SlideShare a Scribd company logo
1 of 12
Download to read offline
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 
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. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
10 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 
Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 
to 20% reduced LDLc lowering 
with pravastatin (4) or simvasta-tin 
(5) due to an alternatively 
spliced HMGCR transcript that 
produces a version of HMGCR 
that is less sensitive to simvasta-tin 
inhibition (6). This haplotype 
was not identified in a GWAS 
with atorvastatin (7), and there-fore, 
whether this association 
holds for other statins is unclear. 
APOE. Two variants, rs7412 
and rs429358, define 3 haplotypes, 
namely, 2, 3, and 4, that are 
associated with lipid, cognitive, 
and thrombotic traits. In general, 
the magnitude of LDLc lowering 
is greatest for carriers of 2, fol-lowed 
by 3, and 4 haplotypes, 
respectively. The influence of 
APOE haplotypes appears to be 
consistent across variety of statin 
types (4,7,8) and doses (8), and results in mild (15%) 
attenuations of LDLc lowering. 
Additional loci. Information on ABCB1 and ABCG2 can 
be found in the Online Appendix. 
Reduction in cardiovascular events. Statins prevent car-diovascular 
disease by lowering LDLc and potential pleio-tropic 
effects. There are few genetic predictors of these 
pleiotropic benefits, but the rs20455 polymorphism in 
kinesin-like protein 6 (KIF6, an underlying disease gene) 
may be a candidate. Carriers of the risk allele may receive a 
greater benefit from statin therapy compared to noncarriers 
despite equal LDLc, triglycerides, or C-reactive protein 
reduction (9,10). However, subsequent genome-wide meta-analysis 
of large randomized placebo-controlled statin trials 
found no evidence for association with coronary artery 
disease (11) or any differential treatment benefit with statin 
therapy (12,13). 
Statin-induced musculoskeletal side effects. Statins have 
a well-defined safety profile, but come with a small risk of 
musculoskeletal side effects (14). Genetic variants in the 
hepatic transporter, SLCO1B1, influence the risk of adverse 
events (Table 2); it does not appear that cytochrome P450 
(CYP) SNPs influence statin-induced side effects. 
SLCO1B1. The solute carrier organic anion transporter 
family, member 1B1 gene (SLCO1B1, also referred to as 
SLC21A6, OATP-C, or OATP1B1) harbors many genetic 
variants, and each variant is numerically and sequentially 
labeled 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 the 
localization of this transporter to the hepatocyte plasma 
membrane (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- to 
3-fold increased risk of CK- negative myopathy (19,20). 
In trials of randomly assigned statins as well as in 
observational studies, the risk for myopathy with *5 depends 
on 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 the 
clearance of these statins (16–18,24) and thus appear to be 
statin-specific. 
Adherence to statin therapy. Often, statin therapy is 
hampered 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) observed 
that carriers of the SLCO1B1*5 allele have a higher rate of 
statin nonadherence (20,25). 
Clinical implications for statin pharmacogenetics. It is 
unlikely that genetic testing for statin efficacy will enter 
clinical care since the magnitude of associations is small 
(10% to 15% differences in LDLc lowering), and 
physicians can reasonably forecast the magnitude of 
LDLc lowering based on statin type, dose, and baseline 
LDLc. 
In contrast, statin-induced side effects and nonadherence 
are less predictable. While the current level of evidence 
surrounding SLCO1B1*5 may not support prospective 
genotyping at this time, the test is currently offered on 
consumer-directed whole genome genotyping platforms 
(e.g., 23andMe, deCODEME, and so on). A potential strat-egy 
for prospective SLCO1B1*5 testing might offer carriers 
either pravastatin or rosuvastatin or fluvastatin as first-line 
Abbreviations 
and Acronyms 
ACS  acute coronary 
syndrome(s) 
BP  blood pressure 
CYP  cytochrome P450 
GWAS  genome-wide 
association study 
HR  heart rate 
INR  international 
normalized ratio 
LDLc  low-density 
lipoprotein cholesterol 
LVEF  left ventricular 
ejection fraction 
MI  myocardial infarction 
PCI  percutaneous 
coronary intervention 
RF  reduced function 
SNP  single nucleotide 
polymorphism 
SoTuarbclees1of PShoaurmrcaecsoogfenPehtaicrmVaacroiagteionnetic Variation 
Category Description Types of Genes Examples 
Pharmacokinetic Variability in concentration of drug at site of 
drug effect 
Drug-metabolizing enzymes 
Drug transporters 
Warfarin: CYP2C9 
Clopidogrel: CYP2C19 
Simvastatin: SLCO1B1 
Metoprolol: CYP2D6 
Pharmacodynamic Variability in drug ability to influence its target Transmembrane receptors 
Intracellular enzymes 
Clopidogrel: P2RY12 
Simvastatin: HMGCR 
Metoprolol: ADBR1 
Underlying disease mechanisms Variability in disease being treated Often downstream or independent of drug target Hydrochlorothiazide: ADD1 
Simvastatin: APOE 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 11 
July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics 
Figure 1 Categories of Major Cardiovascular Pharmacogenetic Variants 
Genetic variants that influence the response to cardiovascular agents fall into 3 broad categories: 1) pharmacokinetic; 2) pharmacodynamic; and 
3) underlying disease mechanism. 
agents, which seem to depend the least on SLCO1B1 for their 
clearance. 
Thienopyridines 
Thienopyridines are effective in patients with acute coronary 
syndrome (ACS) and percutaneous coronary intervention 
(PCI); however, some patients remain at risk for death, 
myocardial infarction (MI), and stent thrombosis. Platelet 
function 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-netic); 
other loci, CYP2C9 (pharmacokinetic), PON1 
(pharmacokinetic), and P2RY12 (pharmacodynamic) may 
also influence response (Table 3). 
Platelet function in response to clopidogrel. CYP2C19. 
Clopidogrel is an inactive prodrug activated by several 
enzymes, including CYP2C19, to an active metabolite that 
inhibits the platelet adenosine diphosphate receptor, 
P2RY12 (29). CYP2C19 *2 (rs4244285) is the most com-mon 
reduced-function (RF) variant; additional, rare variants 
mimic the *2 allele: *3 [rs4986893], *4 [rs28399504], *5 
[rs56337013]) (30). An individual person’s genotype can 
be characterized in 3 ways: 1) presence of at least 1 RF 
allele (carrier vs. noncarrier); 2) the number of RF alleles 
(i.e., 0, 1, 2); or 3) the predicted, total CYP2C19 enzymatic 
activity (Table 4). Carriers of *2—also referred to as 
intermediate metabolizers or poor metabolizers—produce 
lower active metabolite and have attenuated platelet inhibi-tion 
(31). In general, there is a gene-dose effect where 
increasing number of RF alleles (i.e., 0, 1, 2, or extensive 
metabolizers, intermediate metabolizers, and poor metabo-lizers) 
predicts a decreasing amount of platelet inhibition 
(27,31,32). Carriers of another variant *17 (rs3758581, 
ultrametabolizers) exhibit increased CYP2C19 activity, pro-duce 
more active metabolite, and improved platelet inhibi-tion, 
in most reports (33,34). 
Higher loading and maintenance doses (e.g., 1,200 mg 
and 150 mg/day) appear, in part, to overcome the effects 
of the *2 allele (30,32,35,36), although not completely 
(37), and can require up to 300 mg/day (38). Ticlopidine 
(39), prasugrel (31,40,41), and ticagrelor (42) all produce 
uniform platelet inhibition in *2 carriers and noncarriers. 
ABCB1. The adenosine triphosphate-binding cassette, sub-family 
B (MDR/TAP), member 1 (ABCB1) gene encodes 
an apical membrane protein in enterocytes and hepatocytes 
and serves to reduce bioavailability. Clopidogrel appears to be 
handled by this transporter, and 3 SNPs—C1236T (rs1128503), 
G2677T (rs2032582), and C3435T (rs1045642)—capture the 
common genetic variation at this locus. Persons who carry a 
T allele at each SNPs (i.e., the T-T-T haplotype) produced 
reduced active metabolite (43) in an initial report. Despite 
this initial observation, the link to reduced platelet inhibi-tion 
has been difficult to establish (27,30), although may be 
present for persons who carry 2 copies of the T-T-T 
haplotype (44). As with CYP2C19 variants, prasugrel- 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
12 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 
Clinical 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 at 
ABCB1, P2RY12, CYP2C9, and PON1 can be found in the 
Online Appendix. 
Clinical response to clopidogrel. CYP2C19. In parallel 
with the platelet function data, the CYP2C19*2 allele is 
associated 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 (poor 
metabolizers) experience a 1.8-fold increase. This pattern 
also 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 genetic 
associations with platelet function are mirrored in the 
clinical response to clopidogrel in the setting of PCI. These 
observations formed the foundation for updating the clopi-dogrel 
label by the Food and Drug Administration to 
include pharmacogenetic information. Similarly, the gain of 
function variant, CYP2C19*17, is associated with increased 
risk of bleeding (50), and protection from ischemic events 
(51) CYP2C19*2 carriers treated with prasugrel or ticagrelor 
do not show a heightened risk of cardiovascular death, MI, 
stroke, or stent thrombosis (40,52). 
ABCB1. Carriers of 2 copies of the ABCB1 T-T-T haplo-type 
treated with clopidogrel are at an increased risk for 
subsequent death, MI, or stroke compared to persons who 
carried none, thus mirroring the platelet function data 
(44,45,52). Genetic variation at the ABCB1 locus does not 
appear to influence prasugrel- or ticagrelor-treated patients 
(40,52). 
Clinical implications. The current state of evidence reflects 
a strong and consistent association with the CYP2C19*2 allele 
and an increased risk for cardiovascular outcomes in ACS/ 
PCI patients treated with clopidogrel, although not other 
settings (51). Alternative antiplatelet agents (i.e., prasugrel 
and ticagrelor) mitigate the adverse risk of CYP2C19*2- 
associated adverse events. Although CYP2C19 genotyping is 
commercially available, whether genotype-guided therapy 
will improve outcomes or reduce costs is unknown. Con-sensus 
statements (53) currently do not recommend routine 
testing. However, there is sufficient evidence to support 
physicians who may choose to pursue CYP2C19*2 testing in 
selected patients: 1) for diagnosis in patients with compli-cations 
of clopidogrel therapy such as stent thrombosis in 
compliant clopidogrel users; or 2) for the choice of dual 
antiplatelet therapy in the ACS/PCI setting where the 
physician believes that additional information regarding 
the risk/benefit profile for clopidogrel will influence the 
choice of drug therapy (54). Outside of these scenarios, 
the ACS/PCI setting, or situations where there are ample 
data to guide drug choice (e.g., ST-segment elevation MI 
[55], diabetic patients [56], age 75 years, or prior 
MTaainblGee2neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRSetsaptoinnsse to Statins 
Caucasians Africans Asians Type of Study Effect of Variant Statin Type Ref. # 
Gene Variant(s) 
Statin Response/ 
Risk Allele/Haplotype Frequencies 
Class effect 7,8,145–147 
by alleles at rs7412 and 
rs429358 
LDLc lowering/GWAS, CG 2LDL reduction with 3 vs. 2 
APOE 2, 3, and 4 haplotypes defined 
2LDL reduction with 4 vs. 2 
2: 15%–20%* 
3: 90%–95% 
4: 20%–30% 
3% 6% LDLc lowering/CG 2LDL reduction in H7 carriers Simvastatin 4,5 
HMGCR H7 haplotype defined by alleles at 
rs17244841, rs17238540, 
and rs3846662 
Simvastatin, atorvastatin 19,20 
1In CK positive symptoms 
1In CK negative symptoms with 
SLCO1B1 *5: rs4149056 15% 1% 13% Musculoskeletal side 
effects/GWAS, CG 
risk allele 
Simvastatin, atorvastatin 20,25 
SLCO1B1 *5: rs4149056 15% 1% 13% Nonadherence/CG 1Risk of nonadherence with 
risk allele 
*Represents Caucasian population. 
CG  candidate gene association study; CK  creatine kinase; GWAS  genome-wide association study; LDLc  low-density lipoprotein cholesterol. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 13 
July 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 a 
person is found to be a poor metabolizer (Table 4), then an 
alternative to clopidogrel, such as prasugrel or ticagrelor, 
should be considered. This seems preferable over increased 
clopidogrel doses, which have not shown benefit over 
standard dose clopidogrel (57–59). The greatest uncertainty 
is in the intermediate metabolizer group (i.e., those with 1 
loss of function allele) and is where integration of other 
clinical risk factors such as diabetes, body mass index, cost, 
and other bleeding and thrombosis risk factors need to be 
considered in determining the therapy with the optimal 
risk:benefit profile. 
Aspirin 
Aspirin irreversibly inhibits prostaglandin G/H synthase 1 
(PTGS1, or COX-1) and the conversion of arachidonic acid 
to thromboxane. With adequate dosing and compliance, 
aspirin is capable of completely inhibiting COX-1 in 99% 
of persons; thus, true “aspirin resistance” is rare (60). 
However, alternate agonists such as adenosine diphosphate 
and collagen can produce robust aggregation in the face of 
complete COX-1 inhibition (61), a response that demon-strates 
wide interindividual variability (62), heritability 
(63,64), and at high levels, association with future cardio-vascular 
events (65,66), therefore making aspirin a candidate 
for pharmacogenetic discovery. Because aspirin uniformly 
inhibits its target, COX-1, there are no pharmacokinetic or 
pharmacodynamic genetic considerations. Instead, investi-gators 
have focused on platelet function loci (underlying 
disease) and LPA (underlying disease). 
Variants in several platelet genes, namely, PEAR1, 
ITGB3, VAV3, GPVI, F2R, and GP1BA, are associated with 
platelet function in response to aspirin (67–70). One with 
moderate evidence is rs5918 in ITGB3 where carriers of the 
risk allele have heightened platelet function on aspirin 
(71–73). Recent GWAS of platelet function in response to 
aspirin have identified additional genetic loci that have yet 
to be replicated (74). Finally, the most robust association 
comes from a large study that found carriers of the minor 
allele for an intronic SNP, rs12041331, in PEAR1 have 
higher PEAR1 platelet protein content and heightened 
platelet function in response to aspirin (75). 
The link to an increased risk of clinical events in aspirin 
users has not been established despite several large studies 
(76,77), although the PEAR1 variant rs12041331 has not 
yet been tested. An uncommon variant in LPA (rs3798220) 
seems to modify the protective effects of aspirin: carriers 
had a more than 2-fold reduction in the risk for cardio-vascular 
disease with aspirin, whereas noncarriers (95% 
of Caucasians) had none in a large placebo-controlled 
clinical trial (78). 
Clinical implications. Because of the lack of consistent or 
preliminary associations with the variants described in the 
preceding text, there is currently no role for genetic testing 
for aspirin. 
Warfarin 
Three loci contribute to the heterogeneity in response: 
CYP2C9 (pharmacokinetic), VKORC1 (pharmacodynamic), 
and CYP4F2 (pharmacodynamic) (Table 5). 
Warfarin dose requirements. CYP2C9. CYP2C9 is re-sponsible 
for S-warfarin (the more active enantiomer) clear-ance. 
The 2 most common reduced-function variants are 
MTaainblGee3neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRCelsoppoidnosgeretol Clopidogrel 
Gene Variant(s) 
Risk Allele/ 
Haplotype Frequencies 
Drug Response/Type of Study 
Effect of Variant 
Caucasians Africans Asians With Risk Allele Ref. # 
CYP2C19 *2 (rs4244285) 16% 14% 27% Drug concentration, platelet function, 
recurrent MI, stent thrombosis/ 
GWAS, DMET, CG 
2Active metabolite concentration 
2Inhibition of platelet function 
1Risk of recurrent MI, stroke, 
stent thrombosis 
Graded risk with 0, 1, or 2 alleles 
31,32,45–47,52 
CYP2C19 *17 (rs3758581) 5% 10% 6% Drug concentration, platelet function, 
bleeding/CG 
1Active metabolite concentration 
1Inhibition of platelet function 
1Risk bleeding 
32,33,50 
ABCB1 T-T-T haplotype defined by 
T allele at C1236T 
(rs1128503), G2677T 
(rs2032582), and 
C3435T (rs1045642) 
71% 12% 65% Drug concentration, platelet function, 
recurrent MI, stroke, death/CG 
2Active metabolite concentration 
2No change in inhibition of 
platelet function 
1Risk of recurrent MI, stroke, 
death (in carriers of 2 T-T-T 
haplotypes) 
27,30,43,45,48,49,52 
MI  myocardial infarction; DMET  drug metabolizing enzyme and transporter panel; other abbreviations as in Table 2. 
CYTPab2lCe149 DipClYoPty2pCe1C9laDsispilfioctyapteionC*lassification* 
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 
*Adapted from Ingelman-Sundberg et al. (147) and Scott et al. (148). 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
14 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 
Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 
*2 (rs1799853) and *3 (rs1057910). The CYP2C9*2 allele 
reduces warfarin clearance by 30% and the *3 allele by 
90% (79), which translate into 19% and 33% reductions 
per allele in warfarin dose requirements compared to 
noncarriers (80). 
VKORC1. Vitamin K epoxide reductase complex, subunit 1 
(VKORC1) is warfarin’s target (81,82), and resequencing 
this gene identified 2 haplotypes (A and B defined by the 
alleles at 5 SNPS: rs7196161, rs9923231, rs9934438, 
rs8050894, and rs2359612) that influence VKORC1 gene 
expression and predict warfarin dose requirements (83). Of 
these 5 SNPs, an A allele at rs9923231 (also referred to as 
1639 GA and the 3673 SNP) in the promoter region is 
responsible for reducing: VKORC1 gene expression (84). 
Therefore, carriers of an A allele at rs9923231 (or the A 
haplotype) require a 30% per allele reduction in warfarin 
dose requirements (85). 
CYP4F2. Several GWAS have not only confirmed the 
observations above but also have identified a novel associa-tion 
between rs2108622 in CYP4F2 and reduced hepatic 
CYP4F2, higher levels of hepatic vitamin K, and higher 
warfarin dose requirements (86–90). 
Additional loci. Additional information regarding CALU 
and GGCX can be found in the Online Appendix. 
Clinical response to warfarin. RETROSPECTIVE STUDIES. 
Standard dosing algorithms (i.e., a 5 mg or 10 mg loading 
dose followed by titration based on the INR) lead to an 
increased risk of an out-of-range INR (4.0) or a delay in 
the time-to-therapeutic range INR in carriers of the 
CYP2C9*2 or CYP2C9*3 alleles (91,92), the A haplotype or 
A allele of the 1639 SNP in VKORC1 (93), or the T allele 
in CYP4F2 (94). Importantly, only variants in CYP2C9 have 
been linked to the risk of bleeding (91,92). 
PROSPECTIVE STUDIES OF TAILORED WARFARIN THERAPY. 
Small randomized pilot studies, parallel cohort studies, and 
single arm studies with or without historical controls have 
prospectively explored genetically guided warfarin therapy. 
Whereas some studies suggest a benefit over standard 
therapy (95–97), others failed to show any significant 
advantage (98) except for smaller and fewer dosing changes 
and INR measurements (99). Definitive clinical trials are 
ongoing to assess the benefit of incorporating genetics into 
warfarin therapy. 
Clinical implications. There are strong genetic associa-tions 
surrounding CYP2C9, VKORC1, and CYP4F2 variants 
that influence the response to warfarin. Commercial testing 
and algorithms that assist in the interpretation of genotypes 
are available. Therefore, there is evidence to justify and tools 
to enable genotype-guided warfarin therapy. Until large 
scale trials demonstrate a benefit for routine testing, physi-cians 
may choose to pursue testing in selected patients who 
they feel may benefit by: 1) diagnosing those with compli-cations 
from warfarin therapy (e.g., hemorrhage); 2) pre-dicting 
dose for those at high risk of bleeding (e.g., 
“triple-therapy” with aspirin, clopidogrel, and warfarin); or 
3) weighing the costs of newer anticoagulants against 
warfarin. 
Diuretics 
Blood pressure–lowering response. The adducin 1 (alpha) 
gene (ADD1, underlying disease) is implicated in animal 
and human sodium handling (100), and persons with a 
Gly460Trp substitution are more salt sensitive and have 
enhanced blood pressure (BP) lowering with thiazide di-uretics 
(100). 
Clinical outcomes. In an observational study, patients 
with Trp460 who were treated with thiazide diuretics 
experienced a greater protection from MI and stroke than 
patients with Gly460 (101). However, this difference in the 
protective effects of diuretics could not be replicated in 
several larger studies (102–104). 
Clinical implications. Given the inconsistent associations 
and lack of a commercially available test, the use of this 
genetic variant in guiding treatment options in hypertension 
is limited. 
MTaainblGee5neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRWesaprfoanrisne to Warfarin 
Gene Variant(s) 
Risk Allele Frequencies 
Drug Response/ 
Type of Study 
Effect of Variant in 
Caucasians Africans Asians Carriers of Risk Allele Ref. # 
CYP2C9 *2 (rs1799853) 
*3 (rs1057910) 
*2: 10% 
*3: 6% 
1% 
1% 
1% 
4% 
Drug concentration, warfarin 
dose requirements, 
out-of-range INR values, 
hemorrhage/GWAS, CG 
2Clearance of S-warfarin 
2Dose requirements 
1Risk of out-of-range INR values 
1Time to stable, therapeutic INR 
1Risk of hemorrhage 
79,80,88,89,91–93 
VKORC1 1639 (rs9923231)* 60% 98% 2% Warfarin dose requirements, 
out-of-range INR values/ 
linkage (human/animal), 
GWAS, CG 
2Dose requirements 
1Risk of out-of-range INR values 
1Time to stable, therapeutic INR 
81–83,88,89 
CYP4F2 rs2108622 23% 6% 25% Warfarin dose requirements/ 
GWAS, DMET 
1Dose requirements 87–89 
*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. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 15 
July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics 
Beta-Blockers 
Beta-adrenergic receptor antagonists (or beta-blockers) are 
a diverse class of agents that primarily antagonize the beta-1 
adrenergic receptor, encoded by ADBR1. Accordingly, 
changes in heart rate (HR), BP, and left ventricular ejection 
fraction (LVEF) are considered the surrogate responses in 
place 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 common 
RF variant CYP2D6*4 (rs3892097) results in the absence of 
CYP2D6 activity. Carriers have a higher systemic exposure 
to metoprolol (105), which translates into a greater reduc-tion 
in HR and BP (106,107). Beta-blockers such as 
atenolol and carvedilol (108) do not require CYP2D6 for 
their metabolism. 
ADRB1. Ethnic differences in the dose response for pro-pranolol 
(109) motivated pharmacodynamic genetic inves-tigations 
of beta-blockers. Two variants in ADBR1, the 
Ser49Gly (rs1801252) and Arg389Gly (rs1801253), lead to 
impaired down-regulation (110) and higher signal transduc-tion, 
respectively (111). Therefore, carriers of either variant 
have enhanced, beta-1-receptor activity and more beta-blocker 
sensitivity. Healthy volunteers and patients with 
hypertension who carry 2 Arg389 variants have a greater 
HR (112) or BP (113) reduction mainly with metoprolol, 
although not with all beta-blockers (114). 
In patients with systolic heart failure treated with either 
metoprolol or carvedilol (115), but not bucindolol (116), 
carriers of 2 copies of the ADBR1 Arg389 variant had 
significantly greater improvements in LVEF compared to 
the Gly389 carriers. 
ADRB2. Genetic variation in the beta-2 adrenergic receptor 
(ADBR2) centers around 2 variants: Arg16Gly (rs1042713) and 
Glu27Gln (rs1042714). Receptors with Gly16 versus Arg16 
have enhanced down-regulation of ADBR2. In contrast, 
receptors with Glu27 appear to be resistant to down-regulation 
(117). Extension to variation in BP or HR 
lowering in response to beta-blocker therapy has not been 
demonstrated (114). 
GRK5. Downstream of the beta-1-adrenergic receptor are 
G-protein coupled receptor kinases responsible for desensi-tization 
of the beta-1-adrenergic receptor. A Glu41Leu 
genetic variant in 1 of these kinases, G protein-coupled 
receptor kinase 5 (GRK5), is more prevalent in African 
Americans. The Leu41 more effectively uncoupled 
isoproterenol-stimulated responses than GRK5-Q41, thus 
producing a pharmacological-like “beta-blockade” in mice 
(118), although with no differences in atenolol-induced HR 
reduction in humans (119). 
MTaainblGee6neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRBeestpao-AnsderetnoerBgeictaR-AedcreepnteorrgAicntRaegcoenpistotsr Antagonists 
Caucasians Africans Asians Carriers of Risk Allele Beta-Blocker Type Ref. # 
Risk Allele Frequencies 
Drug Response/Type of Study 
Gene Variant(s) 
Effect of Variant in 
Metoprolol 105,106,127,128 
2Clearance 
1Reduction in BP and HR 
1Risk of bradycardia 
CYP2D6 *4 (rs3892097) 18% 23% Drug concentration, 
HR and BP reduction/CG 
113,115,116 
ADRB1 Ser49Gly (rs1801252) 20% 1% 1% HR, BP reduction/CG 2Reduction in BP and HR Metoprolol 112 
ADRB1 Arg389Gly (rs1801253) 31% 41% 20% HR, improvement in LVEF, 
Metoprolol (BP/HR/LVEF) 
Carvedilol (LVEF) 
Bucindolol (hospitalization/death) 
2Reduction in BP and HR 
2Improvement in LVEF 
BP reduction, hospitalization 
or death in CHF/CG 
BP  blood pressure; CG  candidate gene association study; HR  heart rate; LVEF  left ventricular ejection fraction. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
16 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 
Clinical 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 reduction 
in the time to first hospitalization or death when treated 
with bucindolol (116) but not carvedilol or metoprolol 
(120–125) compared to Gly389 carriers. Whether these 
differences are due to drug specific effects (i.e., bucindolol vs. 
metoprolol) or the play of chance has not been adequately 
tested. In patients with chronic coronary artery disease 
randomly assigned to verapamil or atenolol, carriers of at 
least 1 copy of the Ser49/ Arg389 haplotype had a 9- versus 
2-fold worsened prognosis when assigned to verapamil 
versus atenolol (126) despite equivalent BP and HR control 
in both groups. 
ADRB2. One study examined the influence of genetic 
variation at ADRB2 and its influence on ACS patients and 
found those homozygous for both the Arg16 and Glu27 
alleles had a 20% rate of subsequent death versus 6% of 
those homozygous for both Gly16 and Gln27 (125). This 
trend has been observed in some studies of patients with 
heart failure (124), although not all (122,123,126). 
GRK5. Extension of the associations of the Glu41Leu 
variant to clinical outcomes of patients was demonstrated in 
1 study where Leu41 carriers exhibited improvement in sur-vival 
compared to Glu41 carriers (118), although not in 
another, 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 Arg389 
variant have: 1) enhanced reduction in HR and BP; 2) larger 
improvements in LVEF; and 3) longer survival when 
treated with chronic beta-blocker therapy compared to 
persons with the Gly389 variant. Although it is unlikely that 
beta-blocker therapy will ever be withheld for carriers of the 
Gly389 variant, a potential application of these findings 
would 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 Gly389 
variant. 
Because certain beta-blockers such as atenolol and carve-dilol 
are minimally handled by CYP2D6 (131), these may be 
reasonable alternates for carriers of CYP2D6*4 with 
metoprolol-induced bradycardia. Commercial testing for 
CYP2D6*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 with 
altered 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 been 
observed (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 to 
be related to variation in its pharmacokinetics, the induction 
of drug-induced lupus-associated autoantibodies (138,139), 
but not symptomatic, drug-induced lupus (138,139) has 
been linked to the slow acetylator status. 
Propafenone. The hepatic hydroxylation by CYP2D6 of 
propafenone into 5-hydroxypropafenone demonstrates wide 
interindividual variability (140). Carriers of CYP2D6*4 (a 
reduced function allele) have reduced propafenone clearance 
compared to noncarriers (140–142). 
Antiarrhythmic efficacy. At low doses of propafenone, 
CYP2D6*4 carriers have greater reduction in exercise- or 
isoproterenol-induced HR compared to noncarriers (143), 
although not with higher doses or without the “stress” of 
exercise/isoproterenol (140,142,143). Further, CYP2D6*4 
carriers have enhanced suppression of atrial and ventricular 
arrhythmias compared to noncarriers in some studies 
(141,144), although not in all (140,142) Therefore, there are 
insufficient data to make any conclusions regarding 
CYP2D6 genotype and antiarrhythmic efficacy. 
Toxicity. Central nervous system side effects with 
propafenone, as well as excessive beta-blockade, are correlated 
with 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 do 
not sufficiently translate to consistent associations of mean-ingful 
clinical outcomes. Therefore, there is currently no 
role for pharmacogenetic testing in the clinical use of these 
medications. 
Future Directions 
The data to date support many common variants that alter 
the 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 variable 
clinical effects are also candidates for a pharmacogenetic approach. 
Novel systems approaches may elucidate additional deter-minants 
of drug response. Broad “omics” approaches have 
several advantages over traditional genetics-based research, 
for example, 1) responding to the environment, and 2) 
elucidating how the activity of entire pathways, instead of 
individual genes, influence drug response. 
The field of pharmacogenomics would benefit from the 
development of thresholds of evidence for testing and 
coverage by insurance. With the expanding number and 
complexity of “-omic” markers of drug response is an equally 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 17 
July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics 
profound evidentiary gap between association studies and 
those that demonstrate clinical utility. It may be unrealistic 
to wait (or to require) a prospective, events-driven random-ized 
controlled trial of each drug-marker combination. Even 
when such trials are conducted, the results may be made 
obsolete by new observations and/or approval of new drugs. 
Therefore, genetic substudies of clinical trials and registries 
may become the highest levels of evidence for the potential 
benefits of using pharmacogenetic testing. Such standards 
are common in many fields of medicine where there is often 
a lack of randomized clinical trial data. Observational 
studies and comparative effectiveness research will lay the 
foundation 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: 
deepak.voora@duke.edu or geoffrey.ginsburg@duke.edu. 
REFERENCES 
1. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrel 
in patients with acute coronary syndromes. N Engl J Med 2009;361: 
1045–57. 
2. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus 
clopidogrel in patients with acute coronary syndromes. N Engl J Med 
2007;357:2001–15. 
3. Simon JA, Lin F, Hulley SB, et al. Phenotypic predictors of response 
to simvastatin therapy among African-Americans and Caucasians: 
the Cholesterol and Pharmacogenetics (CAP) study. Am J Cardiol 
2006;97:843–50. 
4. Chasman DI, Posada D, Subrahmanyan L, Cook NR, Stanton VP 
Jr., 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 the 
3-hydroxyl-3-methylglutaryl coenzyme a reductase gene is associated 
with racial differences in low-density lipoprotein cholesterol response 
to simvastatin treatment. Circulation 2008;117:1537– 44. 
6. Medina MW, Gao F, Ruan W, Rotter JI, Krauss RM. Alternative 
splicing of 3-hydroxy-3-methylglutaryl coenzyme A reductase is 
associated with plasma low-density lipoprotein cholesterol response 
to 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 in 
the Treating to New Targets (TNT) cohort. Circ Cardiovasc Genet 
2009;2:173– 81. 
8. Voora D, Shah SH, Reed CR, et al. Pharmacogenetic predictors of 
statin-mediated low-density lipoprotein cholesterol reduction and 
dose response. Circ Cardiovasc Genet 2008;1:100–6. 
9. Iakoubova OA, Sabatine MS, Rowland CM, et al. Polymorphism in 
KIF6 gene and benefit from statins after acute coronary syndromes: 
results from the PROVE IT-TIMI 22 study. J Am Coll Cardiol 
2008;51:449 –55. 
10. Iakoubova OA, Tong CH, Rowland CM, et al. Association of the 
Trp719Arg polymorphism in kinesin-like protein 6 with myocardial 
infarction and coronary heart disease in 2 prospective trials: the 
CARE and WOSCOPS trials. J Am Coll Cardiol 2008;51:435– 43. 
11. Assimes TL, Ho´lm H, Kathiresan S, et al. Lack of association 
between the Trp719Arg polymorphism in kinesin-like protein-6 and 
coronary artery disease in 19 case-control studies. J Am Coll Cardiol 
2010;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 Cardiol 
2011;57:2000 –7. 
13. Ridker PM, MacFadyen JG, Glynn RJ, Chasman DI. Kinesin-Like 
Protein 6 (KIF6) polymorphism and the efficacy of rosuvastatin in 
primary prevention/clinical perspective. Circ Cardiovasc Genet 2011; 
4:312–7. 
14. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of 
cholesterol-lowering treatment: prospective meta-analysis of data 
from 90,056 participants in 14 randomised trials of statins. Lancet 
2005;366:1267–78. 
15. Kameyama Y, Yamashita K, Kobayashi K, Hosokawa M, Chiba K. 
Functional characterization of SLCO1B1 (OATP-C) variants, 
SLCO1B1*5, SLCO1B1*15 and SLCO1B1*15C1007G, by using 
transient expression systems of HeLa and HEK293 cells. Pharma-cogenet 
Genomics 2005;15:513–22. 
16. Pasanen MK, Neuvonen M, Neuvonen PJ, Niemi M. SLCO1B1 
polymorphism markedly affects the pharmacokinetics of simvastatin 
acid. Pharmacogenet Genomics 2006;16:873–9. 
17. Pasanen MK, Fredrikson H, Neuvonen PJ, Niemi M. Different 
effects of SLCO1B1 polymorphism on the pharmacokinetics of 
atorvastatin and rosuvastatin. Clin Pharmacol Ther 2007;82:726 –33. 
18. Niemi M, Pasanen MK, Neuvonen PJ. SLCO1B1 polymorphism and 
sex 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 and 
statin-induced myopathy—a genomewide study. N Engl J Med 
2008;359:789 –99. 
20. Voora D, Shah SH, Spasojevic I, et al. The SLCO1B1*5 genetic 
variant is associated with statin-induced side effects. J Am Coll 
Cardiol 2009;54:1609 –16. 
21. Puccetti L, Ciani F, Auteri A. Genetic involvement in statins 
induced 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 to 
moderate muscular symptoms with high-dosage statin therapy in 
hyperlipidemic patients—the PRIMO study. Cardiovasc Drugs Ther 
2005;19:403–14. 
23. Jacqueline Suk Danik, MacFadyen J, Nyberg F, Ridker P. Lack of 
association between polymorphisms in the slco1b1 gene and clinical 
myalgia 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 of 
OATP1B1 genotype on the pharmacokinetics of rosuvastatin in 
Koreans. 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 intolerance 
in routinely treated individuals with type 2 diabetes: a Go-DARTS 
study. Clin Pharmacol Ther 2010;89:210–6. 
26. Gurbel PA, Bliden KP, Guyer K, et al. Platelet reactivity in patients 
and 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 of 
cytochrome P450 2C19 genotype with the antiplatelet effect and 
clinical efficacy of clopidogrel therapy. JAMA 2009;302:849 –57. 
28. Breet NJ, van Werkum JW, Bouman HJ, et al. Comparison of 
platelet function tests in predicting clinical outcome in patients 
undergoing coronary stent implantation. JAMA 2010;303:754–62. 
29. Savi P, Pereillo JM, Uzabiaga MF, et al. Identification and biological 
activity of the active metabolite of clopidogrel. Thromb Haemost 
2000;84:891– 6. 
30. Gladding P, Webster M, Zeng I, et al. The pharmacogenetics and 
pharmacodynamics of clopidogrel response: an analysis from the 
PRINC (Plavix Response in Coronary Intervention) trial. J Am Coll 
Cardiol Intv 2008;1:620 –7. 
31. Brandt JT, Close SL, Iturria SJ, et al. Common polymorphisms of 
CYP2C19 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 polymorphisms 
and the impact of a higher clopidogrel dose regimen on active 
metabolite 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. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
18 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 
Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 
34. Frére C, Cuisset T, Gaborit B, Alessi MC, Hulot JS. The 
CYP2C19*17 allele is associated with better platelet response to 
clopidogrel 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 increased 
clopidogrel maintenance dose or lansoprazole co-administration on 
the antiplatelet response to clopidogrel in CYP2C19-genotyped 
healthy subjects. J Thromb Haemost 2010;8:610 –3. 
36. Gladding P, White H, Voss J, et al. Pharmacogenetic testing for 
clopidogrel using the rapid INFINITI analyzer: a dose-escalation 
study. J Am Coll Cardiol Intv 2009;2:1095–101. 
37. Jeong Y-H, Kim I-S, Park Y, et al. Carriage of cytochrome 2C19 
polymorphism is associated with risk of high post-treatment platelet 
reactivity on high maintenance-dose clopidogrel of 150 mg/day: 
results of the ACCEL-DOUBLE (Accelerated Platelet Inhibition by 
a 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 clopidogrel 
resistance? Circulation 2009;119:2854 –7. 
39. Maeda A, Ando H, Asai T, et al. Differential impacts of CYP2C19 
gene polymorphisms on the antiplatelet effects of clopidogrel and 
ticlopidine. Clin Pharmacol Ther 2011;89:229 –33. 
40. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P450 genetic 
polymorphisms and the response to prasugrel. Relationship to phar-macokinetic, 
pharmacodynamic, and clinical outcomes. Circulation 
2009;119:2553– 60. 
41. Varenhorst C, James S, Erlinge D, et al. Genetic variation of 
CYP2C19 affects both pharmacokinetic and pharmacodynamic re-sponses 
to clopidogrel but not prasugrel in aspirin-treated patients 
with coronary artery disease. Eur Heart J 2009;30:1744 –52. 
42. Tantry US, Bliden KP, Wei C, et al. First analysis of the relation 
between CYP2C19 genotype and pharmacodynamics in patients 
treated with ticagrelor versus clopidogrel/clinical perspective. Circ 
Cardiovasc Genet 2010;3:556–66. 
43. Taubert D, von Beckerath N, Grimberg G, et al. Impact of 
P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther 
2006;80:486 –501. 
44. Mega JL, Close SL, Wiviott SD, et al. Genetic variants in ABCB1 
and CYP2C19 and cardiovascular outcomes after treatment with 
clopidogrel 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 determinants 
of response to clopidogrel and cardiovascular events. N Engl J Med 
2009;360:363–75. 
46. Collet J-P, Hulot J-S, Pena A, et al. Cytochrome P450 2C19 
polymorphism 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 2C19 
loss-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 in 
clopidogrel-treated patients according to cytochrome P450 2C19*2 
loss-of-function allele or proton pump inhibitor coadministration: a 
systematic meta-analysis. J Am Coll Cardiol 2010;56:134–43. 
49. Mega JL, Simon T, Collet J-P, et al. Reduced-function CYP2C19 
genotype and risk of adverse clinical outcomes among patients treated 
with clopidogrel predominantly for PCI: a meta-analysis. JAMA 
2010;304:1821–30. 
50. Sibbing D, Koch W, Gebhard D, et al. Cytochrome 2C19*17 allelic 
variant, platelet aggregation, bleeding events, and stent thrombosis in 
clopidogrel-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 on 
outcomes of clopidogrel treatment. N Engl J Med 2010;363:1704–14. 
52. Wallentin L, James S, Storey RF, et al. Effect of CYP2C19 and 
ABCB1 single nucleotide polymorphisms on outcomes of treatment 
with ticagrelor versus clopidogrel for acute coronary syndromes: a 
genetic substudy of the PLATO trial. Lancet 2010;376:1320–8. 
53. Holmes DR Jr., Dehmer GJ, Kaul S, Leifer D, O’Gara PT, Stein 
CM. ACCF/AHA clopidogrel clinical alert: approaches to the FDA 
“boxed warning”: a report of the American College of Cardiology 
Foundation Task Force on Clinical Expert Consensus Documents 
and the American Heart Association. J Am Coll Cardiol 2010;56: 
321–41. 
54. Scott SA, Sangkuhl K, Gardner EE, et al. Clinical pharmacogenetics 
implementation consortium guidelines for cytochrome P450-2C19 
(CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther 
2011;90:328 –32. 
55. Montalescot G, Wiviott SD, Braunwald E, et al. Prasugrel compared 
with clopidogrel in patients undergoing percutaneous coronary inter-vention 
for ST-elevation myocardial infarction (TRITON-TIMI 
38): double-blind, randomised controlled trial. Lancet 2009;373: 
723–31. 
56. Wiviott SD, Braunwald E, Angiolillo DJ, et al. Greater clinical 
benefit of more intensive oral antiplatelet therapy with prasugrel in 
patients with diabetes mellitus in the trial to assess improvement 
in therapeutic outcomes by optimizing platelet inhibition with 
prasugrel—Thrombolysis in Myocardial Infarction 38. Circulation 
2008;118:1626 –36. 
57. The CURRENT-OASIS 7 Investigators. Dose comparisons of 
clopidogrel and aspirin in acute coronary syndromes. N Engl J Med 
2010;363:930–42. 
58. Price MJ, Berger PB, Teirstein PS, et al. Standard- vs high-dose 
clopidogrel based on platelet function testing after percutaneous 
coronary intervention. JAMA 2011;305:1097–105. 
59. Parodi G, Marcucci R, Valenti R, et al. High residual platelet 
reactivity after clopidogrel loading and long-term cardiovascular 
events among patients with acute coronary syndromes undergoing 
PCI. JAMA 2011;306:1215–23. 
60. Becker DM, Segal J, Vaidya D, et al. Sex differences in platelet 
reactivity and response to low-dose aspirin therapy. JAMA 2006;295: 
1420–7. 
61. Gurbel PA, Bliden KP, DiChiara J, et al. Evaluation of dose-related 
effects 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 in 
aspirin-treated cardiovascular patients. Thromb Haemost 2011;106: 
466–74. 
63. Faraday N, Yanek LR, Mathias R, et al. Heritability of platelet 
responsiveness to aspirin in activation pathways directly and indirectly 
related to cyclooxygenase-1. Circulation 2007;115:2490–6. 
64. Shen H, Herzog W, Drolet M, et al. Aspirin resistance in healthy 
drug-naive men versus women (from the Heredity and Phenotype 
Intervention Heart Study). Am J Cardiol 2009;104:606 –12. 
65. Frelinger AL III, Li Y, Linden MD, et al. Association of 
cyclooxygenase-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, Huisman 
MV. Association of laboratory-defined aspirin resistance with a 
higher risk of recurrent cardiovascular events: a systematic review and 
meta-analysis. Arch Intern Med 2007;167:1593–9. 
67. Lepantalo A, Mikkelsson J, Resendiz JC, et al. Polymorphisms of 
COX-1 and GPVI associate with the antiplatelet effect of aspirin in 
coronary artery disease patients. Thromb Haemost 2006;95:253–9. 
68. Smith SM, Judge HM, Peters G, et al. PAR-1 genotype influences 
platelet aggregation and procoagulant responses in patients with 
coronary 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 aspirin 
is under polygenic control of variants in the VAV3 and phospholipase 
C gamma 2 (PLCG2) genes (abstr). Circulation 2008;118 Suppl:326. 
70. Herrera-Galeano JE, Becker DM, Wilson AF, et al. A novel variant 
in the platelet endothelial aggregation receptor-1 gene is associated 
with increased platelet aggregability. Arterioscler Thromb Vasc Biol 
2008;28:1484 –90. 
71. Undas A, Brummel K, Musial J, Mann KG, Szczeklik A. PlA2 
polymorphism of {beta}3 integrins is associated with enhanced 
thrombin generation and impaired antithrombotic action of aspirin at 
the 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. Lancet 
1998;351:1253. 
73. Cooke GE, Liu-Stratton Y, Ferketich AK, et al. Effect of platelet 
antigen polymorphism on platelet inhibition by aspirin, clopidogrel, 
or their combination. J Am Coll Cardiol 2006;47:541– 6. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 19 
July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics 
74. Mathias R, Kim Y, Sung H, et al. A combined genome-wide linkage 
and association approach to find susceptibility loci for platelet function 
phenotypes in European American and African American families with 
coronary artery disease. BMC Med Genomics 2010;3:22. 
75. Faraday N, Yanek LR, Yang XP, et al. Identification of a specific 
intronic 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, Hamon 
M. Effect of PlA1/A2 glycoprotein IIIa gene polymorphism on the 
long-term outcome after successful coronary stenting. Thromb J 
2007;5:19 –25. 
77. Voora D, Horton J, Shah SH, Shaw LK, Newby LK. Polymorphisms 
associated with in vitro aspirin resistance are not associated with 
clinical outcomes in patients with coronary artery disease who report 
regular aspirin use. Am Heart J 2011;162:166 –72.e1. 
78. Chasman DI, Shiffman D, Zee RY, et al. Polymorphism in the 
apolipoprotein(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 genetic 
basis for idiosyncratic toxicity of warfarin and phenytoin. Epilepsy 
Res 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 cause 
warfarin 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. Nature 
2004;427:541– 4. 
83. Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 
haplotypes on transcriptional regulation and warfarin dose. N Engl 
J Med 2005;352:2285–93. 
84. Wang D, Chen H, Momary KM, Cavallari LH, Johnson JA, Sadee 
W. Regulatory polymorphism in vitamin K epoxide reductase com-plex 
subunit 1 (VKORC1) affects gene expression and warfarin dose 
requirement. 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 variant 
alters required warfarin dose. Blood 2008;111:4106 –12. 
88. Takeuchi F, McGinnis R, Bourgeois S, et al. A genome-wide 
association study confirms VKORC1, CYP2C9, and CYP4F2 as 
principal genetic determinants of warfarin dose. PLoS Genet 2009; 
5:e1000433. 
89. Cooper GM, Johnson JA, Langaee TY, et al. A genome-wide scan 
for common genetic variants with a large influence on warfarin 
maintenance 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 warfarin 
dose in carriers of the V433M variant. Mol Pharmacol 2009;75: 
1337–46. 
91. Higashi MK, Veenstra DL, Kondo LM, et al. Association between 
CYP2C9 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 CYP2C9 
and VKORC1 1173C/T genotype on the risk of hemorrhagic 
complications 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 of 
response to warfarin during initial anticoagulation. N Engl J Med 
2008;358:999 –1008. 
94. Zhang JE, Jorgensen AL, Alfirevic A, et al. Effects of CYP4F2 
genetic polymorphisms and haplotypes on clinical outcomes in 
patients initiated on warfarin therapy. Pharmacogenet Genomics 
2009;19:781–9. 
95. Caraco Y, Blotnick S, Muszkat M. CYP2C9 genotype-guided 
warfarin 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 genotyping 
reduces 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 dosing 
compared with standard of care practices in an orthopedic surgery 
population: a prospective, parallel cohort study. Ther Drug Monit 
2010;32:338–45. 
98. Voora D, Eby C, Linder MW, et al. Prospective dosing of warfarin 
based on cytochrome P-450 2C9 genotype. Thromb Haemost 
2005;93:700 –5. 
99. Anderson JL, Horne BD, Stevens SM, et al. Randomized trial of 
genotype-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 or 
stroke 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 in 
high-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 J 
2008;156:397– 404. 
104. Schelleman H, Klungel OH, Witteman JC, et al. Diuretic-gene 
interaction 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 genotype 
on metoprolol metabolism persists during long-term treatment. 
Pharmacogenetics 2002;12:465–72. 
106. Rau T, Wuttke H, Michels LM, et al. Impact of the CYP2D6 
genotype 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 CYP2D6 
genotype on metoprolol plasma concentration and beta-adrenergic 
inhibition 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 human 
cytochrome P450 enzymes involved in the metabolism of R()- and 
S(-)-carvedilol. Drug Metab Dispos 1997;25:970 –7. 
109. Venter CP, Joubert PH. Ethnic differences in response to beta 
1-adrenoceptor blockade by propranolol. J Cardiovasc Pharmacol 
1984;6:361– 4. 
110. Rathz DA, Brown KM, Kramer LA, Liggett SB. Amino acid 49 
polymorphisms 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-function 
polymorphism in a G-protein coupling domain of the human 
beta1-adrenergic receptor. J Biol Chem 1999;274:12670–4. 
112. Liu J, Liu ZQ, Tan ZR, et al. Gly389Arg polymorphism of 
beta1-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, Pauly 
DF. Beta 1-adrenergic receptor polymorphisms and antihypertensive 
response 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. Beta 
1-adrenergic receptor polymorphisms confer differential function and 
predisposition 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 alters 
cardiac 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 physiology 
of human adrenergic receptor polymorphisms. Annu Rev Pharmacol 
Toxicol 2003;43:381– 411. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
20 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 
Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 
118. Liggett SB, Cresci S, Kelly RJ, et al. A GRK5 polymorphism that 
inhibits beta-adrenergic receptor signaling is protective in heart 
failure. Nat Med 2008;14:510 –7. 
119. Kurnik D, Cunningham AJ, Sofowora GG, et al. GRK5 Gln41Leu 
polymorphism is not associated with sensitivity to beta(1)-adrenergic 
blockade in humans. Pharmacogenomics 2009;10:1581–7. 
120. White HL, de Boer RA, Maqbool A, et al. An evaluation of the 
beta-1 adrenergic receptor Arg389Gly polymorphism in individuals 
with heart failure: a MERIT-HF sub-study. Eur J Heart Fail 
2003;5:463– 8. 
121. Cresci S, Kelly RJ, Cappola TP, et al. Clinical and genetic modifiers 
of 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 association 
between adrenergic receptor genotypes and survival in heart failure 
patients treated with carvedilol or metoprolol. J Am Coll Cardiol 
2008;52:644 –51. 
123. de Groote P, Lamblin N, Helbecque N, et al. The impact of 
beta-adrenoreceptor gene polymorphisms on survival in patients with 
congestive 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 in 
patients 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 patients 
receiving 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 treatment 
outcomes in hypertension. Clin Pharmacol Ther 2008;84:715–21. 
127. Wuttke H, Rau T, Heide R, et al. Increased frequency of cytochrome 
P450 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 the 
CYP2D6 gene is associated with a lower heart rate and blood 
pressure in beta-blocker users. Clin Pharmacol Ther 2009;85:45–50. 
129. Fux R, Morike K, Prohmer AM, et al. Impact of CYP2D6 genotype 
on adverse effects during treatment with metoprolol: a prospective 
clinical study. Clin Pharmacol Ther 2005;78:378–87. 
130. Zineh I, Beitelshees AL, Gaedigk A, et al. Pharmacokinetics and 
CYP2D6 genotypes do not predict metoprolol adverse events or 
efficacy 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 in 
drug biotransformation. Anal Bioanal Chem 2008;392:1093–108. 
132. Xu P, Jiang ZP, Zhang BK, Tu JY, Li HD. Impact of MDR1 
haplotypes derived from C1236T, G2677T/A and C3435T on the 
pharmacokinetics of single-dose oral digoxin in healthy Chinese 
volunteers. Pharmacology 2008;82:221–7. 
133. Verstuyft C, Schwab M, Schaeffeler E, et al. Digoxin pharmacoki-netics 
and MDR1 genetic polymorphisms. Eur J Clin Pharmacol 
2003;58:809 –12. 
134. Zhao LM, He XJ, Qiu F, Sun YX, Li-Ling J. Influence of ABCB1 
gene 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, Sonawane 
B. Genetic polymorphism in N-acetyltransferase (NAT): population 
distribution of NAT1 and NAT2 activity. J Toxicol Environ Health 
B Crit Rev 2009;12:440 –72. 
136. Sabbagh A, Darlu P. SNP selection at the NAT2 locus for an 
accurate 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, Oates 
JA. Effect of acetylator phenotype on the rate at which procainamide 
induces antinuclear antibodies and the lupus syndrome. N Engl 
J Med 1978;298:1157–9. 
139. Mongey AB, Sim E, Risch A, Hess E. Acetylation status is associated 
with serological changes but not clinically significant disease in 
patients receiving procainamide. J Rheumatol 1999;26:1721– 6. 
140. Siddoway L, Thompson K, McAllister C, et al. Polymorphism of 
propafenone 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. Effect 
of CYP2D6*10 genotype on propafenone pharmacodynamics in 
Chinese patients with ventricular arrhythmia. Acta Pharmacol Sin 
2002;23:1040–4. 
142. Morike K, Kivisto KT, Schaeffeler E, et al. Propafenone for the 
prevention of atrial tachyarrhythmias after cardiac surgery: a random-ized, 
double-blind placebo-controlled trial. Clin Pharmacol Ther 
2008;84:104 –10. 
143. Lee JT, Kroemer HK, Silberstein DJ, et al. The role of genetically 
determined polymorphic drug metabolism in the beta-blockade 
produced by propafenone. N Engl J Med 1990;322:1764–8. 
144. Jazwinska-Tarnawska E, Orzechowska-Juzwenko K, Niewinski P, et 
al. The influence of CYP2D6 polymorphism on the antiarrhythmic 
efficacy of propafenone in patients with paroxysmal atrial fibrillation 
during 3 months propafenone prophylactic treatment. Int J Clin 
Pharmacol Ther 2001;39:288 –92. 
145. Thompson JF, Man M, Johnson KJ, et al. An association study of 43 
SNPs 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 statin 
therapy. 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 Pharmacogenetics 
Implementation Consortium guidelines for cytochrome P450-2C19 
(CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther 
2011;90:328 –32. 
Key Words: clopidogrel y pharmacogenetics y polymorphism y statins 
y warfarin. 
APPENDIX 
For additional loci, see the online version of this article. 
Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014

More Related Content

What's hot

The biomarker of choice in association of dyslipidemia with osteoporosis: A c...
The biomarker of choice in association of dyslipidemia with osteoporosis: A c...The biomarker of choice in association of dyslipidemia with osteoporosis: A c...
The biomarker of choice in association of dyslipidemia with osteoporosis: A c...SriramNagarajan17
 
A common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organsA common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organsKevin Jaglinski
 
Relapsed AML: Steve Kornblau
Relapsed AML: Steve KornblauRelapsed AML: Steve Kornblau
Relapsed AML: Steve Kornblauspa718
 
Galena presentation 11 jan 17
Galena presentation   11 jan 17Galena presentation   11 jan 17
Galena presentation 11 jan 17Galenabio
 
Herbal medicines in cardiac patients
Herbal medicines in cardiac patientsHerbal medicines in cardiac patients
Herbal medicines in cardiac patientsU2009
 
Galena presentation 9 feb 17
Galena presentation   9 feb 17Galena presentation   9 feb 17
Galena presentation 9 feb 17Galenabio
 
Evaluating the efficacy of different platelet rich plasma regimens for manage...
Evaluating the efficacy of different platelet rich plasma regimens for manage...Evaluating the efficacy of different platelet rich plasma regimens for manage...
Evaluating the efficacy of different platelet rich plasma regimens for manage...anestesiaudec
 
Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?
Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?
Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?Valentina Corona
 
translational biotechnology-Amgen -Geeta Iyer -4-48-16
translational biotechnology-Amgen -Geeta Iyer -4-48-16translational biotechnology-Amgen -Geeta Iyer -4-48-16
translational biotechnology-Amgen -Geeta Iyer -4-48-16Geeta Iyer
 
stem cells applications in hurler syndrome
stem cells applications in hurler syndrome stem cells applications in hurler syndrome
stem cells applications in hurler syndrome ZiadoonAlyaqoobi
 
Anticoagulación en Mieloma Múltiple
Anticoagulación en Mieloma MúltipleAnticoagulación en Mieloma Múltiple
Anticoagulación en Mieloma Múltipleevelyn-mejia
 
Fgf-23 and mortality risk in CKD
Fgf-23 and mortality risk in CKDFgf-23 and mortality risk in CKD
Fgf-23 and mortality risk in CKDarunchawlamd
 

What's hot (19)

The biomarker of choice in association of dyslipidemia with osteoporosis: A c...
The biomarker of choice in association of dyslipidemia with osteoporosis: A c...The biomarker of choice in association of dyslipidemia with osteoporosis: A c...
The biomarker of choice in association of dyslipidemia with osteoporosis: A c...
 
A common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organsA common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organs
 
Relapsed AML: Steve Kornblau
Relapsed AML: Steve KornblauRelapsed AML: Steve Kornblau
Relapsed AML: Steve Kornblau
 
Galena presentation 11 jan 17
Galena presentation   11 jan 17Galena presentation   11 jan 17
Galena presentation 11 jan 17
 
Herbal medicines in cardiac patients
Herbal medicines in cardiac patientsHerbal medicines in cardiac patients
Herbal medicines in cardiac patients
 
Horse versus rabbit atg
Horse versus rabbit atgHorse versus rabbit atg
Horse versus rabbit atg
 
Galena presentation 9 feb 17
Galena presentation   9 feb 17Galena presentation   9 feb 17
Galena presentation 9 feb 17
 
Chasman
ChasmanChasman
Chasman
 
Evaluating the efficacy of different platelet rich plasma regimens for manage...
Evaluating the efficacy of different platelet rich plasma regimens for manage...Evaluating the efficacy of different platelet rich plasma regimens for manage...
Evaluating the efficacy of different platelet rich plasma regimens for manage...
 
bioequivalence
bioequivalencebioequivalence
bioequivalence
 
Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?
Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?
Chloroquine or hydroxychloroquine for Covid-19: why might they be hazardous?
 
translational biotechnology-Amgen -Geeta Iyer -4-48-16
translational biotechnology-Amgen -Geeta Iyer -4-48-16translational biotechnology-Amgen -Geeta Iyer -4-48-16
translational biotechnology-Amgen -Geeta Iyer -4-48-16
 
Posterl Asgt
Posterl AsgtPosterl Asgt
Posterl Asgt
 
stem cells applications in hurler syndrome
stem cells applications in hurler syndrome stem cells applications in hurler syndrome
stem cells applications in hurler syndrome
 
158 genomic and proteomic risk factors
158 genomic and proteomic risk factors158 genomic and proteomic risk factors
158 genomic and proteomic risk factors
 
Strive Teleconf Presentation Aug10 2005
Strive Teleconf Presentation Aug10 2005Strive Teleconf Presentation Aug10 2005
Strive Teleconf Presentation Aug10 2005
 
Anticoagulación en Mieloma Múltiple
Anticoagulación en Mieloma MúltipleAnticoagulación en Mieloma Múltiple
Anticoagulación en Mieloma Múltiple
 
2015 Resume
2015 Resume2015 Resume
2015 Resume
 
Fgf-23 and mortality risk in CKD
Fgf-23 and mortality risk in CKDFgf-23 and mortality risk in CKD
Fgf-23 and mortality risk in CKD
 

Viewers also liked

Executive Summary DM and Pharmacogenetics
Executive Summary DM and PharmacogeneticsExecutive Summary DM and Pharmacogenetics
Executive Summary DM and PharmacogeneticsArmie Pacheco
 
clinica maternal happy baby
clinica maternal happy baby clinica maternal happy baby
clinica maternal happy baby natinicole
 
Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...
Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...
Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...Matthias Samwald
 
Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...
Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...
Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...Matthias Samwald
 
Genetic Profiling Debate
Genetic Profiling DebateGenetic Profiling Debate
Genetic Profiling DebateShaza El Nemr
 
WHMS PGx Presentation
WHMS PGx PresentationWHMS PGx Presentation
WHMS PGx PresentationOrion Cuffe
 
Pharmacogenetics
PharmacogeneticsPharmacogenetics
PharmacogeneticsDr. Almas A
 
Pharmacogenetics of antipsychotic and antidepressent
Pharmacogenetics of antipsychotic and antidepressentPharmacogenetics of antipsychotic and antidepressent
Pharmacogenetics of antipsychotic and antidepressentismail sadek
 
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics Zohaib HUSSAIN
 
Ecuaciones con mas de una funcion trigonometrica
Ecuaciones con mas de una funcion trigonometricaEcuaciones con mas de una funcion trigonometrica
Ecuaciones con mas de una funcion trigonometricaCipriano Arboleda
 
Pharmacogenetics & Teratogenicity
Pharmacogenetics & TeratogenicityPharmacogenetics & Teratogenicity
Pharmacogenetics & TeratogenicityDr Renju Ravi
 
Pharmacogenetics devang
Pharmacogenetics devangPharmacogenetics devang
Pharmacogenetics devangDevang Parikh
 

Viewers also liked (20)

FINAL MCP - PROJECT VEGGIES
FINAL MCP - PROJECT VEGGIESFINAL MCP - PROJECT VEGGIES
FINAL MCP - PROJECT VEGGIES
 
Executive Summary DM and Pharmacogenetics
Executive Summary DM and PharmacogeneticsExecutive Summary DM and Pharmacogenetics
Executive Summary DM and Pharmacogenetics
 
clinica maternal happy baby
clinica maternal happy baby clinica maternal happy baby
clinica maternal happy baby
 
Cameo_Fall2015PDF
Cameo_Fall2015PDFCameo_Fall2015PDF
Cameo_Fall2015PDF
 
Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...
Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...
Medinfo2013 - An RDF/OWL Knowledge Base for Query Answering and Decision Supp...
 
Presentation1
Presentation1Presentation1
Presentation1
 
CPIC Guidelines
CPIC GuidelinesCPIC Guidelines
CPIC Guidelines
 
Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...
Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...
Genomic CDS: an example of a complex ontology for pharmacogenetics and clinic...
 
Genetic Profiling Debate
Genetic Profiling DebateGenetic Profiling Debate
Genetic Profiling Debate
 
Dr. Chasman on Pharmacogenetics of Statin Therapies
Dr. Chasman on Pharmacogenetics of Statin TherapiesDr. Chasman on Pharmacogenetics of Statin Therapies
Dr. Chasman on Pharmacogenetics of Statin Therapies
 
WHMS PGx Presentation
WHMS PGx PresentationWHMS PGx Presentation
WHMS PGx Presentation
 
Pharmacogenetics
PharmacogeneticsPharmacogenetics
Pharmacogenetics
 
Pharmacogenetics of antipsychotic and antidepressent
Pharmacogenetics of antipsychotic and antidepressentPharmacogenetics of antipsychotic and antidepressent
Pharmacogenetics of antipsychotic and antidepressent
 
Nz pep lecture_jan2016
Nz pep lecture_jan2016Nz pep lecture_jan2016
Nz pep lecture_jan2016
 
Pharmacogenetics
PharmacogeneticsPharmacogenetics
Pharmacogenetics
 
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
Pharmacogenomics, Pharmacogenetics and Pharmacokinetics
 
Ecuaciones con mas de una funcion trigonometrica
Ecuaciones con mas de una funcion trigonometricaEcuaciones con mas de una funcion trigonometrica
Ecuaciones con mas de una funcion trigonometrica
 
Pharmacogenetics & Teratogenicity
Pharmacogenetics & TeratogenicityPharmacogenetics & Teratogenicity
Pharmacogenetics & Teratogenicity
 
Pharmacogenetics devang
Pharmacogenetics devangPharmacogenetics devang
Pharmacogenetics devang
 
Snp
SnpSnp
Snp
 

Similar to Key Pharmacogenetic Variants for Cardiovascular Drugs

2007aquilante-persmed
2007aquilante-persmed2007aquilante-persmed
2007aquilante-persmedpharmdude
 
Pharmacogenetics and Pharmacogenomics
Pharmacogenetics and PharmacogenomicsPharmacogenetics and Pharmacogenomics
Pharmacogenetics and PharmacogenomicsDr. Prashant Shukla
 
Statin drugs are they worth the risks
Statin drugs are they worth the risksStatin drugs are they worth the risks
Statin drugs are they worth the risksDIPAK PATADE
 
Drug drug gene interactions
Drug drug gene interactionsDrug drug gene interactions
Drug drug gene interactionsDRMOHITKHER
 
Gut_Bagatto_ Classic
Gut_Bagatto_ ClassicGut_Bagatto_ Classic
Gut_Bagatto_ ClassicJoseph Gut
 
Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...
Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...
Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...Mgfamiliar Net
 
AHA.Statin Toxicity.pdf
AHA.Statin Toxicity.pdfAHA.Statin Toxicity.pdf
AHA.Statin Toxicity.pdfAhFr1
 
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...wackysavior4064
 
Renal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdf
Renal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdfRenal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdf
Renal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdfHaramaya University
 
Raised Lipid Profile In Rheumatoid Arthritis- A Risk For CVD
Raised Lipid Profile In Rheumatoid Arthritis- A Risk For CVDRaised Lipid Profile In Rheumatoid Arthritis- A Risk For CVD
Raised Lipid Profile In Rheumatoid Arthritis- A Risk For CVDiosrjce
 
An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...
An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...
An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...Oxford Cancer Biomarkers
 
Update on genetics and molecular biology.pdf
Update on genetics and molecular biology.pdfUpdate on genetics and molecular biology.pdf
Update on genetics and molecular biology.pdfssuser5b0f5e
 
Present and Future Impact of Cytogenetics on Acute Myeloid Leukemia
Present and Future Impact of Cytogenetics on Acute Myeloid LeukemiaPresent and Future Impact of Cytogenetics on Acute Myeloid Leukemia
Present and Future Impact of Cytogenetics on Acute Myeloid Leukemialarriva
 
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...M. Luisetto Pharm.D.Spec. Pharmacology
 
Pharmacogenetics seminar
Pharmacogenetics seminarPharmacogenetics seminar
Pharmacogenetics seminarSiva Joy
 

Similar to Key Pharmacogenetic Variants for Cardiovascular Drugs (20)

2007aquilante-persmed
2007aquilante-persmed2007aquilante-persmed
2007aquilante-persmed
 
Pharmacogenetics and Pharmacogenomics
Pharmacogenetics and PharmacogenomicsPharmacogenetics and Pharmacogenomics
Pharmacogenetics and Pharmacogenomics
 
Statin drugs are they worth the risks
Statin drugs are they worth the risksStatin drugs are they worth the risks
Statin drugs are they worth the risks
 
Osu lesko 6 oct final
Osu lesko 6 oct finalOsu lesko 6 oct final
Osu lesko 6 oct final
 
Drug drug gene interactions
Drug drug gene interactionsDrug drug gene interactions
Drug drug gene interactions
 
Gut_Bagatto_ Classic
Gut_Bagatto_ ClassicGut_Bagatto_ Classic
Gut_Bagatto_ Classic
 
Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...
Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...
Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient- Centered App...
 
AHA.Statin Toxicity.pdf
AHA.Statin Toxicity.pdfAHA.Statin Toxicity.pdf
AHA.Statin Toxicity.pdf
 
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
Emerging MRA-Based Treatments for End-Stage Renal Disease (ESRD) Patients on ...
 
Renal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdf
Renal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdfRenal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdf
Renal and Cardiovascular Outcomes with Efpeglenatide in Type 2 Diabetes.pdf
 
Raised Lipid Profile In Rheumatoid Arthritis- A Risk For CVD
Raised Lipid Profile In Rheumatoid Arthritis- A Risk For CVDRaised Lipid Profile In Rheumatoid Arthritis- A Risk For CVD
Raised Lipid Profile In Rheumatoid Arthritis- A Risk For CVD
 
An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...
An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...
An evaluation of the clinical utility of a panel of variants in DPYD and ENOS...
 
Genomics in CVD
Genomics in CVDGenomics in CVD
Genomics in CVD
 
Update on genetics and molecular biology.pdf
Update on genetics and molecular biology.pdfUpdate on genetics and molecular biology.pdf
Update on genetics and molecular biology.pdf
 
Metastatic prostate cancer, nejm 2018
Metastatic prostate cancer, nejm 2018Metastatic prostate cancer, nejm 2018
Metastatic prostate cancer, nejm 2018
 
Present and Future Impact of Cytogenetics on Acute Myeloid Leukemia
Present and Future Impact of Cytogenetics on Acute Myeloid LeukemiaPresent and Future Impact of Cytogenetics on Acute Myeloid Leukemia
Present and Future Impact of Cytogenetics on Acute Myeloid Leukemia
 
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...Jpcr mauro luisetto  et al amyotrophic lateral sclerosis and endogenous esoge...
Jpcr mauro luisetto et al amyotrophic lateral sclerosis and endogenous esoge...
 
Adverse Drug Reaction
Adverse Drug ReactionAdverse Drug Reaction
Adverse Drug Reaction
 
Literature Evaluation.pptx
Literature Evaluation.pptxLiterature Evaluation.pptx
Literature Evaluation.pptx
 
Pharmacogenetics seminar
Pharmacogenetics seminarPharmacogenetics seminar
Pharmacogenetics seminar
 

Recently uploaded

Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...Miss joya
 
Call Girls Budhwar Peth 7001305949 All Area Service COD available Any Time
Call Girls Budhwar Peth 7001305949 All Area Service COD available Any TimeCall Girls Budhwar Peth 7001305949 All Area Service COD available Any Time
Call Girls Budhwar Peth 7001305949 All Area Service COD available Any Timevijaych2041
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Miss joya
 
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceCollege Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceNehru place Escorts
 
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiCall Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiNehru place Escorts
 
Hemostasis Physiology and Clinical correlations by Dr Faiza.pdf
Hemostasis Physiology and Clinical correlations by Dr Faiza.pdfHemostasis Physiology and Clinical correlations by Dr Faiza.pdf
Hemostasis Physiology and Clinical correlations by Dr Faiza.pdfMedicoseAcademics
 
Housewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment Booking
Housewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment BookingHousewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment Booking
Housewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment Bookingnarwatsonia7
 
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort ServiceCall Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Serviceparulsinha
 
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...narwatsonia7
 
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...narwatsonia7
 
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls ServiceCall Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Servicesonalikaur4
 
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...narwatsonia7
 
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowSonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowRiya Pathan
 
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...narwatsonia7
 
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...narwatsonia7
 
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableVip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableNehru place Escorts
 

Recently uploaded (20)

Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
 
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
 
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
College Call Girls Pune Mira 9907093804 Short 1500 Night 6000 Best call girls...
 
Call Girls Budhwar Peth 7001305949 All Area Service COD available Any Time
Call Girls Budhwar Peth 7001305949 All Area Service COD available Any TimeCall Girls Budhwar Peth 7001305949 All Area Service COD available Any Time
Call Girls Budhwar Peth 7001305949 All Area Service COD available Any Time
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
 
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
 
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort ServiceCollege Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
College Call Girls Vyasarpadi Whatsapp 7001305949 Independent Escort Service
 
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service ChennaiCall Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
Call Girls Service Chennai Jiya 7001305949 Independent Escort Service Chennai
 
Hemostasis Physiology and Clinical correlations by Dr Faiza.pdf
Hemostasis Physiology and Clinical correlations by Dr Faiza.pdfHemostasis Physiology and Clinical correlations by Dr Faiza.pdf
Hemostasis Physiology and Clinical correlations by Dr Faiza.pdf
 
Housewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment Booking
Housewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment BookingHousewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment Booking
Housewife Call Girls Hoskote | 7001305949 At Low Cost Cash Payment Booking
 
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort ServiceCall Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
 
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
 
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
 
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
 
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls ServiceCall Girls Thane Just Call 9910780858 Get High Class Call Girls Service
Call Girls Thane Just Call 9910780858 Get High Class Call Girls Service
 
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
 
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowSonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
 
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
 
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
 
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableVip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
 

Key Pharmacogenetic Variants for Cardiovascular Drugs

  • 1. 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 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. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 2. 10 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 to 20% reduced LDLc lowering with pravastatin (4) or simvasta-tin (5) due to an alternatively spliced HMGCR transcript that produces a version of HMGCR that is less sensitive to simvasta-tin inhibition (6). This haplotype was not identified in a GWAS with atorvastatin (7), and there-fore, whether this association holds for other statins is unclear. APOE. Two variants, rs7412 and rs429358, define 3 haplotypes, namely, 2, 3, and 4, that are associated with lipid, cognitive, and thrombotic traits. In general, the magnitude of LDLc lowering is greatest for carriers of 2, fol-lowed by 3, and 4 haplotypes, respectively. The influence of APOE haplotypes appears to be consistent across variety of statin types (4,7,8) and doses (8), and results in mild (15%) attenuations of LDLc lowering. Additional loci. Information on ABCB1 and ABCG2 can be found in the Online Appendix. Reduction in cardiovascular events. Statins prevent car-diovascular disease by lowering LDLc and potential pleio-tropic effects. There are few genetic predictors of these pleiotropic benefits, but the rs20455 polymorphism in kinesin-like protein 6 (KIF6, an underlying disease gene) may be a candidate. Carriers of the risk allele may receive a greater benefit from statin therapy compared to noncarriers despite equal LDLc, triglycerides, or C-reactive protein reduction (9,10). However, subsequent genome-wide meta-analysis of large randomized placebo-controlled statin trials found no evidence for association with coronary artery disease (11) or any differential treatment benefit with statin therapy (12,13). Statin-induced musculoskeletal side effects. Statins have a well-defined safety profile, but come with a small risk of musculoskeletal side effects (14). Genetic variants in the hepatic transporter, SLCO1B1, influence the risk of adverse events (Table 2); it does not appear that cytochrome P450 (CYP) SNPs influence statin-induced side effects. SLCO1B1. The solute carrier organic anion transporter family, member 1B1 gene (SLCO1B1, also referred to as SLC21A6, OATP-C, or OATP1B1) harbors many genetic variants, and each variant is numerically and sequentially labeled 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 the localization of this transporter to the hepatocyte plasma membrane (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- to 3-fold increased risk of CK- negative myopathy (19,20). In trials of randomly assigned statins as well as in observational studies, the risk for myopathy with *5 depends on 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 the clearance of these statins (16–18,24) and thus appear to be statin-specific. Adherence to statin therapy. Often, statin therapy is hampered 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) observed that carriers of the SLCO1B1*5 allele have a higher rate of statin nonadherence (20,25). Clinical implications for statin pharmacogenetics. It is unlikely that genetic testing for statin efficacy will enter clinical care since the magnitude of associations is small (10% to 15% differences in LDLc lowering), and physicians can reasonably forecast the magnitude of LDLc lowering based on statin type, dose, and baseline LDLc. In contrast, statin-induced side effects and nonadherence are less predictable. While the current level of evidence surrounding SLCO1B1*5 may not support prospective genotyping at this time, the test is currently offered on consumer-directed whole genome genotyping platforms (e.g., 23andMe, deCODEME, and so on). A potential strat-egy for prospective SLCO1B1*5 testing might offer carriers either pravastatin or rosuvastatin or fluvastatin as first-line Abbreviations and Acronyms ACS acute coronary syndrome(s) BP blood pressure CYP cytochrome P450 GWAS genome-wide association study HR heart rate INR international normalized ratio LDLc low-density lipoprotein cholesterol LVEF left ventricular ejection fraction MI myocardial infarction PCI percutaneous coronary intervention RF reduced function SNP single nucleotide polymorphism SoTuarbclees1of PShoaurmrcaecsoogfenPehtaicrmVaacroiagteionnetic Variation Category Description Types of Genes Examples Pharmacokinetic Variability in concentration of drug at site of drug effect Drug-metabolizing enzymes Drug transporters Warfarin: CYP2C9 Clopidogrel: CYP2C19 Simvastatin: SLCO1B1 Metoprolol: CYP2D6 Pharmacodynamic Variability in drug ability to influence its target Transmembrane receptors Intracellular enzymes Clopidogrel: P2RY12 Simvastatin: HMGCR Metoprolol: ADBR1 Underlying disease mechanisms Variability in disease being treated Often downstream or independent of drug target Hydrochlorothiazide: ADD1 Simvastatin: APOE Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 3. JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 11 July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics Figure 1 Categories of Major Cardiovascular Pharmacogenetic Variants Genetic variants that influence the response to cardiovascular agents fall into 3 broad categories: 1) pharmacokinetic; 2) pharmacodynamic; and 3) underlying disease mechanism. agents, which seem to depend the least on SLCO1B1 for their clearance. Thienopyridines Thienopyridines are effective in patients with acute coronary syndrome (ACS) and percutaneous coronary intervention (PCI); however, some patients remain at risk for death, myocardial infarction (MI), and stent thrombosis. Platelet function 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-netic); other loci, CYP2C9 (pharmacokinetic), PON1 (pharmacokinetic), and P2RY12 (pharmacodynamic) may also influence response (Table 3). Platelet function in response to clopidogrel. CYP2C19. Clopidogrel is an inactive prodrug activated by several enzymes, including CYP2C19, to an active metabolite that inhibits the platelet adenosine diphosphate receptor, P2RY12 (29). CYP2C19 *2 (rs4244285) is the most com-mon reduced-function (RF) variant; additional, rare variants mimic the *2 allele: *3 [rs4986893], *4 [rs28399504], *5 [rs56337013]) (30). An individual person’s genotype can be characterized in 3 ways: 1) presence of at least 1 RF allele (carrier vs. noncarrier); 2) the number of RF alleles (i.e., 0, 1, 2); or 3) the predicted, total CYP2C19 enzymatic activity (Table 4). Carriers of *2—also referred to as intermediate metabolizers or poor metabolizers—produce lower active metabolite and have attenuated platelet inhibi-tion (31). In general, there is a gene-dose effect where increasing number of RF alleles (i.e., 0, 1, 2, or extensive metabolizers, intermediate metabolizers, and poor metabo-lizers) predicts a decreasing amount of platelet inhibition (27,31,32). Carriers of another variant *17 (rs3758581, ultrametabolizers) exhibit increased CYP2C19 activity, pro-duce more active metabolite, and improved platelet inhibi-tion, in most reports (33,34). Higher loading and maintenance doses (e.g., 1,200 mg and 150 mg/day) appear, in part, to overcome the effects of the *2 allele (30,32,35,36), although not completely (37), and can require up to 300 mg/day (38). Ticlopidine (39), prasugrel (31,40,41), and ticagrelor (42) all produce uniform platelet inhibition in *2 carriers and noncarriers. ABCB1. The adenosine triphosphate-binding cassette, sub-family B (MDR/TAP), member 1 (ABCB1) gene encodes an apical membrane protein in enterocytes and hepatocytes and serves to reduce bioavailability. Clopidogrel appears to be handled by this transporter, and 3 SNPs—C1236T (rs1128503), G2677T (rs2032582), and C3435T (rs1045642)—capture the common genetic variation at this locus. Persons who carry a T allele at each SNPs (i.e., the T-T-T haplotype) produced reduced active metabolite (43) in an initial report. Despite this initial observation, the link to reduced platelet inhibi-tion has been difficult to establish (27,30), although may be present for persons who carry 2 copies of the T-T-T haplotype (44). As with CYP2C19 variants, prasugrel- Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 4. 12 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 Clinical 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 at ABCB1, P2RY12, CYP2C9, and PON1 can be found in the Online Appendix. Clinical response to clopidogrel. CYP2C19. In parallel with the platelet function data, the CYP2C19*2 allele is associated 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 (poor metabolizers) experience a 1.8-fold increase. This pattern also 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 genetic associations with platelet function are mirrored in the clinical response to clopidogrel in the setting of PCI. These observations formed the foundation for updating the clopi-dogrel label by the Food and Drug Administration to include pharmacogenetic information. Similarly, the gain of function variant, CYP2C19*17, is associated with increased risk of bleeding (50), and protection from ischemic events (51) CYP2C19*2 carriers treated with prasugrel or ticagrelor do not show a heightened risk of cardiovascular death, MI, stroke, or stent thrombosis (40,52). ABCB1. Carriers of 2 copies of the ABCB1 T-T-T haplo-type treated with clopidogrel are at an increased risk for subsequent death, MI, or stroke compared to persons who carried none, thus mirroring the platelet function data (44,45,52). Genetic variation at the ABCB1 locus does not appear to influence prasugrel- or ticagrelor-treated patients (40,52). Clinical implications. The current state of evidence reflects a strong and consistent association with the CYP2C19*2 allele and an increased risk for cardiovascular outcomes in ACS/ PCI patients treated with clopidogrel, although not other settings (51). Alternative antiplatelet agents (i.e., prasugrel and ticagrelor) mitigate the adverse risk of CYP2C19*2- associated adverse events. Although CYP2C19 genotyping is commercially available, whether genotype-guided therapy will improve outcomes or reduce costs is unknown. Con-sensus statements (53) currently do not recommend routine testing. However, there is sufficient evidence to support physicians who may choose to pursue CYP2C19*2 testing in selected patients: 1) for diagnosis in patients with compli-cations of clopidogrel therapy such as stent thrombosis in compliant clopidogrel users; or 2) for the choice of dual antiplatelet therapy in the ACS/PCI setting where the physician believes that additional information regarding the risk/benefit profile for clopidogrel will influence the choice of drug therapy (54). Outside of these scenarios, the ACS/PCI setting, or situations where there are ample data to guide drug choice (e.g., ST-segment elevation MI [55], diabetic patients [56], age 75 years, or prior MTaainblGee2neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRSetsaptoinnsse to Statins Caucasians Africans Asians Type of Study Effect of Variant Statin Type Ref. # Gene Variant(s) Statin Response/ Risk Allele/Haplotype Frequencies Class effect 7,8,145–147 by alleles at rs7412 and rs429358 LDLc lowering/GWAS, CG 2LDL reduction with 3 vs. 2 APOE 2, 3, and 4 haplotypes defined 2LDL reduction with 4 vs. 2 2: 15%–20%* 3: 90%–95% 4: 20%–30% 3% 6% LDLc lowering/CG 2LDL reduction in H7 carriers Simvastatin 4,5 HMGCR H7 haplotype defined by alleles at rs17244841, rs17238540, and rs3846662 Simvastatin, atorvastatin 19,20 1In CK positive symptoms 1In CK negative symptoms with SLCO1B1 *5: rs4149056 15% 1% 13% Musculoskeletal side effects/GWAS, CG risk allele Simvastatin, atorvastatin 20,25 SLCO1B1 *5: rs4149056 15% 1% 13% Nonadherence/CG 1Risk of nonadherence with risk allele *Represents Caucasian population. CG candidate gene association study; CK creatine kinase; GWAS genome-wide association study; LDLc low-density lipoprotein cholesterol. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 5. JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 13 July 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 a person is found to be a poor metabolizer (Table 4), then an alternative to clopidogrel, such as prasugrel or ticagrelor, should be considered. This seems preferable over increased clopidogrel doses, which have not shown benefit over standard dose clopidogrel (57–59). The greatest uncertainty is in the intermediate metabolizer group (i.e., those with 1 loss of function allele) and is where integration of other clinical risk factors such as diabetes, body mass index, cost, and other bleeding and thrombosis risk factors need to be considered in determining the therapy with the optimal risk:benefit profile. Aspirin Aspirin irreversibly inhibits prostaglandin G/H synthase 1 (PTGS1, or COX-1) and the conversion of arachidonic acid to thromboxane. With adequate dosing and compliance, aspirin is capable of completely inhibiting COX-1 in 99% of persons; thus, true “aspirin resistance” is rare (60). However, alternate agonists such as adenosine diphosphate and collagen can produce robust aggregation in the face of complete COX-1 inhibition (61), a response that demon-strates wide interindividual variability (62), heritability (63,64), and at high levels, association with future cardio-vascular events (65,66), therefore making aspirin a candidate for pharmacogenetic discovery. Because aspirin uniformly inhibits its target, COX-1, there are no pharmacokinetic or pharmacodynamic genetic considerations. Instead, investi-gators have focused on platelet function loci (underlying disease) and LPA (underlying disease). Variants in several platelet genes, namely, PEAR1, ITGB3, VAV3, GPVI, F2R, and GP1BA, are associated with platelet function in response to aspirin (67–70). One with moderate evidence is rs5918 in ITGB3 where carriers of the risk allele have heightened platelet function on aspirin (71–73). Recent GWAS of platelet function in response to aspirin have identified additional genetic loci that have yet to be replicated (74). Finally, the most robust association comes from a large study that found carriers of the minor allele for an intronic SNP, rs12041331, in PEAR1 have higher PEAR1 platelet protein content and heightened platelet function in response to aspirin (75). The link to an increased risk of clinical events in aspirin users has not been established despite several large studies (76,77), although the PEAR1 variant rs12041331 has not yet been tested. An uncommon variant in LPA (rs3798220) seems to modify the protective effects of aspirin: carriers had a more than 2-fold reduction in the risk for cardio-vascular disease with aspirin, whereas noncarriers (95% of Caucasians) had none in a large placebo-controlled clinical trial (78). Clinical implications. Because of the lack of consistent or preliminary associations with the variants described in the preceding text, there is currently no role for genetic testing for aspirin. Warfarin Three loci contribute to the heterogeneity in response: CYP2C9 (pharmacokinetic), VKORC1 (pharmacodynamic), and CYP4F2 (pharmacodynamic) (Table 5). Warfarin dose requirements. CYP2C9. CYP2C9 is re-sponsible for S-warfarin (the more active enantiomer) clear-ance. The 2 most common reduced-function variants are MTaainblGee3neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRCelsoppoidnosgeretol Clopidogrel Gene Variant(s) Risk Allele/ Haplotype Frequencies Drug Response/Type of Study Effect of Variant Caucasians Africans Asians With Risk Allele Ref. # CYP2C19 *2 (rs4244285) 16% 14% 27% Drug concentration, platelet function, recurrent MI, stent thrombosis/ GWAS, DMET, CG 2Active metabolite concentration 2Inhibition of platelet function 1Risk of recurrent MI, stroke, stent thrombosis Graded risk with 0, 1, or 2 alleles 31,32,45–47,52 CYP2C19 *17 (rs3758581) 5% 10% 6% Drug concentration, platelet function, bleeding/CG 1Active metabolite concentration 1Inhibition of platelet function 1Risk bleeding 32,33,50 ABCB1 T-T-T haplotype defined by T allele at C1236T (rs1128503), G2677T (rs2032582), and C3435T (rs1045642) 71% 12% 65% Drug concentration, platelet function, recurrent MI, stroke, death/CG 2Active metabolite concentration 2No change in inhibition of platelet function 1Risk of recurrent MI, stroke, death (in carriers of 2 T-T-T haplotypes) 27,30,43,45,48,49,52 MI myocardial infarction; DMET drug metabolizing enzyme and transporter panel; other abbreviations as in Table 2. CYTPab2lCe149 DipClYoPty2pCe1C9laDsispilfioctyapteionC*lassification* 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 *Adapted from Ingelman-Sundberg et al. (147) and Scott et al. (148). Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 6. 14 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 *2 (rs1799853) and *3 (rs1057910). The CYP2C9*2 allele reduces warfarin clearance by 30% and the *3 allele by 90% (79), which translate into 19% and 33% reductions per allele in warfarin dose requirements compared to noncarriers (80). VKORC1. Vitamin K epoxide reductase complex, subunit 1 (VKORC1) is warfarin’s target (81,82), and resequencing this gene identified 2 haplotypes (A and B defined by the alleles at 5 SNPS: rs7196161, rs9923231, rs9934438, rs8050894, and rs2359612) that influence VKORC1 gene expression and predict warfarin dose requirements (83). Of these 5 SNPs, an A allele at rs9923231 (also referred to as 1639 GA and the 3673 SNP) in the promoter region is responsible for reducing: VKORC1 gene expression (84). Therefore, carriers of an A allele at rs9923231 (or the A haplotype) require a 30% per allele reduction in warfarin dose requirements (85). CYP4F2. Several GWAS have not only confirmed the observations above but also have identified a novel associa-tion between rs2108622 in CYP4F2 and reduced hepatic CYP4F2, higher levels of hepatic vitamin K, and higher warfarin dose requirements (86–90). Additional loci. Additional information regarding CALU and GGCX can be found in the Online Appendix. Clinical response to warfarin. RETROSPECTIVE STUDIES. Standard dosing algorithms (i.e., a 5 mg or 10 mg loading dose followed by titration based on the INR) lead to an increased risk of an out-of-range INR (4.0) or a delay in the time-to-therapeutic range INR in carriers of the CYP2C9*2 or CYP2C9*3 alleles (91,92), the A haplotype or A allele of the 1639 SNP in VKORC1 (93), or the T allele in CYP4F2 (94). Importantly, only variants in CYP2C9 have been linked to the risk of bleeding (91,92). PROSPECTIVE STUDIES OF TAILORED WARFARIN THERAPY. Small randomized pilot studies, parallel cohort studies, and single arm studies with or without historical controls have prospectively explored genetically guided warfarin therapy. Whereas some studies suggest a benefit over standard therapy (95–97), others failed to show any significant advantage (98) except for smaller and fewer dosing changes and INR measurements (99). Definitive clinical trials are ongoing to assess the benefit of incorporating genetics into warfarin therapy. Clinical implications. There are strong genetic associa-tions surrounding CYP2C9, VKORC1, and CYP4F2 variants that influence the response to warfarin. Commercial testing and algorithms that assist in the interpretation of genotypes are available. Therefore, there is evidence to justify and tools to enable genotype-guided warfarin therapy. Until large scale trials demonstrate a benefit for routine testing, physi-cians may choose to pursue testing in selected patients who they feel may benefit by: 1) diagnosing those with compli-cations from warfarin therapy (e.g., hemorrhage); 2) pre-dicting dose for those at high risk of bleeding (e.g., “triple-therapy” with aspirin, clopidogrel, and warfarin); or 3) weighing the costs of newer anticoagulants against warfarin. Diuretics Blood pressure–lowering response. The adducin 1 (alpha) gene (ADD1, underlying disease) is implicated in animal and human sodium handling (100), and persons with a Gly460Trp substitution are more salt sensitive and have enhanced blood pressure (BP) lowering with thiazide di-uretics (100). Clinical outcomes. In an observational study, patients with Trp460 who were treated with thiazide diuretics experienced a greater protection from MI and stroke than patients with Gly460 (101). However, this difference in the protective effects of diuretics could not be replicated in several larger studies (102–104). Clinical implications. Given the inconsistent associations and lack of a commercially available test, the use of this genetic variant in guiding treatment options in hypertension is limited. MTaainblGee5neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRWesaprfoanrisne to Warfarin Gene Variant(s) Risk Allele Frequencies Drug Response/ Type of Study Effect of Variant in Caucasians Africans Asians Carriers of Risk Allele Ref. # CYP2C9 *2 (rs1799853) *3 (rs1057910) *2: 10% *3: 6% 1% 1% 1% 4% Drug concentration, warfarin dose requirements, out-of-range INR values, hemorrhage/GWAS, CG 2Clearance of S-warfarin 2Dose requirements 1Risk of out-of-range INR values 1Time to stable, therapeutic INR 1Risk of hemorrhage 79,80,88,89,91–93 VKORC1 1639 (rs9923231)* 60% 98% 2% Warfarin dose requirements, out-of-range INR values/ linkage (human/animal), GWAS, CG 2Dose requirements 1Risk of out-of-range INR values 1Time to stable, therapeutic INR 81–83,88,89 CYP4F2 rs2108622 23% 6% 25% Warfarin dose requirements/ GWAS, DMET 1Dose requirements 87–89 *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. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 7. JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 15 July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics Beta-Blockers Beta-adrenergic receptor antagonists (or beta-blockers) are a diverse class of agents that primarily antagonize the beta-1 adrenergic receptor, encoded by ADBR1. Accordingly, changes in heart rate (HR), BP, and left ventricular ejection fraction (LVEF) are considered the surrogate responses in place 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 common RF variant CYP2D6*4 (rs3892097) results in the absence of CYP2D6 activity. Carriers have a higher systemic exposure to metoprolol (105), which translates into a greater reduc-tion in HR and BP (106,107). Beta-blockers such as atenolol and carvedilol (108) do not require CYP2D6 for their metabolism. ADRB1. Ethnic differences in the dose response for pro-pranolol (109) motivated pharmacodynamic genetic inves-tigations of beta-blockers. Two variants in ADBR1, the Ser49Gly (rs1801252) and Arg389Gly (rs1801253), lead to impaired down-regulation (110) and higher signal transduc-tion, respectively (111). Therefore, carriers of either variant have enhanced, beta-1-receptor activity and more beta-blocker sensitivity. Healthy volunteers and patients with hypertension who carry 2 Arg389 variants have a greater HR (112) or BP (113) reduction mainly with metoprolol, although not with all beta-blockers (114). In patients with systolic heart failure treated with either metoprolol or carvedilol (115), but not bucindolol (116), carriers of 2 copies of the ADBR1 Arg389 variant had significantly greater improvements in LVEF compared to the Gly389 carriers. ADRB2. Genetic variation in the beta-2 adrenergic receptor (ADBR2) centers around 2 variants: Arg16Gly (rs1042713) and Glu27Gln (rs1042714). Receptors with Gly16 versus Arg16 have enhanced down-regulation of ADBR2. In contrast, receptors with Glu27 appear to be resistant to down-regulation (117). Extension to variation in BP or HR lowering in response to beta-blocker therapy has not been demonstrated (114). GRK5. Downstream of the beta-1-adrenergic receptor are G-protein coupled receptor kinases responsible for desensi-tization of the beta-1-adrenergic receptor. A Glu41Leu genetic variant in 1 of these kinases, G protein-coupled receptor kinase 5 (GRK5), is more prevalent in African Americans. The Leu41 more effectively uncoupled isoproterenol-stimulated responses than GRK5-Q41, thus producing a pharmacological-like “beta-blockade” in mice (118), although with no differences in atenolol-induced HR reduction in humans (119). MTaainblGee6neticMAasinsoGceianteitoincsAWssitohcitahteioRnsesWpoitnhstehteoRBeestpao-AnsderetnoerBgeictaR-AedcreepnteorrgAicntRaegcoenpistotsr Antagonists Caucasians Africans Asians Carriers of Risk Allele Beta-Blocker Type Ref. # Risk Allele Frequencies Drug Response/Type of Study Gene Variant(s) Effect of Variant in Metoprolol 105,106,127,128 2Clearance 1Reduction in BP and HR 1Risk of bradycardia CYP2D6 *4 (rs3892097) 18% 23% Drug concentration, HR and BP reduction/CG 113,115,116 ADRB1 Ser49Gly (rs1801252) 20% 1% 1% HR, BP reduction/CG 2Reduction in BP and HR Metoprolol 112 ADRB1 Arg389Gly (rs1801253) 31% 41% 20% HR, improvement in LVEF, Metoprolol (BP/HR/LVEF) Carvedilol (LVEF) Bucindolol (hospitalization/death) 2Reduction in BP and HR 2Improvement in LVEF BP reduction, hospitalization or death in CHF/CG BP blood pressure; CG candidate gene association study; HR heart rate; LVEF left ventricular ejection fraction. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 8. 16 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 Clinical 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 reduction in the time to first hospitalization or death when treated with bucindolol (116) but not carvedilol or metoprolol (120–125) compared to Gly389 carriers. Whether these differences are due to drug specific effects (i.e., bucindolol vs. metoprolol) or the play of chance has not been adequately tested. In patients with chronic coronary artery disease randomly assigned to verapamil or atenolol, carriers of at least 1 copy of the Ser49/ Arg389 haplotype had a 9- versus 2-fold worsened prognosis when assigned to verapamil versus atenolol (126) despite equivalent BP and HR control in both groups. ADRB2. One study examined the influence of genetic variation at ADRB2 and its influence on ACS patients and found those homozygous for both the Arg16 and Glu27 alleles had a 20% rate of subsequent death versus 6% of those homozygous for both Gly16 and Gln27 (125). This trend has been observed in some studies of patients with heart failure (124), although not all (122,123,126). GRK5. Extension of the associations of the Glu41Leu variant to clinical outcomes of patients was demonstrated in 1 study where Leu41 carriers exhibited improvement in sur-vival compared to Glu41 carriers (118), although not in another, 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 Arg389 variant have: 1) enhanced reduction in HR and BP; 2) larger improvements in LVEF; and 3) longer survival when treated with chronic beta-blocker therapy compared to persons with the Gly389 variant. Although it is unlikely that beta-blocker therapy will ever be withheld for carriers of the Gly389 variant, a potential application of these findings would 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 Gly389 variant. Because certain beta-blockers such as atenolol and carve-dilol are minimally handled by CYP2D6 (131), these may be reasonable alternates for carriers of CYP2D6*4 with metoprolol-induced bradycardia. Commercial testing for CYP2D6*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 with altered 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 been observed (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 to be related to variation in its pharmacokinetics, the induction of drug-induced lupus-associated autoantibodies (138,139), but not symptomatic, drug-induced lupus (138,139) has been linked to the slow acetylator status. Propafenone. The hepatic hydroxylation by CYP2D6 of propafenone into 5-hydroxypropafenone demonstrates wide interindividual variability (140). Carriers of CYP2D6*4 (a reduced function allele) have reduced propafenone clearance compared to noncarriers (140–142). Antiarrhythmic efficacy. At low doses of propafenone, CYP2D6*4 carriers have greater reduction in exercise- or isoproterenol-induced HR compared to noncarriers (143), although not with higher doses or without the “stress” of exercise/isoproterenol (140,142,143). Further, CYP2D6*4 carriers have enhanced suppression of atrial and ventricular arrhythmias compared to noncarriers in some studies (141,144), although not in all (140,142) Therefore, there are insufficient data to make any conclusions regarding CYP2D6 genotype and antiarrhythmic efficacy. Toxicity. Central nervous system side effects with propafenone, as well as excessive beta-blockade, are correlated with 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 do not sufficiently translate to consistent associations of mean-ingful clinical outcomes. Therefore, there is currently no role for pharmacogenetic testing in the clinical use of these medications. Future Directions The data to date support many common variants that alter the 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 variable clinical effects are also candidates for a pharmacogenetic approach. Novel systems approaches may elucidate additional deter-minants of drug response. Broad “omics” approaches have several advantages over traditional genetics-based research, for example, 1) responding to the environment, and 2) elucidating how the activity of entire pathways, instead of individual genes, influence drug response. The field of pharmacogenomics would benefit from the development of thresholds of evidence for testing and coverage by insurance. With the expanding number and complexity of “-omic” markers of drug response is an equally Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 9. JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 17 July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics profound evidentiary gap between association studies and those that demonstrate clinical utility. It may be unrealistic to wait (or to require) a prospective, events-driven random-ized controlled trial of each drug-marker combination. Even when such trials are conducted, the results may be made obsolete by new observations and/or approval of new drugs. Therefore, genetic substudies of clinical trials and registries may become the highest levels of evidence for the potential benefits of using pharmacogenetic testing. Such standards are common in many fields of medicine where there is often a lack of randomized clinical trial data. Observational studies and comparative effectiveness research will lay the foundation 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: deepak.voora@duke.edu or geoffrey.ginsburg@duke.edu. REFERENCES 1. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2009;361: 1045–57. 2. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2007;357:2001–15. 3. Simon JA, Lin F, Hulley SB, et al. Phenotypic predictors of response to simvastatin therapy among African-Americans and Caucasians: the Cholesterol and Pharmacogenetics (CAP) study. Am J Cardiol 2006;97:843–50. 4. Chasman DI, Posada D, Subrahmanyan L, Cook NR, Stanton VP Jr., 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 the 3-hydroxyl-3-methylglutaryl coenzyme a reductase gene is associated with racial differences in low-density lipoprotein cholesterol response to simvastatin treatment. Circulation 2008;117:1537– 44. 6. Medina MW, Gao F, Ruan W, Rotter JI, Krauss RM. Alternative splicing of 3-hydroxy-3-methylglutaryl coenzyme A reductase is associated with plasma low-density lipoprotein cholesterol response to 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 in the Treating to New Targets (TNT) cohort. Circ Cardiovasc Genet 2009;2:173– 81. 8. Voora D, Shah SH, Reed CR, et al. Pharmacogenetic predictors of statin-mediated low-density lipoprotein cholesterol reduction and dose response. Circ Cardiovasc Genet 2008;1:100–6. 9. Iakoubova OA, Sabatine MS, Rowland CM, et al. Polymorphism in KIF6 gene and benefit from statins after acute coronary syndromes: results from the PROVE IT-TIMI 22 study. J Am Coll Cardiol 2008;51:449 –55. 10. Iakoubova OA, Tong CH, Rowland CM, et al. Association of the Trp719Arg polymorphism in kinesin-like protein 6 with myocardial infarction and coronary heart disease in 2 prospective trials: the CARE and WOSCOPS trials. J Am Coll Cardiol 2008;51:435– 43. 11. Assimes TL, Ho´lm H, Kathiresan S, et al. Lack of association between the Trp719Arg polymorphism in kinesin-like protein-6 and coronary artery disease in 19 case-control studies. J Am Coll Cardiol 2010;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 Cardiol 2011;57:2000 –7. 13. Ridker PM, MacFadyen JG, Glynn RJ, Chasman DI. Kinesin-Like Protein 6 (KIF6) polymorphism and the efficacy of rosuvastatin in primary prevention/clinical perspective. Circ Cardiovasc Genet 2011; 4:312–7. 14. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 2005;366:1267–78. 15. Kameyama Y, Yamashita K, Kobayashi K, Hosokawa M, Chiba K. Functional characterization of SLCO1B1 (OATP-C) variants, SLCO1B1*5, SLCO1B1*15 and SLCO1B1*15C1007G, by using transient expression systems of HeLa and HEK293 cells. Pharma-cogenet Genomics 2005;15:513–22. 16. Pasanen MK, Neuvonen M, Neuvonen PJ, Niemi M. SLCO1B1 polymorphism markedly affects the pharmacokinetics of simvastatin acid. Pharmacogenet Genomics 2006;16:873–9. 17. Pasanen MK, Fredrikson H, Neuvonen PJ, Niemi M. Different effects of SLCO1B1 polymorphism on the pharmacokinetics of atorvastatin and rosuvastatin. Clin Pharmacol Ther 2007;82:726 –33. 18. Niemi M, Pasanen MK, Neuvonen PJ. SLCO1B1 polymorphism and sex 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 and statin-induced myopathy—a genomewide study. N Engl J Med 2008;359:789 –99. 20. Voora D, Shah SH, Spasojevic I, et al. The SLCO1B1*5 genetic variant is associated with statin-induced side effects. J Am Coll Cardiol 2009;54:1609 –16. 21. Puccetti L, Ciani F, Auteri A. Genetic involvement in statins induced 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 to moderate muscular symptoms with high-dosage statin therapy in hyperlipidemic patients—the PRIMO study. Cardiovasc Drugs Ther 2005;19:403–14. 23. Jacqueline Suk Danik, MacFadyen J, Nyberg F, Ridker P. Lack of association between polymorphisms in the slco1b1 gene and clinical myalgia 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 of OATP1B1 genotype on the pharmacokinetics of rosuvastatin in Koreans. 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 intolerance in routinely treated individuals with type 2 diabetes: a Go-DARTS study. Clin Pharmacol Ther 2010;89:210–6. 26. Gurbel PA, Bliden KP, Guyer K, et al. Platelet reactivity in patients and 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 of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA 2009;302:849 –57. 28. Breet NJ, van Werkum JW, Bouman HJ, et al. Comparison of platelet function tests in predicting clinical outcome in patients undergoing coronary stent implantation. JAMA 2010;303:754–62. 29. Savi P, Pereillo JM, Uzabiaga MF, et al. Identification and biological activity of the active metabolite of clopidogrel. Thromb Haemost 2000;84:891– 6. 30. Gladding P, Webster M, Zeng I, et al. The pharmacogenetics and pharmacodynamics of clopidogrel response: an analysis from the PRINC (Plavix Response in Coronary Intervention) trial. J Am Coll Cardiol Intv 2008;1:620 –7. 31. Brandt JT, Close SL, Iturria SJ, et al. Common polymorphisms of CYP2C19 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 polymorphisms and the impact of a higher clopidogrel dose regimen on active metabolite 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. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 10. 18 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 34. Frére C, Cuisset T, Gaborit B, Alessi MC, Hulot JS. The CYP2C19*17 allele is associated with better platelet response to clopidogrel 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 increased clopidogrel maintenance dose or lansoprazole co-administration on the antiplatelet response to clopidogrel in CYP2C19-genotyped healthy subjects. J Thromb Haemost 2010;8:610 –3. 36. Gladding P, White H, Voss J, et al. Pharmacogenetic testing for clopidogrel using the rapid INFINITI analyzer: a dose-escalation study. J Am Coll Cardiol Intv 2009;2:1095–101. 37. Jeong Y-H, Kim I-S, Park Y, et al. Carriage of cytochrome 2C19 polymorphism is associated with risk of high post-treatment platelet reactivity on high maintenance-dose clopidogrel of 150 mg/day: results of the ACCEL-DOUBLE (Accelerated Platelet Inhibition by a 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 clopidogrel resistance? Circulation 2009;119:2854 –7. 39. Maeda A, Ando H, Asai T, et al. Differential impacts of CYP2C19 gene polymorphisms on the antiplatelet effects of clopidogrel and ticlopidine. Clin Pharmacol Ther 2011;89:229 –33. 40. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P450 genetic polymorphisms and the response to prasugrel. Relationship to phar-macokinetic, pharmacodynamic, and clinical outcomes. Circulation 2009;119:2553– 60. 41. Varenhorst C, James S, Erlinge D, et al. Genetic variation of CYP2C19 affects both pharmacokinetic and pharmacodynamic re-sponses to clopidogrel but not prasugrel in aspirin-treated patients with coronary artery disease. Eur Heart J 2009;30:1744 –52. 42. Tantry US, Bliden KP, Wei C, et al. First analysis of the relation between CYP2C19 genotype and pharmacodynamics in patients treated with ticagrelor versus clopidogrel/clinical perspective. Circ Cardiovasc Genet 2010;3:556–66. 43. Taubert D, von Beckerath N, Grimberg G, et al. Impact of P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther 2006;80:486 –501. 44. Mega JL, Close SL, Wiviott SD, et al. Genetic variants in ABCB1 and CYP2C19 and cardiovascular outcomes after treatment with clopidogrel 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 determinants of response to clopidogrel and cardiovascular events. N Engl J Med 2009;360:363–75. 46. Collet J-P, Hulot J-S, Pena A, et al. Cytochrome P450 2C19 polymorphism 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 2C19 loss-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 in clopidogrel-treated patients according to cytochrome P450 2C19*2 loss-of-function allele or proton pump inhibitor coadministration: a systematic meta-analysis. J Am Coll Cardiol 2010;56:134–43. 49. Mega JL, Simon T, Collet J-P, et al. Reduced-function CYP2C19 genotype and risk of adverse clinical outcomes among patients treated with clopidogrel predominantly for PCI: a meta-analysis. JAMA 2010;304:1821–30. 50. Sibbing D, Koch W, Gebhard D, et al. Cytochrome 2C19*17 allelic variant, platelet aggregation, bleeding events, and stent thrombosis in clopidogrel-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 on outcomes of clopidogrel treatment. N Engl J Med 2010;363:1704–14. 52. Wallentin L, James S, Storey RF, et al. Effect of CYP2C19 and ABCB1 single nucleotide polymorphisms on outcomes of treatment with ticagrelor versus clopidogrel for acute coronary syndromes: a genetic substudy of the PLATO trial. Lancet 2010;376:1320–8. 53. Holmes DR Jr., Dehmer GJ, Kaul S, Leifer D, O’Gara PT, Stein CM. ACCF/AHA clopidogrel clinical alert: approaches to the FDA “boxed warning”: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents and the American Heart Association. J Am Coll Cardiol 2010;56: 321–41. 54. Scott SA, Sangkuhl K, Gardner EE, et al. Clinical pharmacogenetics implementation consortium guidelines for cytochrome P450-2C19 (CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther 2011;90:328 –32. 55. Montalescot G, Wiviott SD, Braunwald E, et al. Prasugrel compared with clopidogrel in patients undergoing percutaneous coronary inter-vention for ST-elevation myocardial infarction (TRITON-TIMI 38): double-blind, randomised controlled trial. Lancet 2009;373: 723–31. 56. Wiviott SD, Braunwald E, Angiolillo DJ, et al. Greater clinical benefit of more intensive oral antiplatelet therapy with prasugrel in patients with diabetes mellitus in the trial to assess improvement in therapeutic outcomes by optimizing platelet inhibition with prasugrel—Thrombolysis in Myocardial Infarction 38. Circulation 2008;118:1626 –36. 57. The CURRENT-OASIS 7 Investigators. Dose comparisons of clopidogrel and aspirin in acute coronary syndromes. N Engl J Med 2010;363:930–42. 58. Price MJ, Berger PB, Teirstein PS, et al. Standard- vs high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention. JAMA 2011;305:1097–105. 59. Parodi G, Marcucci R, Valenti R, et al. High residual platelet reactivity after clopidogrel loading and long-term cardiovascular events among patients with acute coronary syndromes undergoing PCI. JAMA 2011;306:1215–23. 60. Becker DM, Segal J, Vaidya D, et al. Sex differences in platelet reactivity and response to low-dose aspirin therapy. JAMA 2006;295: 1420–7. 61. Gurbel PA, Bliden KP, DiChiara J, et al. Evaluation of dose-related effects 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 in aspirin-treated cardiovascular patients. Thromb Haemost 2011;106: 466–74. 63. Faraday N, Yanek LR, Mathias R, et al. Heritability of platelet responsiveness to aspirin in activation pathways directly and indirectly related to cyclooxygenase-1. Circulation 2007;115:2490–6. 64. Shen H, Herzog W, Drolet M, et al. Aspirin resistance in healthy drug-naive men versus women (from the Heredity and Phenotype Intervention Heart Study). Am J Cardiol 2009;104:606 –12. 65. Frelinger AL III, Li Y, Linden MD, et al. Association of cyclooxygenase-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, Huisman MV. Association of laboratory-defined aspirin resistance with a higher risk of recurrent cardiovascular events: a systematic review and meta-analysis. Arch Intern Med 2007;167:1593–9. 67. Lepantalo A, Mikkelsson J, Resendiz JC, et al. Polymorphisms of COX-1 and GPVI associate with the antiplatelet effect of aspirin in coronary artery disease patients. Thromb Haemost 2006;95:253–9. 68. Smith SM, Judge HM, Peters G, et al. PAR-1 genotype influences platelet aggregation and procoagulant responses in patients with coronary 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 aspirin is under polygenic control of variants in the VAV3 and phospholipase C gamma 2 (PLCG2) genes (abstr). Circulation 2008;118 Suppl:326. 70. Herrera-Galeano JE, Becker DM, Wilson AF, et al. A novel variant in the platelet endothelial aggregation receptor-1 gene is associated with increased platelet aggregability. Arterioscler Thromb Vasc Biol 2008;28:1484 –90. 71. Undas A, Brummel K, Musial J, Mann KG, Szczeklik A. PlA2 polymorphism of {beta}3 integrins is associated with enhanced thrombin generation and impaired antithrombotic action of aspirin at the 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. Lancet 1998;351:1253. 73. Cooke GE, Liu-Stratton Y, Ferketich AK, et al. Effect of platelet antigen polymorphism on platelet inhibition by aspirin, clopidogrel, or their combination. J Am Coll Cardiol 2006;47:541– 6. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 11. JACC Vol. 60, No. 1, 2012 Voora and Ginsburg 19 July 3, 2012:9–20 Clinical Cardiovascular Pharmacogenetics 74. Mathias R, Kim Y, Sung H, et al. A combined genome-wide linkage and association approach to find susceptibility loci for platelet function phenotypes in European American and African American families with coronary artery disease. BMC Med Genomics 2010;3:22. 75. Faraday N, Yanek LR, Yang XP, et al. Identification of a specific intronic 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, Hamon M. Effect of PlA1/A2 glycoprotein IIIa gene polymorphism on the long-term outcome after successful coronary stenting. Thromb J 2007;5:19 –25. 77. Voora D, Horton J, Shah SH, Shaw LK, Newby LK. Polymorphisms associated with in vitro aspirin resistance are not associated with clinical outcomes in patients with coronary artery disease who report regular aspirin use. Am Heart J 2011;162:166 –72.e1. 78. Chasman DI, Shiffman D, Zee RY, et al. Polymorphism in the apolipoprotein(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 genetic basis for idiosyncratic toxicity of warfarin and phenytoin. Epilepsy Res 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 cause warfarin 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. Nature 2004;427:541– 4. 83. Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med 2005;352:2285–93. 84. Wang D, Chen H, Momary KM, Cavallari LH, Johnson JA, Sadee W. Regulatory polymorphism in vitamin K epoxide reductase com-plex subunit 1 (VKORC1) affects gene expression and warfarin dose requirement. 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 variant alters required warfarin dose. Blood 2008;111:4106 –12. 88. Takeuchi F, McGinnis R, Bourgeois S, et al. A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. PLoS Genet 2009; 5:e1000433. 89. Cooper GM, Johnson JA, Langaee TY, et al. A genome-wide scan for common genetic variants with a large influence on warfarin maintenance 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 warfarin dose in carriers of the V433M variant. Mol Pharmacol 2009;75: 1337–46. 91. Higashi MK, Veenstra DL, Kondo LM, et al. Association between CYP2C9 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 CYP2C9 and VKORC1 1173C/T genotype on the risk of hemorrhagic complications 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 of response to warfarin during initial anticoagulation. N Engl J Med 2008;358:999 –1008. 94. Zhang JE, Jorgensen AL, Alfirevic A, et al. Effects of CYP4F2 genetic polymorphisms and haplotypes on clinical outcomes in patients initiated on warfarin therapy. Pharmacogenet Genomics 2009;19:781–9. 95. Caraco Y, Blotnick S, Muszkat M. CYP2C9 genotype-guided warfarin 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 genotyping reduces 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 dosing compared with standard of care practices in an orthopedic surgery population: a prospective, parallel cohort study. Ther Drug Monit 2010;32:338–45. 98. Voora D, Eby C, Linder MW, et al. Prospective dosing of warfarin based on cytochrome P-450 2C9 genotype. Thromb Haemost 2005;93:700 –5. 99. Anderson JL, Horne BD, Stevens SM, et al. Randomized trial of genotype-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 or stroke 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 in high-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 J 2008;156:397– 404. 104. Schelleman H, Klungel OH, Witteman JC, et al. Diuretic-gene interaction 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 genotype on metoprolol metabolism persists during long-term treatment. Pharmacogenetics 2002;12:465–72. 106. Rau T, Wuttke H, Michels LM, et al. Impact of the CYP2D6 genotype 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 CYP2D6 genotype on metoprolol plasma concentration and beta-adrenergic inhibition 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 human cytochrome P450 enzymes involved in the metabolism of R()- and S(-)-carvedilol. Drug Metab Dispos 1997;25:970 –7. 109. Venter CP, Joubert PH. Ethnic differences in response to beta 1-adrenoceptor blockade by propranolol. J Cardiovasc Pharmacol 1984;6:361– 4. 110. Rathz DA, Brown KM, Kramer LA, Liggett SB. Amino acid 49 polymorphisms 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-function polymorphism in a G-protein coupling domain of the human beta1-adrenergic receptor. J Biol Chem 1999;274:12670–4. 112. Liu J, Liu ZQ, Tan ZR, et al. Gly389Arg polymorphism of beta1-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, Pauly DF. Beta 1-adrenergic receptor polymorphisms and antihypertensive response 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. Beta 1-adrenergic receptor polymorphisms confer differential function and predisposition 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 alters cardiac 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 physiology of human adrenergic receptor polymorphisms. Annu Rev Pharmacol Toxicol 2003;43:381– 411. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014
  • 12. 20 Voora and Ginsburg JACC Vol. 60, No. 1, 2012 Clinical Cardiovascular Pharmacogenetics July 3, 2012:9–20 118. Liggett SB, Cresci S, Kelly RJ, et al. A GRK5 polymorphism that inhibits beta-adrenergic receptor signaling is protective in heart failure. Nat Med 2008;14:510 –7. 119. Kurnik D, Cunningham AJ, Sofowora GG, et al. GRK5 Gln41Leu polymorphism is not associated with sensitivity to beta(1)-adrenergic blockade in humans. Pharmacogenomics 2009;10:1581–7. 120. White HL, de Boer RA, Maqbool A, et al. An evaluation of the beta-1 adrenergic receptor Arg389Gly polymorphism in individuals with heart failure: a MERIT-HF sub-study. Eur J Heart Fail 2003;5:463– 8. 121. Cresci S, Kelly RJ, Cappola TP, et al. Clinical and genetic modifiers of 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 association between adrenergic receptor genotypes and survival in heart failure patients treated with carvedilol or metoprolol. J Am Coll Cardiol 2008;52:644 –51. 123. de Groote P, Lamblin N, Helbecque N, et al. The impact of beta-adrenoreceptor gene polymorphisms on survival in patients with congestive 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 in patients 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 patients receiving 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 treatment outcomes in hypertension. Clin Pharmacol Ther 2008;84:715–21. 127. Wuttke H, Rau T, Heide R, et al. Increased frequency of cytochrome P450 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 the CYP2D6 gene is associated with a lower heart rate and blood pressure in beta-blocker users. Clin Pharmacol Ther 2009;85:45–50. 129. Fux R, Morike K, Prohmer AM, et al. Impact of CYP2D6 genotype on adverse effects during treatment with metoprolol: a prospective clinical study. Clin Pharmacol Ther 2005;78:378–87. 130. Zineh I, Beitelshees AL, Gaedigk A, et al. Pharmacokinetics and CYP2D6 genotypes do not predict metoprolol adverse events or efficacy 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 in drug biotransformation. Anal Bioanal Chem 2008;392:1093–108. 132. Xu P, Jiang ZP, Zhang BK, Tu JY, Li HD. Impact of MDR1 haplotypes derived from C1236T, G2677T/A and C3435T on the pharmacokinetics of single-dose oral digoxin in healthy Chinese volunteers. Pharmacology 2008;82:221–7. 133. Verstuyft C, Schwab M, Schaeffeler E, et al. Digoxin pharmacoki-netics and MDR1 genetic polymorphisms. Eur J Clin Pharmacol 2003;58:809 –12. 134. Zhao LM, He XJ, Qiu F, Sun YX, Li-Ling J. Influence of ABCB1 gene 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, Sonawane B. Genetic polymorphism in N-acetyltransferase (NAT): population distribution of NAT1 and NAT2 activity. J Toxicol Environ Health B Crit Rev 2009;12:440 –72. 136. Sabbagh A, Darlu P. SNP selection at the NAT2 locus for an accurate 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, Oates JA. Effect of acetylator phenotype on the rate at which procainamide induces antinuclear antibodies and the lupus syndrome. N Engl J Med 1978;298:1157–9. 139. Mongey AB, Sim E, Risch A, Hess E. Acetylation status is associated with serological changes but not clinically significant disease in patients receiving procainamide. J Rheumatol 1999;26:1721– 6. 140. Siddoway L, Thompson K, McAllister C, et al. Polymorphism of propafenone 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. Effect of CYP2D6*10 genotype on propafenone pharmacodynamics in Chinese patients with ventricular arrhythmia. Acta Pharmacol Sin 2002;23:1040–4. 142. Morike K, Kivisto KT, Schaeffeler E, et al. Propafenone for the prevention of atrial tachyarrhythmias after cardiac surgery: a random-ized, double-blind placebo-controlled trial. Clin Pharmacol Ther 2008;84:104 –10. 143. Lee JT, Kroemer HK, Silberstein DJ, et al. The role of genetically determined polymorphic drug metabolism in the beta-blockade produced by propafenone. N Engl J Med 1990;322:1764–8. 144. Jazwinska-Tarnawska E, Orzechowska-Juzwenko K, Niewinski P, et al. The influence of CYP2D6 polymorphism on the antiarrhythmic efficacy of propafenone in patients with paroxysmal atrial fibrillation during 3 months propafenone prophylactic treatment. Int J Clin Pharmacol Ther 2001;39:288 –92. 145. Thompson JF, Man M, Johnson KJ, et al. An association study of 43 SNPs 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 statin therapy. 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 Pharmacogenetics Implementation Consortium guidelines for cytochrome P450-2C19 (CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther 2011;90:328 –32. Key Words: clopidogrel y pharmacogenetics y polymorphism y statins y warfarin. APPENDIX For additional loci, see the online version of this article. Downloaded From: http://content.onlinejacc.org/pdfAccess.ashx?url=/data/Journals/JAC/24346/ on 09/05/2014