur-vival compared to Glu41 carriers (118), although not inanother, larger study (121).Adverse events. Metoprolol-induced adverse events (e.g.,bradycardia) are associated with CYP2D6*4 in 2 studies,each numbering �1,000 treated patients (127,128), al-though not in smaller studies (129,130).Clinical implications. In general, carriers of the Arg389variant have: 1) enhanced reduction in HR and BP; 2) largerimprovements in LVEF; and 3) longer survival whentreated with chronic beta-blocker therapy compared topersons with the Gly389 variant. Although it is unlikely thatbeta-blocker therapy will ever be withheld for carriers of theGly389 variant, a potential application of these findingswould be to consider advanced heart failure therapies (e.g.,left ventricular assist devices, biventricular pacing, or trans-plantation) at an earlier stage in patients with the Gly389variant.

Because certain beta-blockers such as atenolol and carve-dilol are minimally handled by CYP2D6 (131), these may bereasonable alternates for carriers of CYP2D6*4 withmetoprolol-induced bradycardia. Commercial testing forCYP2D6*4 is available (e.g., Labcorp).

Antiarrhythmic Drugs

Digoxin and calcium-channel blockers. Many antiar-rhythmics are known ABCB1 (described in preceding text)substrates including verapamil, diltiazem, and digoxin. Ge-netic variation in ABCB1 is inconsistently associated withaltered pharmacokinetic profiles (132–134), and there are

no reports of different clinical outcomes.

Procainamide. Procainamide is rapidly converted by acet-ylation into N-acetylprocainamide (NAPA, an active me-tabolite) by hepatic acetyltransferases (NAT2, primarily).Variability in hepatic acetylation capacity has long beenobserved (135), and “slow acetylators” produce less NAPA.Common genetic variants that decrease NAT2 activity are*5/rs1801280, *6/rs1799930, *7/rs1799931, and *14/rs1801279 (136,137). Although efficacy does not appear tobe related to variation in its pharmacokinetics, the inductionof drug-induced lupus-associated autoantibodies (138,139),but not symptomatic, drug-induced lupus (138,139) hasbeen linked to the slow acetylator status.Propafenone. The hepatic hydroxylation by CYP2D6 ofpropafenone into 5-hydroxypropafenone demonstrates wideinterindividual variability (140). Carriers of CYP2D6*4 (areduced function allele) have reduced propafenone clearancecompared to noncarriers (140–142).Antiarrhythmic efficacy. At low doses of propafenone,CYP2D6*4 carriers have greater reduction in exercise- orisoproterenol-induced HR compared to noncarriers (143),although not with higher doses or without the “stress” ofexercise/isoproterenol (140,142,143). Further, CYP2D6*4carriers have enhanced suppression of atrial and ventriculararrhythmias compared to noncarriers in some studies(141,144), although not in all (140,142) Therefore, there areinsufficient data to make any conclusions regardingCYP2D6 genotype and antiarrhythmic efficacy.Toxicity. Central nervous system side effects withpropafenone, as well as excessive beta-blockade, are correlatedwith a higher concentration of systemic propafenone and,accordingly, the CYP2D6 genotype (140).Clinical implications. Antiarrhythmic drug pharmacoge-netics represents a mixed collection of associations that donot sufficiently translate to consistent associations of mean-ingful clinical outcomes. Therefore, there is currently norole for pharmacogenetic testing in the clinical use of thesemedications.

Future Directions

The data to date support many common variants that alterthe pharmacologic properties of cardiovascular agents.Newer medications such as prasugrel, ticagrelor, and apxi-ban as well as older medications such as enalapril, spirono-lactone, and angiotensin-receptor blockers that have variableclinical effects are also candidates for a pharmacogenetic approach.Novel systems approaches may elucidate additional deter-minants of drug response. Broad “omics” approaches haveseveral advantages over traditional genetics-based research,for example, 1) responding to the environment, and 2)elucidating how the activity of entire pathways, instead ofindividual genes, influence drug response.

The field of pharmacogenomics would benefit from thedevelopment of thresholds of evidence for testing andcoverage by insurance. With the expanding number and

complexity of “-omic” markers of drug response is an equally
Page 9: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

17JACC Vol. 60, No. 1, 2012 Voora and GinsburgJuly 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics

profound evidentiary gap between association studies andthose that demonstrate clinical utility. It may be unrealisticto wait (or to require) a prospective, events-driven random-ized controlled trial of each drug-marker combination. Evenwhen such trials are conducted, the results may be madeobsolete by new observations and/or approval of new drugs.Therefore, genetic substudies of clinical trials and registriesmay become the highest levels of evidence for the potentialbenefits of using pharmacogenetic testing. Such standardsare common in many fields of medicine where there is oftena lack of randomized clinical trial data. Observationalstudies and comparative effectiveness research will lay thefoundation for broad-based recommendations and transla-tion of pharmacogenetic observations into a clinical para-digm of personalized medicine to improve health outcomes.

Reprint requests and correspondence: Dr. Deepak Voora or Dr.Geoffrey S. Ginsburg, Institute for Genome Sciences & Policy,Center for Genomic Medicine, 101 Science Drive, Duke Univer-sity, DUMC Box 3382, Durham, North Carolina 27708. E-mail:[email protected] or [email protected].

REFERENCES

1. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrelin patients with acute coronary syndromes. N Engl J Med 2009;361:1045–57.

2. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versusclopidogrel in patients with acute coronary syndromes. N Engl J Med2007;357:2001–15.

3. Simon JA, Lin F, Hulley SB, et al. Phenotypic predictors of responseto simvastatin therapy among African-Americans and Caucasians:the Cholesterol and Pharmacogenetics (CAP) study. Am J Cardiol2006;97:843–50.

4. Chasman DI, Posada D, Subrahmanyan L, Cook NR, Stanton VPJr., Ridker PM. Pharmacogenetic study of statin therapy and choles-terol reduction. JAMA 2004;291:2821–7.

5. Krauss RM, Mangravite LM, Smith JD, et al. Variation in the3-hydroxyl-3-methylglutaryl coenzyme a reductase gene is associatedwith racial differences in low-density lipoprotein cholesterol responseto simvastatin treatment. Circulation 2008;117:1537–44.

6. Medina MW, Gao F, Ruan W, Rotter JI, Krauss RM. Alternativesplicing of 3-hydroxy-3-methylglutaryl coenzyme A reductase isassociated with plasma low-density lipoprotein cholesterol responseto simvastatin. Circulation 2008;118:355–62.

7. Thompson JF, Hyde CL, Wood LS, et al. Comprehensive whole-genome and candidate gene analysis for response to statin therapy inthe Treating to New Targets (TNT) cohort. Circ Cardiovasc Genet2009;2:173–81.

8. Voora D, Shah SH, Reed CR, et al. Pharmacogenetic predictors ofstatin-mediated low-density lipoprotein cholesterol reduction anddose response. Circ Cardiovasc Genet 2008;1:100–6.

9. Iakoubova OA, Sabatine MS, Rowland CM, et al. Polymorphism inKIF6 gene and benefit from statins after acute coronary syndromes:results from the PROVE IT-TIMI 22 study. J Am Coll Cardiol2008;51:449–55.

10. Iakoubova OA, Tong CH, Rowland CM, et al. Association of theTrp719Arg polymorphism in kinesin-like protein 6 with myocardialinfarction and coronary heart disease in 2 prospective trials: theCARE and WOSCOPS trials. J Am Coll Cardiol 2008;51:435–43.

11. Assimes TL, Holm H, Kathiresan S, et al. Lack of associationbetween the Trp719Arg polymorphism in kinesin-like protein-6 andcoronary artery disease in 19 case-control studies. J Am Coll Cardiol2010;56:1552–63.

12. Hopewell JC, Parish S, Clarke R, et al. No impact of KIF6 genotype

on vascular risk and statin response among 18,348 randomized

patients in the Heart Protection Study. J Am Coll Cardiol2011;57:2000–7.

13. Ridker PM, MacFadyen JG, Glynn RJ, Chasman DI. Kinesin-LikeProtein 6 (KIF6) polymorphism and the efficacy of rosuvastatin inprimary prevention/clinical perspective. Circ Cardiovasc Genet 2011;4:312–7.

14. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety ofcholesterol-lowering treatment: prospective meta-analysis of datafrom 90,056 participants in 14 randomised trials of statins. Lancet2005;366:1267–78.

15. Kameyama Y, Yamashita K, Kobayashi K, Hosokawa M, Chiba K.Functional characterization of SLCO1B1 (OATP-C) variants,SLCO1B1*5, SLCO1B1*15 and SLCO1B1*15�C1007G, by usingtransient expression systems of HeLa and HEK293 cells. Pharma-cogenet Genomics 2005;15:513–22.

16. Pasanen MK, Neuvonen M, Neuvonen PJ, Niemi M. SLCO1B1polymorphism markedly affects the pharmacokinetics of simvastatinacid. Pharmacogenet Genomics 2006;16:873–9.

17. Pasanen MK, Fredrikson H, Neuvonen PJ, Niemi M. Differenteffects of SLCO1B1 polymorphism on the pharmacokinetics ofatorvastatin and rosuvastatin. Clin Pharmacol Ther 2007;82:726–33.

18. Niemi M, Pasanen MK, Neuvonen PJ. SLCO1B1 polymorphism andsex affect the pharmacokinetics of pravastatin but not fluvastatin[ast].Clin Pharmacol Ther 2006;80:356–66.

19. Link E, Parish S, Armitage J, et al. SLCO1B1 variants andstatin-induced myopathy—a genomewide study. N Engl J Med2008;359:789–99.

20. Voora D, Shah SH, Spasojevic I, et al. The SLCO1B1*5 geneticvariant is associated with statin-induced side effects. J Am CollCardiol 2009;54:1609–16.

21. Puccetti L, Ciani F, Auteri A. Genetic involvement in statinsinduced myopathy. Preliminary data from an observational case-control study. Atherosclerosis 2010;211:28–9.

22. Bruckert E, Hayem G, Dejager S, Yau C, Begaud B. Mild tomoderate muscular symptoms with high-dosage statin therapy inhyperlipidemic patients—the PRIMO study. Cardiovasc Drugs Ther2005;19:403–14.

23. Jacqueline Suk Danik, MacFadyen J, Nyberg F, Ridker P. Lack ofassociation between polymorphisms in the slco1b1 gene and clinicalmyalgia following rosuvastatin therapy. J Am Coll Cardiol 2012;59:1621, doi:10.1016/S0735-1097(12)61622-2.

24. Choi JH, Lee MG, Cho JY, Lee JE, Kim KH, Park K. Influence ofOATP1B1 genotype on the pharmacokinetics of rosuvastatin inKoreans. Clin Pharmacol Ther 2007;83:251–7.

25. Donnelly LA, Doney ASF, Tavendale R, et al. Common nonsyn-onymous substitutions in SLCO1B1 predispose to statin intolerancein routinely treated individuals with type 2 diabetes: a Go-DARTSstudy. Clin Pharmacol Ther 2010;89:210–6.

26. Gurbel PA, Bliden KP, Guyer K, et al. Platelet reactivity in patientsand recurrent events post-stenting: results of the PREPARE POST-STENTING study. J Am Coll Cardiol 2005;46:1820–6.

27. Shuldiner AR, O’Connell JR, Bliden KP, et al. Association ofcytochrome P450 2C19 genotype with the antiplatelet effect andclinical efficacy of clopidogrel therapy. JAMA 2009;302:849–57.

28. Breet NJ, van Werkum JW, Bouman HJ, et al. Comparison ofplatelet function tests in predicting clinical outcome in patientsundergoing coronary stent implantation. JAMA 2010;303:754–62.

29. Savi P, Pereillo JM, Uzabiaga MF, et al. Identification and biologicalactivity of the active metabolite of clopidogrel. Thromb Haemost2000;84:891–6.

30. Gladding P, Webster M, Zeng I, et al. The pharmacogenetics andpharmacodynamics of clopidogrel response: an analysis from thePRINC (Plavix Response in Coronary Intervention) trial. J Am CollCardiol Intv 2008;1:620–7.

31. Brandt JT, Close SL, Iturria SJ, et al. Common polymorphisms ofCYP2C19 and CYP2C9 affect the pharmacokinetic and pharmaco-dynamic response to clopidogrel but not prasugrel. J Thromb Hae-most 2007;5:2429–36.

32. Simon T, Bhatt DL, Bergougnan L, et al. Genetic polymorphismsand the impact of a higher clopidogrel dose regimen on activemetabolite exposure and antiplatelet response in healthy subjects.Clin Pharmacol Ther 2011;90:287–95.

33. Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 polymor-

phisms and response to clopidogrel. N Engl J Med 2009;360:354–62.
Page 10: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

18 Voora and Ginsburg JACC Vol. 60, No. 1, 2012Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20

34. Frére C, Cuisset T, Gaborit B, Alessi MC, Hulot JS. TheCYP2C19*17 allele is associated with better platelet response toclopidogrel in patients admitted for non-ST acute coronary syn-drome. J Thromb Haemost 2009;7:1409–11.

35. Hulot JS, Wuerzner G, Bachelot-Loza C, et al. Effect of an increasedclopidogrel maintenance dose or lansoprazole co-administration onthe antiplatelet response to clopidogrel in CYP2C19-genotypedhealthy subjects. J Thromb Haemost 2010;8:610–3.

36. Gladding P, White H, Voss J, et al. Pharmacogenetic testing forclopidogrel using the rapid INFINITI analyzer: a dose-escalationstudy. J Am Coll Cardiol Intv 2009;2:1095–101.

37. Jeong Y-H, Kim I-S, Park Y, et al. Carriage of cytochrome 2C19polymorphism is associated with risk of high post-treatment plateletreactivity on high maintenance-dose clopidogrel of 150 mg/day:results of the ACCEL-DOUBLE (Accelerated Platelet Inhibition bya Double Dose of Clopidogrel According to Gene Polymorphism)study. J Am Coll Cardiol Intv 2010;3:731–41.

38. Pena A, Collet J-P, Hulot J-S, et al. Can we override clopidogrelresistance? Circulation 2009;119:2854–7.

39. Maeda A, Ando H, Asai T, et al. Differential impacts of CYP2C19gene polymorphisms on the antiplatelet effects of clopidogrel andticlopidine. Clin Pharmacol Ther 2011;89:229–33.

40. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P450 geneticpolymorphisms and the response to prasugrel. Relationship to phar-macokinetic, pharmacodynamic, and clinical outcomes. Circulation2009;119:2553–60.

41. Varenhorst C, James S, Erlinge D, et al. Genetic variation ofCYP2C19 affects both pharmacokinetic and pharmacodynamic re-sponses to clopidogrel but not prasugrel in aspirin-treated patientswith coronary artery disease. Eur Heart J 2009;30:1744–52.

42. Tantry US, Bliden KP, Wei C, et al. First analysis of the relationbetween CYP2C19 genotype and pharmacodynamics in patientstreated with ticagrelor versus clopidogrel/clinical perspective. CircCardiovasc Genet 2010;3:556–66.

43. Taubert D, von Beckerath N, Grimberg G, et al. Impact ofP-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther2006;80:486–501.

44. Mega JL, Close SL, Wiviott SD, et al. Genetic variants in ABCB1and CYP2C19 and cardiovascular outcomes after treatment withclopidogrel and prasugrel in the TRITON-TIMI 38 trial: a phar-macogenetic analysis. Lancet 2010;376:1312–9.

45. Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinantsof response to clopidogrel and cardiovascular events. N Engl J Med2009;360:363–75.

46. Collet J-P, Hulot J-S, Pena A, et al. Cytochrome P450 2C19polymorphism in young patients treated with clopidogrel after myo-cardial infarction: a cohort study. Lancet 2009;373:309–17.

47. Sibbing D, Stegherr J, Latz W, et al. Cytochrome P450 2C19loss-of-function polymorphism and stent thrombosis following per-cutaneous coronary intervention. Eur Heart J 2009;30:916–22.

48. Hulot J-S, Collet J-P, Silvain J, et al. Cardiovascular risk inclopidogrel-treated patients according to cytochrome P450 2C19*2loss-of-function allele or proton pump inhibitor coadministration: asystematic meta-analysis. J Am Coll Cardiol 2010;56:134–43.

49. Mega JL, Simon T, Collet J-P, et al. Reduced-function CYP2C19genotype and risk of adverse clinical outcomes among patients treatedwith clopidogrel predominantly for PCI: a meta-analysis. JAMA2010;304:1821–30.

50. Sibbing D, Koch W, Gebhard D, et al. Cytochrome 2C19*17 allelicvariant, platelet aggregation, bleeding events, and stent thrombosis inclopidogrel-treated patients with coronary stent placement. Circula-tion 2010;121:512–8.

51. Paré G, Mehta SR, Yusuf S, et al. Effects of CYP2C19 genotype onoutcomes of clopidogrel treatment. N Engl J Med 2010;363:1704–14.

52. Wallentin L, James S, Storey RF, et al. Effect of CYP2C19 andABCB1 single nucleotide polymorphisms on outcomes of treatmentwith ticagrelor versus clopidogrel for acute coronary syndromes: agenetic substudy of the PLATO trial. Lancet 2010;376:1320–8.

53. Holmes DR Jr., Dehmer GJ, Kaul S, Leifer D, O’Gara PT, SteinCM. ACCF/AHA clopidogrel clinical alert: approaches to the FDA“boxed warning”: a report of the American College of CardiologyFoundation Task Force on Clinical Expert Consensus Documentsand the American Heart Association. J Am Coll Cardiol 2010;56:

321–41.

54. Scott SA, Sangkuhl K, Gardner EE, et al. Clinical pharmacogeneticsimplementation consortium guidelines for cytochrome P450-2C19(CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther2011;90:328–32.

55. Montalescot G, Wiviott SD, Braunwald E, et al. Prasugrel comparedwith clopidogrel in patients undergoing percutaneous coronary inter-vention for ST-elevation myocardial infarction (TRITON-TIMI38): double-blind, randomised controlled trial. Lancet 2009;373:723–31.

56. Wiviott SD, Braunwald E, Angiolillo DJ, et al. Greater clinicalbenefit of more intensive oral antiplatelet therapy with prasugrel inpatients with diabetes mellitus in the trial to assess improvementin therapeutic outcomes by optimizing platelet inhibition withprasugrel—Thrombolysis in Myocardial Infarction 38. Circulation2008;118:1626 –36.

57. The CURRENT-OASIS 7 Investigators. Dose comparisons ofclopidogrel and aspirin in acute coronary syndromes. N Engl J Med2010;363:930–42.

58. Price MJ, Berger PB, Teirstein PS, et al. Standard- vs high-doseclopidogrel based on platelet function testing after percutaneouscoronary intervention. JAMA 2011;305:1097–105.

59. Parodi G, Marcucci R, Valenti R, et al. High residual plateletreactivity after clopidogrel loading and long-term cardiovascularevents among patients with acute coronary syndromes undergoingPCI. JAMA 2011;306:1215–23.

60. Becker DM, Segal J, Vaidya D, et al. Sex differences in plateletreactivity and response to low-dose aspirin therapy. JAMA 2006;295:1420–7.

61. Gurbel PA, Bliden KP, DiChiara J, et al. Evaluation of dose-relatedeffects of aspirin on platelet function: results from the Aspirin-Induced Platelet Effect (ASPECT) study. Circulation 2007;115:3156–64.

62. Zufferey A, Reny JL, Combescure C, de Moerloose P, Sanchez JC,Fontana P. Platelet reactivity is a stable and global phenomenon inaspirin-treated cardiovascular patients. Thromb Haemost 2011;106:466–74.

63. Faraday N, Yanek LR, Mathias R, et al. Heritability of plateletresponsiveness to aspirin in activation pathways directly and indirectlyrelated to cyclooxygenase-1. Circulation 2007;115:2490–6.

64. Shen H, Herzog W, Drolet M, et al. Aspirin resistance in healthydrug-naive men versus women (from the Heredity and PhenotypeIntervention Heart Study). Am J Cardiol 2009;104:606–12.

65. Frelinger AL III, Li Y, Linden MD, et al. Association ofcyclooxygenase-1-dependent and -independent platelet function as-says with adverse clinical outcomes in aspirin-treated patients pre-senting for cardiac catheterization. Circulation 2009;120:2586–96.

66. Snoep JD, Hovens MM, Eikenboom JC, van der Bom JG, HuismanMV. Association of laboratory-defined aspirin resistance with ahigher risk of recurrent cardiovascular events: a systematic review andmeta-analysis. Arch Intern Med 2007;167:1593–9.

67. Lepantalo A, Mikkelsson J, Resendiz JC, et al. Polymorphisms ofCOX-1 and GPVI associate with the antiplatelet effect of aspirin incoronary artery disease patients. Thromb Haemost 2006;95:253–9.

68. Smith SM, Judge HM, Peters G, et al. PAR-1 genotype influencesplatelet aggregation and procoagulant responses in patients withcoronary artery disease prior to and during clopidogrel therapy.Platelets 2005;16:340–5.

69. Herrera JE, Qayyum R, Faraday N, et al. Platelet response to aspirinis under polygenic control of variants in the VAV3 and phospholipaseC gamma 2 (PLCG2) genes (abstr). Circulation 2008;118 Suppl:326.

70. Herrera-Galeano JE, Becker DM, Wilson AF, et al. A novel variantin the platelet endothelial aggregation receptor-1 gene is associatedwith increased platelet aggregability. Arterioscler Thromb Vasc Biol2008;28:1484–90.

71. Undas A, Brummel K, Musial J, Mann KG, Szczeklik A. PlA2polymorphism of {beta}3 integrins is associated with enhancedthrombin generation and impaired antithrombotic action of aspirin atthe site of microvascular injury. Circulation 2001;104:2666–72.

72. Cooke GE, Bray PF, Hamlington JD, Pham DM, Goldschmidt-Clermont PJ. PlA2 polymorphism and efficacy of aspirin. Lancet1998;351:1253.

73. Cooke GE, Liu-Stratton Y, Ferketich AK, et al. Effect of plateletantigen polymorphism on platelet inhibition by aspirin, clopidogrel,

or their combination. J Am Coll Cardiol 2006;47:541–6.
Page 11: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

19JACC Vol. 60, No. 1, 2012 Voora and GinsburgJuly 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics

74. Mathias R, Kim Y, Sung H, et al. A combined genome-wide linkageand association approach to find susceptibility loci for platelet functionphenotypes in European American and African American families withcoronary artery disease. BMC Med Genomics 2010;3:22.

75. Faraday N, Yanek LR, Yang XP, et al. Identification of a specificintronic PEAR1 gene variant associated with greater platelet aggre-gability and protein expression. Blood 2011;118:3367–75.

76. Le Hello C, Morello R, Lequerrec A, Duarte C, Riddell J, HamonM. Effect of PlA1/A2 glycoprotein IIIa gene polymorphism on thelong-term outcome after successful coronary stenting. Thromb J2007;5:19–25.

77. Voora D, Horton J, Shah SH, Shaw LK, Newby LK. Polymorphismsassociated with in vitro aspirin resistance are not associated withclinical outcomes in patients with coronary artery disease who reportregular aspirin use. Am Heart J 2011;162:166–72.e1.

78. Chasman DI, Shiffman D, Zee RY, et al. Polymorphism in theapolipoprotein(a) gene, plasma lipoprotein(a), cardiovascular disease,and low-dose aspirin therapy. Atherosclerosis 2009;203:371–6.

79. Rettie AE, Haining RL, Bajpai M, Levy RH. A common geneticbasis for idiosyncratic toxicity of warfarin and phenytoin. EpilepsyRes 1999;35:253–5.

80. Gage BF, Eby C, Milligan PE, Banet GA, Duncan JR, McLeod HL.Use of pharmacogenetics and clinical factors to predict the mainte-nance dose of warfarin. Thromb Haemost 2004;91:87–94.

81. Rost S, Fregin A, Ivaskevicius V, et al. Mutations in VKORC1 causewarfarin resistance and multiple coagulation factor deficiency type 2.Nature 2004;427:537–41.

82. Li T, Chang CY, Jin DY, Lin PJ, Khvorova A, Stafford DW.Identification of the gene for vitamin K epoxide reductase. Nature2004;427:541–4.

83. Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1haplotypes on transcriptional regulation and warfarin dose. N EnglJ Med 2005;352:2285–93.

84. Wang D, Chen H, Momary KM, Cavallari LH, Johnson JA, SadeeW. Regulatory polymorphism in vitamin K epoxide reductase com-plex subunit 1 (VKORC1) affects gene expression and warfarin doserequirement. Blood 2008;112:1013–21.

85. The International Warfarin Pharmacogenetics Consortium. Estima-tion of the warfarin dose with clinical and pharmacogenetic data.N Engl J Med 2009;360:753–64.

86. Singh O, Sandanaraj E, Subramanian K, Lee LH, Chowbay B.Influence of CYP4F2 rs2108622 (V433M) on warfarin dose require-ment in Asian patients. Drug Metab Pharmacokinet 2011;26:130–6.

87. Caldwell MD, Awad T, Johnson JA, et al. CYP4F2 genetic variantalters required warfarin dose. Blood 2008;111:4106–12.

88. Takeuchi F, McGinnis R, Bourgeois S, et al. A genome-wideassociation study confirms VKORC1, CYP2C9, and CYP4F2 asprincipal genetic determinants of warfarin dose. PLoS Genet 2009;5:e1000433.

89. Cooper GM, Johnson JA, Langaee TY, et al. A genome-wide scanfor common genetic variants with a large influence on warfarinmaintenance dose. Blood 2008;122:1022–7.

90. McDonald MG, Rieder MJ, Nakano M, Hsia CK, Rettie AE.CYP4F2 is a vitamin K1 oxidase: an explanation for altered warfarindose in carriers of the V433M variant. Mol Pharmacol 2009;75:1337–46.

91. Higashi MK, Veenstra DL, Kondo LM, et al. Association betweenCYP2C9 genetic variants and anticoagulation-related outcomes dur-ing warfarin therapy. JAMA 2002;287:1690–8.

92. Limdi NA, McGwin G, Goldstein JA, et al. Influence of CYP2C9and VKORC1 1173C/T genotype on the risk of hemorrhagiccomplications in African-American and European-American pa-tients on warfarin. Clin Pharmacol Ther 2007;83:312–21.

93. Schwarz UI, Ritchie MD, Bradford Y, et al. Genetic determinants ofresponse to warfarin during initial anticoagulation. N Engl J Med2008;358:999–1008.

94. Zhang JE, Jorgensen AL, Alfirevic A, et al. Effects of CYP4F2genetic polymorphisms and haplotypes on clinical outcomes inpatients initiated on warfarin therapy. Pharmacogenet Genomics2009;19:781–9.

95. Caraco Y, Blotnick S, Muszkat M. CYP2C9 genotype-guidedwarfarin prescribing enhances the efficacy and safety of anticoagula-tion: a prospective randomized controlled study. Clin Pharmacol

Ther 2007;83:460–70.

96. Epstein RS, Moyer TP, Aubert RE, et al. Warfarin genotypingreduces hospitalization rates: results from the MM-WES (Medco-Mayo Warfarin Effectiveness Study). J Am Coll Cardiol 2010;55:2804–12.

97. McMillin GA, Melis R, Wilson A, et al. Gene-based warfarin dosingcompared with standard of care practices in an orthopedic surgerypopulation: a prospective, parallel cohort study. Ther Drug Monit2010;32:338–45.

98. Voora D, Eby C, Linder MW, et al. Prospective dosing of warfarinbased on cytochrome P-450 2C9 genotype. Thromb Haemost2005;93:700–5.

99. Anderson JL, Horne BD, Stevens SM, et al. Randomized trial ofgenotype-guided versus standard warfarin dosing in patients initiat-ing oral anticoagulation. Circulation 2007;116:2563–70.

100. Cusi D, Barlassina C, Azzani T, et al. Polymorphisms of alpha-adducin and salt sensitivity in patients with essential hypertension.Lancet 1997;349:1353–7.

101. Psaty BM, Smith NL, Heckbert SR, et al. Diuretic therapy, the{alpha}-adducin gene variant, and the risk of myocardial infarction orstroke in persons with treated hypertension. JAMA 2002;287:1680–9.

102. Davis BR, Arnett DK, Boerwinkle E, et al. Antihypertensive therapy,the alpha-adducin polymorphism, and cardiovascular disease inhigh-risk hypertensive persons: the Genetics of Hypertension-Associated Treatment Study. Pharmacogenomics J 2007;7:112–22.

103. Gerhard T, Gong Y, Beitelshees AL, et al. Alpha-adducin polymor-phism associated with increased risk of adverse cardiovascular out-comes: results from GENEtic substudy of the International Vera-pamil SR-Trandolapril Study (INVEST-GENES). Am Heart J2008;156:397–404.

104. Schelleman H, Klungel OH, Witteman JC, et al. Diuretic-geneinteraction and the risk of myocardial infarction and stroke. Phar-macogenomics J 2007;7:346–52.

105. Rau T, Heide R, Bergmann K, et al. Effect of the CYP2D6 genotypeon metoprolol metabolism persists during long-term treatment.Pharmacogenetics 2002;12:465–72.

106. Rau T, Wuttke H, Michels LM, et al. Impact of the CYP2D6genotype on the clinical effects of metoprolol: a prospective longitu-dinal study. Clin Pharmacol Ther 2009;85:269–72.

107. Nozawa T, Taguchi M, Tahara K, et al. Influence of CYP2D6genotype on metoprolol plasma concentration and beta-adrenergicinhibition during long-term treatment: a comparison with bisoprolol.J Cardiovasc Pharmacol 2005;46:713–20.

108. Oldham HG, Clarke SE. In vitro identification of the humancytochrome P450 enzymes involved in the metabolism of R(�)- andS(-)-carvedilol. Drug Metab Dispos 1997;25:970–7.

109. Venter CP, Joubert PH. Ethnic differences in response to beta1-adrenoceptor blockade by propranolol. J Cardiovasc Pharmacol1984;6:361–4.

110. Rathz DA, Brown KM, Kramer LA, Liggett SB. Amino acid 49polymorphisms of the human beta1-adrenergic receptor affect agonist-promoted trafficking. J Cardiovasc Pharmacol 2002;39:155–60.

111. Mason DA, Moore JD, Green SA, Liggett SB. A gain-of-functionpolymorphism in a G-protein coupling domain of the humanbeta1-adrenergic receptor. J Biol Chem 1999;274:12670–4.

112. Liu J, Liu ZQ, Tan ZR, et al. Gly389Arg polymorphism ofbeta1-adrenergic receptor is associated with the cardiovascular re-sponse to metoprolol. Clin Pharmacol Ther 2003;74:372–9.

113. Johnson JA, Zineh I, Puckett BJ, McGorray SP, Yarandi HN, PaulyDF. Beta 1-adrenergic receptor polymorphisms and antihypertensiveresponse to metoprolol. Clin Pharmacol Ther 2003;74:44–52.

114. Shin J, Johnson JA. Pharmacogenetics of beta-blockers. Pharmaco-therapy 2007;27:874–87.

115. Mialet Perez J, Rathz DA, Petrashevskaya NN, et al. Beta1-adrenergic receptor polymorphisms confer differential function andpredisposition to heart failure. Nat Med 2003;9:1300–5.

116. Liggett SB, Mialet-Perez J, Thaneemit-Chen S, et al. A polymor-phism within a conserved beta(1)-adrenergic receptor motif alterscardiac function and beta-blocker response in human heart failure.Proc Natl Acad Sci U S A 2006;103:11288–93.

117. Small KM, McGraw DW, Liggett SB. Pharmacology and physiologyof human adrenergic receptor polymorphisms. Annu Rev Pharmacol

Toxicol 2003;43:381–411.
Page 12: Clinical Application of Cardiovascular Pharmacogenetics · Clinical Application of Cardiovascular Pharmacogenetics Deepak Voora, MD, Geoffrey S. Ginsburg, MD, PHD Durham, North Carolina

20 Voora and Ginsburg JACC Vol. 60, No. 1, 2012Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20

118. Liggett SB, Cresci S, Kelly RJ, et al. A GRK5 polymorphism thatinhibits beta-adrenergic receptor signaling is protective in heartfailure. Nat Med 2008;14:510–7.

119. Kurnik D, Cunningham AJ, Sofowora GG, et al. GRK5 Gln41Leupolymorphism is not associated with sensitivity to beta(1)-adrenergicblockade in humans. Pharmacogenomics 2009;10:1581–7.

120. White HL, de Boer RA, Maqbool A, et al. An evaluation of thebeta-1 adrenergic receptor Arg389Gly polymorphism in individualswith heart failure: a MERIT-HF sub-study. Eur J Heart Fail2003;5:463–8.

121. Cresci S, Kelly RJ, Cappola TP, et al. Clinical and genetic modifiersof long-term survival in heart failure. J Am Coll Cardiol 2009;54:432–44.

122. Sehnert AJ, Daniels SE, Elashoff M, et al. Lack of associationbetween adrenergic receptor genotypes and survival in heart failurepatients treated with carvedilol or metoprolol. J Am Coll Cardiol2008;52:644–51.

123. de Groote P, Lamblin N, Helbecque N, et al. The impact ofbeta-adrenoreceptor gene polymorphisms on survival in patients withcongestive heart failure. Eur J Heart Fail 2005;7:966–73.

124. Shin J, Lobmeyer MT, Gong Y, et al. Relation of beta(2)-adrenoceptor haplotype to risk of death and heart transplantation inpatients with heart failure. Am J Cardiol 2007;99:250–5.

125. Lanfear DE, Jones PG, Marsh S, Cresci S, McLeod HL, Spertus JA.Beta2-adrenergic receptor genotype and survival among patientsreceiving beta-blocker therapy after an acute coronary syndrome.JAMA 2005;294:1526–33.

126. Pacanowski MA, Gong Y, Cooper-Dehoff RM, et al. Beta-adrenergic receptor gene polymorphisms and beta-blocker treatmentoutcomes in hypertension. Clin Pharmacol Ther 2008;84:715–21.

127. Wuttke H, Rau T, Heide R, et al. Increased frequency of cytochromeP450 2D6 poor metabolizers among patients with metoprolol-associated adverse effects. Clin Pharmacol Ther 2002;72:429–37.

128. Bijl MJ, Visser LE, van Schaik RH, et al. Genetic variation in theCYP2D6 gene is associated with a lower heart rate and bloodpressure in beta-blocker users. Clin Pharmacol Ther 2009;85:45–50.

129. Fux R, Morike K, Prohmer AM, et al. Impact of CYP2D6 genotypeon adverse effects during treatment with metoprolol: a prospectiveclinical study. Clin Pharmacol Ther 2005;78:378–87.

130. Zineh I, Beitelshees AL, Gaedigk A, et al. Pharmacokinetics andCYP2D6 genotypes do not predict metoprolol adverse events orefficacy in hypertension. Clin Pharmacol Ther 2004;76:536–44.

131. Zanger UM, Turpeinen M, Klein K, Schwab M. Functional phar-macogenetics/genomics of human cytochromes P450 involved indrug biotransformation. Anal Bioanal Chem 2008;392:1093–108.

132. Xu P, Jiang ZP, Zhang BK, Tu JY, Li HD. Impact of MDR1haplotypes derived from C1236T, G2677T/A and C3435T on thepharmacokinetics of single-dose oral digoxin in healthy Chinesevolunteers. Pharmacology 2008;82:221–7.

133. Verstuyft C, Schwab M, Schaeffeler E, et al. Digoxin pharmacoki-netics and MDR1 genetic polymorphisms. Eur J Clin Pharmacol2003;58:809–12.

134. Zhao LM, He XJ, Qiu F, Sun YX, Li-Ling J. Influence of ABCB1gene polymorphisms on the pharmacokinetics of verapamil among

healthy Chinese Han ethnic subjects. Br J Clin Pharmacol 2009;68:395–401.

135. Walker K, Ginsberg G, Hattis D, Johns DO, Guyton KZ, SonawaneB. Genetic polymorphism in N-acetyltransferase (NAT): populationdistribution of NAT1 and NAT2 activity. J Toxicol Environ HealthB Crit Rev 2009;12:440–72.

136. Sabbagh A, Darlu P. SNP selection at the NAT2 locus for anaccurate prediction of the acetylation phenotype. Genet Med 2006;8:76–85.

137. Okumura K, Kita T, Chikazawa S, Komada F, Iwakawa S, Tani-gawara Y. Genotyping of N-acetylation polymorphism and correla-tion with procainamide metabolism. Clin Pharmacol Ther 1997;61:509–17.

138. Woosley RL, Drayer DE, Reidenberg MM, Nies AS, Carr K, OatesJA. Effect of acetylator phenotype on the rate at which procainamideinduces antinuclear antibodies and the lupus syndrome. N EnglJ Med 1978;298:1157–9.

139. Mongey AB, Sim E, Risch A, Hess E. Acetylation status is associatedwith serological changes but not clinically significant disease inpatients receiving procainamide. J Rheumatol 1999;26:1721–6.

140. Siddoway L, Thompson K, McAllister C, et al. Polymorphism ofpropafenone metabolism and disposition in man: clinical and phar-macokinetic consequences. Circulation 1987;75:785–91.

141. Cai WM, Xu J, Chen B, Zhang FM, Huang YZ, Zhang YD. Effectof CYP2D6*10 genotype on propafenone pharmacodynamics inChinese patients with ventricular arrhythmia. Acta Pharmacol Sin2002;23:1040–4.

142. Morike K, Kivisto KT, Schaeffeler E, et al. Propafenone for theprevention of atrial tachyarrhythmias after cardiac surgery: a random-ized, double-blind placebo-controlled trial. Clin Pharmacol Ther2008;84:104–10.

143. Lee JT, Kroemer HK, Silberstein DJ, et al. The role of geneticallydetermined polymorphic drug metabolism in the beta-blockadeproduced by propafenone. N Engl J Med 1990;322:1764–8.

144. Jazwinska-Tarnawska E, Orzechowska-Juzwenko K, Niewinski P, etal. The influence of CYP2D6 polymorphism on the antiarrhythmicefficacy of propafenone in patients with paroxysmal atrial fibrillationduring 3 months propafenone prophylactic treatment. Int J ClinPharmacol Ther 2001;39:288–92.

145. Thompson JF, Man M, Johnson KJ, et al. An association study of 43SNPs in 16 candidate genes with atorvastatin response. Pharmacog-enomics J 2005;5:352–8.

146. Mega JL, Morrow DA, Brown A, Cannon CP, Sabatine MS.Identification of genetic variants associated with response to statintherapy. Arterioscler Thromb Vasc Biol 2009;29:1310–5.

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

148. Scott SA, Sangkuhl K, Gardner EE, et al. Clinical PharmacogeneticsImplementation Consortium guidelines for cytochrome P450-2C19(CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther2011;90:328–32.

Key Words: clopidogrel y pharmacogenetics y polymorphism y statinsy warfarin.

APPENDIX

For additional loci, see the online version of this article.