Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 1.
Published in final edited form as: Pharmacogenomics. 2012 Mar;13(4):465–475. doi: 10.2217/pgs.12.2

Clinical utility of pharmacogenetic biomarkers in cardiovascular therapeutics: a challenge for clinical implementation

Frank S Ong 1,2,*,, Joshua L Deignan 3, Jane Z Kuo 1,2, Kenneth E Bernstein 1, Jerome I Rotter 1,2,4,5, Wayne W Grody 3,5, Kingshuk Das 3,
PMCID: PMC3306231  NIHMSID: NIHMS361643  PMID: 22380001

Abstract

In the past decade, significant strides have been made in the area of cardiovascular pharmacogenomic research, with the discovery of associations between certain genotypes and drug-response phenotypes. While the motivations for personalized and predictive medicine are promising for patient care and support a model of health system efficiency, the implementation of pharmacogenomics for cardiovascular therapeutics on a population scale faces substantial challenges. The greatest obstacle to clinical implementation of cardiovascular pharmacogenetics may be the lack of both reproducibility and agreement about the validity and utility of the findings. In this review, we present the scientific evidence in the literature for diagnostic variants for the US FDA-labeled cardiovascular therapies, namely CYP2C19 and clopidogrel, CYP2C9/VKORC1 and warfarin, and CYP2D6/ADRB1 and β-blockers. We also discuss the effect of HMGCR/LDLR in decreasing the effectiveness of low-density lipoprotein cholesterol with statin therapy, the SLCO1B1 genotype and simvastatin myotoxicity, and ADRB1/ADD1 for antihypertensive response.

Keywords: biomarker, cardiovascular, clinical utility, clopidogrel, drug label, genetics, personalized medicine, pharmacogenetics, predictive medicine, warfarin


In the past decade, significant strides have been made in the area of cardiovascular pharmacogenomic research, with the discovery of association between certain genotypes and drug-response phenotypes. These findings led the US FDA to add pharmacogenomic biomarkers to drug labels of many commonly prescribed drugs. In the cardiovascular and hematology therapeutic areas, these drugs include carvedilol, clopidogrel, isosorbide/hydralazine, metoprolol, prasugrel, pravastatin, propafenone, propranolol, ticagrelor and warfarin [101]. With the aim of aiding the healthcare provider in arriving at a well-informed clinical decision, these labels include boxed warnings, dosage and administration information, precautionary statements, drug interactions, clinical pharmacology information or any combination thereof. While the motivations for personalized and predictive medicine – reduction of serious adverse effects, moderation of variability in treatment efficacy, and augmentation of therapy effectiveness based on individual genetic variation – are promising for patient care and support a model of health system efficiency, the implementation of pharmacogenomics for cardiovascular therapeutics on a population scale faces substantial challenges.

One obvious hurdle is the required threshold of evidence for a particular variant prior to clinical translation. Not only does the evidence for clinical utility have to be robust, an increased value above and beyond current testing practices needs to be clearly demonstrated. The pharmacokinetic or pharmacodynamic effect of a given genotype needs to result in a predictable difference in drug efficacy, response or adverse effects. Given that a particular variant reaches a significant level of evidence in a large clinical trial, the next obstacle would be for the provider to act upon this knowledge with objective practice guidelines and easy access to the genotype of the individual patient. Both the provider and patient would need to have a positive perception and acceptance of genetic testing. The genotyping would need to be of high standards and be performed in a Clinical Laboratory Improvement Amendments (CLIA)-approved laboratory. For implementation at point-of-care, the genomic information would already have to be part of the electronic medical record system, which comes with its own ethical, privacy and security issues [1]. Finally, the compensation for such testing would also need to be addressed.

The greatest obstacle to clinical implementation of cardiovascular pharmacogenetics may be the lack of both reproducibility and agreement about the validity and utility of the findings. In this review, we present the scientific evidence in the literature for diagnostic variants for the FDA-labeled cardiovascular therapies, namely CYP2C19 and clopidogrel, CYP2C9/VKORC1 and warfarin, and CYP2D6/ADRB1 and β-blockers (Table 1). We also discuss the effect of HMGCR/LDLR in decreasing the effectiveness of low-density lipoprotein (LDL)-cholesterol with statin therapy, the SLCO1B1 genotype and simvastatin myotoxicity, and ADRB1/ADD1 for antihypertensive response. Even with the challenges facing the clinical implementation of pharmacogenetics in cardiovascular therapeutics, personalized and predictive medicine in this arena holds great promise, and in a few centers, such as Vanderbilt University Medical Center (TN, USA) and others in the NIH Pharmacogenomics Research Network (PGRN), it has already begun. The prospective findings from these institutions that are pursuing clinical implementation will provide further insight into the challenges and future direction for pharmacogenetics. There are also ongoing prospective randomized clinical trials that will likely result in future revisions to guidelines.

Table 1.

Cardiovascular therapeutics, associated genes and gene variants, associated allele effect and minor allele frequency in different populations.

Drug Associated gene(s) Associated gene variant(s) Associated allele effect Minor allele frequency in different populations
Clopidogrel CYP2C19 CYP2C19*2 (c.681G>A; rs4244285) Loss-of-function, lower antiplatelet effect, higher risk of cardiovascular events CEU 14%; YRI/ASW 14%; CHB/JPT 26–28%
CYP2C19*17 (c.-806C>T; rs12248560) Gain-of-function, increased drug sensitivity, increased adverse bleeding side effect CEU 23%; YRI/ASW 29%; CHB/JPT 0–2%

Warfarin VKORC1/CYP2C9 VKORC1 (−1639G>A; rs9923231) Increased drug sensitivity, reduced dose requirement CEU 40%; YRI/ASW 3–10%; CHB/JPT 91–95%
CYP2C9*2 (p.Arg144Cys; rs1799853) Reduced drug clearance, reduced dose requirement CEU 10%; YRI/ASW 0%; CHB/JPT 0%
CYP2C9*3 (p.Ile359Leu; rs1057910) Reduced drug clearance, reduced dose requirement CEU 6%; YRI/ASW 0–3%; CHB/JPT 2–4%

β-blockers ADRB1 ADRB1 (p.Ser49Gly; rs1801252) More optimal blood pressure control, increased left ventricular ejection fraction CEU data not available; YRI/ASW 1%; CHB/JPT 0%
ADRB1 (p.Arg389Gly; rs1801253) More optimal blood pressure control, increased left ventricular ejection fraction CEU 31%; YRI/ASW 42%; CHB/JPT 15–25%

Statins HMGCR/SLCO1B1 HMGCR H7 (c.451-174A>T; rs17244841) Decreased reduction in LDL-cholesterol Data not available
HMGCR H7 (c.1564--106A>G; rs3846662) Decreased reduction in LDL-cholesterol CEU 43%; YRI/ASW 88–95%; CHB/JPT 54%
HMGCR H7 (c.2298+117T>G; rs17238540) Decreased reduction in LDL-cholesterol Data not available
SLCO1B1*5 (p.Val174Ala; rs4149056) Increased risk of developing myopathy or rhabdomyolysis CEU 15%; YRI/ASW 1–5%; CHB/JPT 12–14%

Data from International HapMap Project Data Rel 28 Phases II and III, August 2010 [116], on NCBI B36 assembly, dbSNP b126.

ASW: African ancestry in southwestern USA; CEU: Utah residents with northern and western European ancestry from the CEPH collection; CHB: Han Chinese in Beijing, China; JPT: Japanese in Tokyo, Japan; LDL: Low-density lipoprotein; YRI: Yoruban in Ibadan, Nigeria.

Cardiovascular therapeutics & genomic variants

CYP2C19 & clopidogrel

Clopidogrel is indicated for patients at high risk for acute coronary syndrome without ST-segment elevation, ST elevation myocardial infarction and prevention of vascular ischemic events in patients with symptomatic atherosclerosis. Aspirin/clopidogrel combination therapy is the standard of care for prevention of thrombosis after placement of intracoronary stent, and clopidogrel is also an alternative antiplatelet drug to aspirin. In fact, in 2007, clopidogrel was the second most widely prescribed drug in the USA and worldwide sales reached US$6.6 billion in 2009.

Clopidogrel is a thienopyridine prodrug, requiring CYP2C19 [2] and other enzymes in the CYP450 superfamily to transform it to an active thiol derivative [3,4]. It is well recognized that the interindividual variation to response of clopidogrel is heritable [511]. Several studies have reproducibly shown that CYP2C19*2 allele (c.681G>A; rs4244285) carriers are associated with a reduced ability to metabolize clopidogrel to form the active metabolite (loss of function), a lower antiplatelet effect, and a higher risk for cardiovascular events [1227]. Meta-analyses have shown that CYP2C19*2 carriers treated with clopidogrel were associated with higher rates of major adverse cardiovascular events and higher risk of stent thrombosis compared with noncarriers [13,26]. There has also been an association with the gain-of-function CYP2C19*17 variant (c.-806C>T; rs12248560) with increased drug sensitivity and adverse bleeding side effects [11,28,29]. In terms of population genetics, there appears to be a strong correlation with ancestry to the heritability of the alleles [11,29]. The loss-of-function CYP2C19*2 allele frequencies range from around 15% in European and African ancestries versus 29% in east Asians [11]. Conversely, the gain-of-function CYP2C19*17 alleles range from 16 to 21% in subjects of European and African ancestries compared with 3% in east Asian subjects [29]. There is also some evidence of loss-of-function *3 variant, contributing to poorer response in east Asians [11]. These results suggest that CYP2C19 testing may be even more imperative in that region of the world; however, there needs to be more education and awareness of pharmacogenomics before implementation can be carried out on a widescale in these populations.

Despite the robust association between the CYP2C19*2 genotype with lower clopidogrel response, large-scale studies with coronary artery disease subjects with lower risks for thromboembolic events failed to show the same association [30]. Therefore, clinical utility may be limited to only patients with high risk for recurrent events [31]. Nevertheless, the FDA approved a boxed warning of clopidogrel in March 2010 (Table 2), advising that alternative antiplatelet therapy be used in patients that are homozygous for the CYP2C19*2 allele [102]. Despite this warning, the FDA has not mandated genetic testing for CYP2C19 status before initiation of clopidogrel, leading to some confusion among healthcare providers regarding clinical implementation and personalized therapy. Furthermore, the American College of Cardiology Foundation and the American Heart Association have recently suggested that in the absence of prospective randomized trials, the evidence is insufficient to recommend genetic testing [32,33]. Recently, the ELEVATE-TIMI 56 (NCT01235351 [103]), a multicenter, randomized, double-blinded trial, showed that among patients with stable cardiovascular disease, tripling the maintenance dose of clopidogrel to 225 mg daily in CYP2C19*2 heterozygotes achieved levels of platelet reactivity similar to the standard 75 mg dose in noncarriers while in CYP2C19*2 homozygotes, even a 300 mg daily dose was unlikely to result in optimal degree of platelet inhibition [34]. It will be years before the data from other ongoing prospective randomized clinical trials of genotype-directed antiplatelet therapy will become available. In the meantime, the NIH-funded Clinical Pharmacogenetics Implementation Consortium (CPIC) of PGRN has published guidelines to recommend use of genetic information to guide clopidogrel therapy, also noting that the evidence is compelling for a relationship between CYP2C19 genotype and clopidogrel response in acute coronary syndrome patients who have undergone percutaneous coronary intervention [35].

Table 2.

US FDA warning label for clopidogrel and warfarin and FDA-approved testing platforms and assays for the analyte(s).

Drug Biomarker(s) Label sections Date of FDA warning FDA-approved pharmacogenetic testing platforms and assays Effective date
Clopidogrel CYP2C19 Boxed warning
Warnings and Precautions
Dosage and Administration
March 2010 Affymetrix GeneChip System 3000DX (Roche AmpliChip® CYP450 Test) January 2005
AutoGenomics Infiniti Analyzer November 2010

Warfarin CYP2C9
VKORC1
Precautions
Dosage and Administration
August 2007, updated January 2010 Verigene® System September 2007
AutoGenomics Infiniti Analyzer January 2008
Cepheid SmartCycler® Dx System (ParagonDx Rapid Genotyping Assay – CYP2C9 and VKORC1) April 2008
Osmetech Molecular Systems eSensor® XT-8 System July 2008
Roche Diagnostics LightCycler® Ver1.2 (TrimGen Corporation eQ-PCR LC Warfarin Genotyping Kit) February 2009
GenMark Diagnostics eSensor® XT-8 System (DNA Genotek Oragene-Dx collection device) December 2011

US FDA drug label.

US FDA Center for Devices and Radiological Health Clinical Laboratory Improvement Amendments database.

CYP2C9, VKORC1 & warfarin

Warfarin is the most widely prescribed oral anticoagulant drug in North America and probably the world. It is indicated for the prophylaxis or treatment of venous thrombosis and pulmonary embolism. It is also indicated for the prophylaxis or treatment of thromboembolic complications associated with atrial fibrillation, cardiac valve replacement and myocardial infarction. In patients with atrial fibrillation, anticoagulation with warfarin is over twice as effective in secondary prevention of stroke as any other alternatives, including other antithrombotic drugs [36] or surgical intervention [37]. Despite its effectiveness, warfarin interacts with many commonly used medications (simvastatin, metronidazole, macrolides and other broad-spectrum antibiotics), alcohol and some foods (particularly fresh plant-based foods containing vitamin K) resulting in either reduced effectiveness or increased toxicity.

Patients on this drug are monitored by a trial and error approach for safety and narrow therapeutic index based on the International Normalized Ratio. The wide interpatient variability in dosing requirement suggests a highly genetic contribution to the pharmacokinetics and pharmacodynamics of the drug [38]. The association of higher risk of bleeding as well as the under- or over-anticoagulation with certain genotypes prompted the FDA to place a warning label on the drug (Table 2) in August 2007 to include information on CYP2C9 and VKORC1 genotypes as predictors of dose response, and in January 2010, the FDA further revised the warning to include specific dosage recommendations for these genotypes [104,105]. However, as with clopidogrel, the agency stopped short of mandating pharmacogenetic testing.

Warfarin is a synthetic derivative of dicoumarol, which is a derivative from coumarin. The related 4-hydroxycoumarin-containing molecules decrease blood coagulation by inhibiting VKORC1, an enzyme that recycles the epoxide and quinine form of oxidized vitamin K to its reduced nonoxidized form after it has participated in the carboxylation of several blood coagulation proteins, mainly prothrombin and factor VII. The onset of effect requires approximately a day before clotting factors being normally made by the liver have time to naturally disappear in metabolism, and the duration of action of a single dose of racemic warfarin is 2–5 days. Multiple linked variants in VKORC1 (−1639G>A; rs99232318 and 1173C>T; rs9934438) have been associated with variability in gene expression, increased sensitivity to warfarin, and reduction in dose requirement [3943]. However, only the VKORC1 −1639G>A is a functional variant [44]. Additional identification of variants in CYP2C9*2 (p.Arg144Cys; rs1799853) and *3 (p.Ile359Leu; rs1057910) alleles are associated with 40–70% reduction in (S)-warfarin clearance and approximately 20–40% lower warfarin dose requirement, respectively [4548]. As in the case of clopidogrel, there are population and racial differences in minor allele frequencies of important variants as well as sensitivity to warfarin dosage requirement [40,49,50]. The CYP2C9*2 and *3 alleles and VKORC1 −1639G>A genotype explain approximately 50–60% of the variability in dosage in Caucasians but only approximately 20–25% of variability in African–Americans [40,48,5154]. The VKORC1 genotype has also been shown to be the strongest predictor of warfarin dosage in Japanese subjects [43,55]. Whether VKORC1 and CYP2C9 variants are the primary contributors to the drug response in African–Americans remains to be seen in studies currently in progress.

Despite the plethora of information from multiple genome-wide association studies, the strong support from the International Warfarin Pharmacogenetics Consortium (IWPC) and CPIC, in addition to the FDA warning labels and the availability of several FDA-approved platforms for warfarin genotyping, the clinical implementation of genetic testing for warfarin dosing has not only been disappointing in practice, but also the current consensus guidelines by the American College of Chest Physicians actually warn against the routine use of genetic data to guide dosing [38]. Similarly, the American College of Medical Genetics endorses testing only in cases of unusual warfarin response [56]. These conclusions have generally cited the suboptimal predictive value of the pharmacogenetic test over large populations [57]. There is a lack of evidence from large-scale prospective randomized trials, but such studies are currently under way – COAG (NCT00839657 [106]), GIFT (NCT01006733 [107]) and EU-PACT (NCT01119300 [108]) trials. Randomized clinical trials thus far have found the benefit of a genotype-guided dosing algorithm as a predictor of final stable dose compared with the clinical-only algorithm; however, the studies have not demonstrated a clear advantage of pharmacogenetic dosing on anticoagulation control [40,51,58,59]. By contrast, two prospective studies show a relationship between reduced hospitalization and better prediction of maintenance dose with the healthcare provider having knowledge of warfarin genetic information [45,60].

The clinical evidence shows that there is clinical and analytical validity of warfarin pharmacogenetics for Caucasians and possibly for other racial groups. However, whether the clinical utility of genetic testing for warfarin has been unequivocally established is still debatable. Even if the evidence is favorable for testing from prospective randomized trials and the guidelines are modified to reflect that, the challenge still remains in obtaining quality (CLIA-approved) clinical laboratory genotyping with quick turnaround time, compared with the turnaround time for international normalized ratio, if not eventually necessitating pre-emptive genotyping information in the medical electronic system for point-of-care access.

CYP2D6, ADRB1 & β-blockers

Clinicians have long known about the relationship of ancestry and drug response, especially in the case of heart failure, exemplified by the improvement in survival in African–Americans treated with hydralazine and nitrate, in addition to standard therapy. Three commonly prescribed β-blockers for heart failure (metoprolol, carvedilol and propranolol) have FDA labeling regarding the CYP2D6 polymorphisms [109111]. However, the evidence for pharmaco-kinetic effect of this genotype does not appear to translate into differences in efficacy, response or adverse effects, where dose titration is based on clinical surrogates such as blood pressure and heart rate.

In heart failure therapeutic pharmaco-genomics, the most robust evidence concerns the association of ADRB1 variants (two common nonsynonymous polymorphisms Ser49Gly; rs1801252 and Arg389Gly; rs1801253) and left ventricular ejection fraction or clinical outcomes mediated by β-blockade [6163]. The BEST (NCT00000560) suggested that Arg389Arg patients had significant benefits from bucindolol, whereas Gly389 carriers had no significant benefit [61]. This has led to probably the first case of pharmacogenomically guided drug development for the treatment of cardiovascular disease. The pharmaceutical company plans to launch a superiority trial in 3200 ADRB1 Arg389Arg patients randomized to metoprolol or bucindolol. Other studies, however, have not documented an association between Arg389Gly genotype and improved outcome with β-blockade [64,65]. There are also studies suggesting association of β-blockers for heart failure with ADRB2, ADRA2C and GRK5 [6668]. Thus, the literature suggests that while some subjects with heart failure may derive little benefit from β-blockade, in other patients, genetic variability in the adrenergic signaling pathway may have an important influence on the benefits of the drug. Nevertheless, it is difficult to clinically implement testing based on the consensus guideline use of β-blockers in patients with heart failure with the suggestion to withhold therapy without providing an alternative therapeutic option.

ADRB1, ADD1 & antihypertensives

As with heart failure and β-blockade, there are consistent data in several clinical studies showing association in differential blood pressure lowering levels with polymorphisms of the ADRB1 gene, namely Ser49Gly and Arg389Gly [63,6971]. In atenolol-treated patients with hypertension, the Ser49/Arg389 haplotype was associated with improved outcome, especially in lower death rate, in comparison with verapamil [71]. It may be that these variants demonstrate enhanced agonist-induced adenylyl cyclase activation by Gly49 compared with Ser49 and by Arg389 compared with Gly389 as shown in vitro. There has been equivocal data regarding the association of Gly460Trp variant in the α-adducin gene ADD1 with response to thiazide [7176]. This polymorphism in ADD1 has also been associated with increased risk of myocardial infarction or stroke during thiazide diuretic treatment [77]. But analyses in both the ALLHAT (NCT00000542 [112]) and the INVEST (NCT00133692 [113]) studies were unable to replicate this finding [78,79]. Nevertheless, this association led to the development of a novel antihypertensive drug class targeting adducin, namely ouabain, which has shown impressive Phase II data regarding blood pressure lowering efficacy [80,81].

HMGCR, LDLR, SLCO1B1 & statins

Another class of widely prescribed drugs for the prevention of cardiovascular events in coronary artery disease is the HMGCR inhibitors. The mechanism by which these inhibitors lower plasma LDL-cholesterol is via attenuation of endogenous production of cholesterol by increasing expression of the LDLR [82]. The H7 haplotype involves three intronic polymorphisms – rs17244841, rs3846662, rs17238540 – in the HMGCR and has been associated with 11–19% decreased reduction in LDL-cholesterol in both ethnically diverse and multiple independent populations [72,8388]. Other haplotypes – HMGCR H2 and LDLR L5 [8688], have also been associated with decreased attenuation of LDL-cholesterol. Additional studies have shown association with variability in statin efficacy with variants in CYP3A4 [89,90]. The FDA recently recommended that the higher dose of 80 mg/day of simvastatin be restricted to patients taking the medication for 12 months or longer [114]. Most patients who are at risk for developing myopathy or fatal rhabdomyolysis would most likely show clinical symptoms of adverse side effects within a few weeks of treatment. The FDA recommendation was based on the finding from the SEARCH (NCT00124072 [115]) consortium, where patients homozygous or heterozygous for a variant (rs4363657) in the SLCO1B1 gene were found to have a 16.9- and 4.5-fold increase, respectively, in developing myopathy or rhabdomyolysis at the 80 mg/day higher dose [91]. SLCO1B1 encodes the uptake transporter organic anion-transporting polypeptide 1B1 [92]. The identified polymorphism tags a known nonsynonymous variant SLCO1B1*5 (p.Val174Ala; rs4149056), which has been replicated in a separate cohort in the original study [91] and subsequently in two other studies [93,94]. Another study also showed that Val174Ala predicted the discontinuation of another popular statin drug, atorvastatin, due to adverse side effects [93]. Cerivastatin-associated rhabdomyolysis has also been associated with this variant in SLCO1B1 [95]. These studies demonstrate how dosage and duration of therapy interact with genetics to produce a drug response phenotype. However, it also appears that a biomarker that can modulate risk may not necessarily be of high clinical utility because of low predictive value (<20% for developing myopathy in patients homozygous for the Val174Ala allele). The SLCO1B1 genotype may predict a large effect on risk of simvastatin-associated myotoxicity in some settings and not in others. Current studies are exploring whether genomic markers have predictive value in identifying subjects with developing coronary events while on statin therapy.

Future perspective

With the ‘$1000’ whole-genome sequencing within reach in the next few years, it will not be the cost of genotyping or sequencing that will deter the progress of personalized and predictive medicine. The greatest obstacle to clinical implementation of cardiovascular pharmaco-genetics may be the lack of both reproducibility and agreement about the validity and utility of the findings. Once the scientific and medical communities are in agreement as to the clinical validity and utility of these pharmacogenetic tests, objective practice guidelines may be developed. Instantaneous access to the genotype information for point-of-care treatment of the individual patient may also be a great obstacle as the genomic information needs to be in the electronic medical record system pre-emptively, which raises its own ethical, legal and social issues. In the end, clinical implementation of pharmacogenetics in the cardiovascular field or any other therapeutic arena on a population-wide scale across cultures, nations and ethnicities may very well be limited by the social acceptance and public perception of genetic testing.

Executive summary.

Background

  • Significant advances in cardiovascular pharmacogenomic research with discovery of association between genotype and drug response phenotype.

  • Challenges include the required threshold of evidence for a particular variant prior to clinical translation, increased value above and beyond current testing practices, genotype predictability in drug efficacy, response, adverse effects, objective practice guidelines, point-of-care access to patient genomic information, compensation and reimbursement, and ethical, legal, privacy and security issues.

Cardiovascular & genomic variants

  • Clopidogrel and CYP2C19:

    • Association of loss-of-function *2 allele carriers with a reduced ability to metabolize clopidogrel to form the active metabolite, a lower antiplatelet effect, and a higher risk for cardiovascular events.

    • Association of the gain-of-function *17 variant with increased drug sensitivity and adverse bleeding side effects.

    • Strong correlation of ancestry to the heritability of the alleles.

    • Some evidence of loss-of-function *3 variant contributing to poorer response in east Asians.

    • Despite the robust association between the CYP2C19*2 genotype and lower clopidogrel response, large-scale studies with coronary artery disease subjects with lower risks for thromboembolic events failed to show the same association. Therefore, clinical utility may be limited to only patients with high risk for recurrent events.

    • Despite the boxed warning, the US FDA has not mandated genetic testing for CYP2C19 status before initiation of clopidogrel, leading to some confusion among healthcare providers regarding clinical implementation.

    • The American College of Cardiology Foundation and the American Heart Association have recently suggested that in the absence of prospective randomized trials, the evidence is insufficient to recommend genetic testing.

    • Recently, the ELEVATE-TIMI 56 (NCT01235351), a multicenter, randomized, double-blinded trial, showed that among patients with stable cardiovascular disease, tripling the maintenance dose of clopidogrel to 225 mg daily in CYP2C19*2 heterozygotes achieved levels of platelet reactivity similar to the standard 75 mg dose in noncarriers while in CYP2C19*2 homozygotes, even a 300 mg daily dose was unlikely to result in optimal degree of platelet inhibition.

  • Warfarin and CYP2C9 and VKORC1:

    • Multiple linked variants in VKORC1 have been associated with variability in gene expression, increased sensitivity to warfarin and reduction in dose requirement. However, only VKORC1 −1639G>A is a functional variant.

    • Additional identification of variants in CYP2C9*2 and *3 alleles are associated with 40–70% reduction in (S)-warfarin clearance and approximately 20–40% lower warfarin dose requirement.

    • Despite the plethora of information from multiple genome-wide association studies, the strong support from the International Warfarin Pharmacogenetics Consortium (IWPC) and Clinical Pharmacogenetics Implementation Consortium (CPIC), in addition to the FDA warning labels and the availability of several FDA-approved platforms for warfarin genotyping, the clinical implementation of genetic testing for warfarin dosing has been disappointing in practice.

    • The current consensus guidelines by the American College of Chest Physicians actually warn against the routine use of genetic data to guide dosing. Similarly, the American College of Medical Genetics endorses testing only in cases of unusual warfarin response.

    • There is a lack of evidence from large-scale prospective randomized trials, but current such studies are underway.

  • β-blockers and CYP2D6 and ADRB1:

    • In heart failure therapeutic pharmacogenomics, the most robust evidence concerns the association of ADRB1 variants (two common nonsynonymous polymorphisms Ser49Gly; rs1801252 and Arg389Gly; rs1801253) and left ventricular ejection fraction or clinical outcomes mediated by β-blockade.

  • Antihypertensives and ADRB1 and ADD1:

    • There are consistent data in several clinical studies showing association of differential blood pressure lowering levels with polymorphisms of the ADRB1 gene, namely Ser49Gly and Arg389Gly.

  • Statins and HMGCR, LDLR and SLCO1B1:

    • The H7 haplotype in the HMGCR has been associated with 11–19% decreased reduction in low-density lipoprotein cholesterol in both ethnically diverse and multiple independent populations; HMGCR H2 and LDLR L5, have also been associated with decreased attenuation of low-density lipoprotein cholesterol.

    • The FDA recommendation that the higher dose of 80 mg/day of simvastatin be restricted to patients taking the medication for 12 months or longer was based on the finding where patients homozygous or heterozygous for a variant (rs4363657) in the SLCO1B1 gene were found to have a 16.9- and 4.5-fold increase, respectively, in developing myopathy or rhabdomyolysis at the 80 mg/day higher dose. The identified polymorphism tags a known nonsynonymous variant SLCO1B1*5.

    • Other studies have shown that variants in SLCO1B1 predict adverse side effects to atorvastatin and cerivastatin-associated rhabdomyolysis.

    • A biomarker that can modulate risk may not necessarily be of high clinical utility because of low predictive value (<20% for developing myopathy in patients homozygous for the Val174Ala allele).

Future perspective

  • The greatest obstacle to clinical implementation of cardiovascular pharmacogenetics may be the lack of both reproducibility and agreement about the clinical validity and utility of the findings.

  • Population-wide implementation across cultures, nations and ethnicities may be limited by the social acceptance and public perception of genetic testing as well as ethical, legal and social issues.

Footnotes

Financial & competing interests disclosure

This work was supported by NIH grant F32 HL105036 (FS Ong). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as:

▪ of interest

▪ of considerable interest

  • 1▪.Ong FS, Grody WW, Deignan JL. Privacy and data management in the era of massively parallel next-generation sequencing. Expert Rev Mol Diagn. 2011;11(5):457–459. doi: 10.1586/erm.11.34. Discusses ethical, privacy and social issues in the era of massively parallel next-generation sequencing. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Savi P, Combalbert J, Gaich C, et al. The antiaggregating activity of clopidogrel is due to a metabolic activation by the hepatic cytochrome P450-1A. Thromb Haemost. 1994;72(2):313–317. [PubMed] [Google Scholar]
  • 3.Turpeinen M, Tolonen A, Uusitalo J, Jalonen J, Pelkonen O, Laine K. Effect of clopidogrel and ticlopidine on cytochrome P450 2B6 activity as measured by bupropion hydroxylation. Clin Pharmacol Ther. 2005;77(6):553–559. doi: 10.1016/j.clpt.2005.02.010. [DOI] [PubMed] [Google Scholar]
  • 4.Richter T, Murdter TE, Heinkele G, et al. Potent mechanism-based inhibition of human CYP2B6 by clopidogrel and ticlopidine. J Pharmacol Exp Ther. 2004;308(1):189–197. doi: 10.1124/jpet.103.056127. [DOI] [PubMed] [Google Scholar]
  • 5.O’Donoghue M, Wiviott SD. Clopidogrel response variability and future therapies: clopidogrel: does one size fit all? Circulation. 2006;114(22):e600–e606. doi: 10.1161/CIRCULATIONAHA.106.643171. [DOI] [PubMed] [Google Scholar]
  • 6.Wiviott SD. Clopidogrel response variability, resistance, or both? Am J Cardiol. 2006;98(10A):18N–24N. doi: 10.1016/j.amjcard.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 7.Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, et al. Variability in individual responsiveness to clopidogrel: clinical implications, management, and future perspectives. J Am Coll Cardiol. 2007;49(14):1505–1516. doi: 10.1016/j.jacc.2006.11.044. [DOI] [PubMed] [Google Scholar]
  • 8.Gurbel PA, Bliden KP, Hiatt BL, O’Connor CM. Clopidogrel for coronary stenting: response variability, drug resistance, and the effect of pretreatment platelet reactivity. Circulation. 2003;107(23):2908–2913. doi: 10.1161/01.CIR.0000072771.11429.83. [DOI] [PubMed] [Google Scholar]
  • 9.Gurbel PA, Lau WC, Bliden KP, Tantry US. Clopidogrel resistance: implications for coronary stenting. Curr Pharm Des. 2006;12(10):1261–1269. doi: 10.2174/138161206776361345. [DOI] [PubMed] [Google Scholar]
  • 10.Gurbel PA, Tantry US. Drug insight: clopidogrel nonresponsiveness. Nat Clin Pract Cardiovasc Med. 2006;3(7):387–395. doi: 10.1038/ncpcardio0602. [DOI] [PubMed] [Google Scholar]
  • 11.Xie HG, Zou JJ, Hu ZY, Zhang JJ, Ye F, Chen SL. Individual variability in the disposition of and response to clopidogrel: pharmacogenomics and beyond. Pharmacol Ther. 2011;129(3):267–289. doi: 10.1016/j.pharmthera.2010.10.001. [DOI] [PubMed] [Google Scholar]
  • 12▪▪.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(8):849–857. doi: 10.1001/jama.2009.1232. Association study that demonstrated CYP2C19*2 genotype was associated with diminished platelet response to clopidogrel treatment and poorer cardiovascular outcomes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sofi F, Giusti B, Marcucci R, Gori AM, Abbate R, Gensini GF. Cytochrome P450 2C19*2 polymorphism and cardiovascular recurrences in patients taking clopidogrel: a meta-analysis. Pharmacogenomics J. 2011;11(3):199–206. doi: 10.1038/tpj.2010.21. [DOI] [PubMed] [Google Scholar]
  • 14.Kim KA, Park PW, Hong SJ, Park JY. The effect of CYP2C19 polymorphism on the pharmacokinetics and pharmacodynamics of clopidogrel: a possible mechanism for clopidogrel resistance. Clin Pharmacol Ther. 2008;84(2):236–242. doi: 10.1038/clpt.2008.20. [DOI] [PubMed] [Google Scholar]
  • 15▪.Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 polymorphisms and response to clopidogrel. N Engl J Med. 2009;360(4):354–362. doi: 10.1056/NEJMoa0809171. Association study that showed carriers of the reduced-function CYP2C19 allele had significantly lower levels of the active metabolite of clopidogrel, diminished platelet inhibition and a higher rate of major adverse cardiovascular events with clopidogrel therapy. [DOI] [PubMed] [Google Scholar]
  • 16.Hulot JS, Bura A, Villard E, et al. Cytochrome P450 2C19 loss-of-function polymorphism is a major determinant of clopidogrel responsiveness in healthy subjects. Blood. 2006;108(7):2244–2247. doi: 10.1182/blood-2006-04-013052. [DOI] [PubMed] [Google Scholar]
  • 17.Umemura K, Furuta T, Kondo K. The common gene variants of CYP2C19 affect pharmacokinetics and pharmacodynamics in an active metabolite of clopidogrel in healthy subjects. J Thromb Haemost. 2008;6(8):1439–1441. doi: 10.1111/j.1538-7836.2008.03050.x. [DOI] [PubMed] [Google Scholar]
  • 18.Brandt JT, Close SL, Iturria SJ, et al. Common polymorphisms of CYP2C19 and CYP2C9 affect the pharmacokinetic and pharmacodynamic response to clopidogrel but not prasugrel. J Thromb Haemost. 2007;5(12):2429–2436. doi: 10.1111/j.1538-7836.2007.02775.x. [DOI] [PubMed] [Google Scholar]
  • 19.Frere C, Cuisset T, Morange PE, et al. Effect of cytochrome p450 polymorphisms on platelet reactivity after treatment with clopidogrel in acute coronary syndrome. Am J Cardiol. 2008;101(8):1088–1093. doi: 10.1016/j.amjcard.2007.11.065. [DOI] [PubMed] [Google Scholar]
  • 20.Geisler T, Schaeffeler E, Dippon J, et al. CYP2C19 and nongenetic factors predict poor responsiveness to clopidogrel loading dose after coronary stent implantation. Pharmacogenomics. 2008;9(9):1251–1259. doi: 10.2217/14622416.9.9.1251. [DOI] [PubMed] [Google Scholar]
  • 21.Giusti B, Gori AM, Marcucci R, et al. Cytochrome P450 2C19 loss-of-function polymorphism, but not CYP3A4 IVS10 + 12G/A and P2Y12 T744C polymorphisms, is associated with response variability to dual antiplatelet treatment in high-risk vascular patients. Pharmacogenet Genomics. 2007;17(12):1057–1064. doi: 10.1097/FPC.0b013e3282f1b2be. [DOI] [PubMed] [Google Scholar]
  • 22.Malek LA, Kisiel B, Spiewak M, et al. Coexisting polymorphisms of P2Y12 and CYP2C19 genes as a risk factor for persistent platelet activation with clopidogrel. Circ J. 2008;72(7):1165–1169. doi: 10.1253/circj.72.1165. [DOI] [PubMed] [Google Scholar]
  • 23.Collet JP, Hulot JS, Pena A, et al. Cytochrome P450 2C19 polymorphism in young patients treated with clopidogrel after myocardial infarction: a cohort study. Lancet. 2009;373(9660):309–317. doi: 10.1016/S0140-6736(08)61845-0. [DOI] [PubMed] [Google Scholar]
  • 24.Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med. 2009;360(4):363–375. doi: 10.1056/NEJMoa0808227. [DOI] [PubMed] [Google Scholar]
  • 25.Trenk D, Hochholzer W, Fromm MF, et al. Cytochrome P450 2C19 681G>A polymorphism and high on-clopidogrel platelet reactivity associated with adverse 1-year clinical outcome of elective percutaneous coronary intervention with drug-eluting or bare-metal stents. J Am Coll Cardiol. 2008;51(20):1925–1934. doi: 10.1016/j.jacc.2007.12.056. [DOI] [PubMed] [Google Scholar]
  • 26.Mega JL, Simon T, Collet JP, 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(16):1821–1830. doi: 10.1001/jama.2010.1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hulot JS, Collet JP, 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(2):134–143. doi: 10.1016/j.jacc.2009.12.071. [DOI] [PubMed] [Google Scholar]
  • 28.Gurbel PA, Bliden KP, Butler K, et al. Response to ticagrelor in clopidogrel nonresponders and responders and effect of switching therapies: the RESPOND study. Circulation. 2010;121(10):1188–1199. doi: 10.1161/CIRCULATIONAHA.109.919456. [DOI] [PubMed] [Google Scholar]
  • 29.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. Circulation. 2010;121(4):512–518. doi: 10.1161/CIRCULATIONAHA.109.885194. [DOI] [PubMed] [Google Scholar]
  • 30.Pare G, Mehta SR, Yusuf S, et al. Effects of CYP2C19 genotype on outcomes of clopidogrel treatment. N Engl J Med. 2010;363(18):1704–1714. doi: 10.1056/NEJMoa1008410. [DOI] [PubMed] [Google Scholar]
  • 31.Fuster V, Sweeny JM. Clopidogrel and the reduced-function CYP2C19 genetic variant: a limited piece of the overall therapeutic puzzle. JAMA. 2010;304(16):1839–1840. doi: 10.1001/jama.2010.1566. [DOI] [PubMed] [Google Scholar]
  • 32.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 endorsed by the Society for Cardiovascular Angiography and Interventions and the Society of Thoracic Surgeons. J Am Coll Cardiol. 2010;56(4):321–341. doi: 10.1016/j.jacc.2010.05.013. [DOI] [PubMed] [Google Scholar]
  • 33.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. Circulation. 2010;122(5):537–557. doi: 10.1161/CIR.0b013e3181ee08ed. [DOI] [PubMed] [Google Scholar]
  • 34▪▪.Mega JL, Hochholzer W, Frelinger AL, 3rd, et al. Dosing clopidogrel based on CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovascular disease. JAMA. 2011;306(20):2221–2228. doi: 10.1001/jama.2011.1703. A recent clinical trial showing that tripling the maintenance dose of clopidogrel to 225 mg daily in CYP2C19*2 heterozygotes achieved levels of platelet reactivity similar to the standard 75 mg dose in noncarriers while in CYP2C19*2 homozygotes, even a 300 mg daily dose was unlikely to result in optimal degree of platelet inhibition. [DOI] [PubMed] [Google Scholar]
  • 35.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(2):328–332. doi: 10.1038/clpt.2011.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361(12):1139–1151. doi: 10.1056/NEJMoa0905561. [DOI] [PubMed] [Google Scholar]
  • 37.Tomek A, Matoska V, Eisert C, Serebruany VL. Optimization of anticoagulation with warfarin for stroke prevention: pharmacogenetic considerations. Am J Ther. 2011;18(3):e55–e66. doi: 10.1097/MJT.0b013e3181cea0cd. [DOI] [PubMed] [Google Scholar]
  • 38.Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G. Pharmacology and management of the vitamin K antagonists: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition) Chest. 2008;133(Suppl 6):160S–198S. doi: 10.1378/chest.08-0670. [DOI] [PubMed] [Google Scholar]
  • 39.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;112(4):1022–1027. doi: 10.1182/blood-2008-01-134247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Klein TE, Altman RB, Eriksson N, et al. Estimation of the warfarin dose with clinical and pharmacogenetic data. N Engl J Med. 2009;360(8):753–764. doi: 10.1056/NEJMoa0809329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Veenstra DL, You JH, Rieder MJ, et al. Association of vitamin K epoxide reductase complex 1 (VKORC1) variants with warfarin dose in a Hong Kong Chinese patient population. Pharmacogenet Genomics. 2005;15(10):687–691. doi: 10.1097/01.fpc.0000174789.77614.68. [DOI] [PubMed] [Google Scholar]
  • 42▪.Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med. 2005;352(22):2285–2293. doi: 10.1056/NEJMoa044503. Showed that VKORC1 haplotypes can be used to stratify patients into low-, intermediate- and high-dose warfarin groups and may explain differences in dose requirements among patients of different ancestries. [DOI] [PubMed] [Google Scholar]
  • 43.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(3):e1000433. doi: 10.1371/journal.pgen.1000433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang D, Chen H, Momary KM, Cavallari LH, Johnson JA, Sadee W. Regulatory polymorphism in vitamin K epoxide reductase complex subunit 1 (VKORC1) affects gene expression and warfarin dose requirement. Blood. 2008;112(4):1013–1021. doi: 10.1182/blood-2008-03-144899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.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(25):2804–2812. doi: 10.1016/j.jacc.2010.03.009. [DOI] [PubMed] [Google Scholar]
  • 46.Sanderson S, Emery J, Higgins J. CYP2C9 gene variants, drug dose, and bleeding risk in warfarin-treated patients: a HuGEnet systematic review and meta-analysis. Genet Med. 2005;7(2):97–104. doi: 10.1097/01.gim.0000153664.65759.cf. [DOI] [PubMed] [Google Scholar]
  • 47.Limdi NA, Arnett DK, Goldstein JA, et al. Influence of CYP2C9 and VKORC1 on warfarin dose, anticoagulation attainment and maintenance among European–Americans and African–Americans. Pharmacogenomics. 2008;9(5):511–526. doi: 10.2217/14622416.9.5.511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.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 patients on warfarin. Clin Pharmacol Ther. 2008;83(2):312–321. doi: 10.1038/sj.clpt.6100290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Goldstein JA, Blaisdell JA, Limdi NA. A potentially deleterious new CYP2C9 polymorphism identified in an African–American patient with major hemorrhage on warfarin therapy. Blood Cells Mol Dis. 2009;42(2):155–158. doi: 10.1016/j.bcmd.2008.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Limdi NA, Wadelius M, Cavallari L, et al. Warfarin pharmacogenetics: a single VKORC1 polymorphism is predictive of dose across 3 racial groups. Blood. 2010;115(18):3827–3834. doi: 10.1182/blood-2009-12-255992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gage BF, Eby C, Johnson JA, et al. Use of pharmacogenetic and clinical factors to predict the therapeutic dose of warfarin. Clin Pharmacol Ther. 2008;84(3):326–331. doi: 10.1038/clpt.2008.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Perera MA, Gamazon E, Cavallari LH, et al. The missing association: sequencing-based discovery of novel SNPs in VKORC1 and CYP2C9 that affect warfarin dose in African–Americans. Clin Pharmacol Ther. 2011;89(3):408–415. doi: 10.1038/clpt.2010.322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Aithal GP, Day CP, Kesteven PJ, Daly AK. Association of polymorphisms in the cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding complications. Lancet. 1999;353(9154):717–719. doi: 10.1016/S0140-6736(98)04474-2. [DOI] [PubMed] [Google Scholar]
  • 54.Wadelius M, Chen LY, Eriksson N, et al. Association of warfarin dose with genes involved in its action and metabolism. Hum Genet. 2007;121(1):23–34. doi: 10.1007/s00439-006-0260-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cha PC, Mushiroda T, Takahashi A, et al. Genome-wide association study identifies genetic determinants of warfarin responsiveness for Japanese. Hum Mol Genet. 2010;19(23):4735–4744. doi: 10.1093/hmg/ddq389. [DOI] [PubMed] [Google Scholar]
  • 56.Flockhart DA, O’Kane D, Williams MS, et al. Pharmacogenetic testing of CYP2C9 and VKORC1 alleles for warfarin. Genet Med. 2008;10(2):139–150. doi: 10.1097/GIM.0b013e318163c35f. [DOI] [PubMed] [Google Scholar]
  • 57.Rosove MH, Grody WW. Should we be applying warfarin pharmacogenetics to clinical practice? No, not now. Ann Intern Med. 2009;151(4):270–273. W295. doi: 10.7326/0003-4819-151-4-200908180-00009. [DOI] [PubMed] [Google Scholar]
  • 58.Anderson JL, Horne BD, Stevens SM, et al. Randomized trial of genotype-guided versus standard warfarin dosing in patients initiating oral anticoagulation. Circulation. 2007;116(22):2563–2570. doi: 10.1161/CIRCULATIONAHA.107.737312. [DOI] [PubMed] [Google Scholar]
  • 59.Johnson JA, Gong L, Whirl-Carrillo M, et al. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin Pharmacol Ther. 2011;90(4):625–629. doi: 10.1038/clpt.2011.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lenzini P, Wadelius M, Kimmel S, et al. Integration of genetic, clinical, and INR data to refine warfarin dosing. Clin Pharmacol Ther. 2010;87(5):572–578. doi: 10.1038/clpt.2010.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Liggett SB, Mialet-Perez J, Thaneemit-Chen S, et al. A polymorphism within a conserved beta(1)-adrenergic receptor motif alters cardiac function and beta-blocker response in human heart failure. Proc Natl Acad Sci USA. 2006;103(30):11288–11293. doi: 10.1073/pnas.0509937103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Davis HM, Johnson JA. Heart failure pharmacogenetics: past, present, and future. Curr Cardiol Rep. 2011;13(3):175–184. doi: 10.1007/s11886-011-0181-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Johnson JA, Liggett SB. Cardiovascular pharmacogenomics of adrenergic receptor signaling: clinical implications and future directions. Clin Pharmacol Ther. 2011;89(3):366–378. doi: 10.1038/clpt.2010.315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.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(5):432–444. doi: 10.1016/j.jacc.2009.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.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(8):644–651. doi: 10.1016/j.jacc.2008.05.022. [DOI] [PubMed] [Google Scholar]
  • 66.Bristow MR, Murphy GA, Krause-Steinrauf H, et al. An alpha2C-adrenergic receptor polymorphism alters the norepinephrine-lowering effects and therapeutic response of the beta-blocker bucindolol in chronic heart failure. Circ Heart Fail. 2010;3(1):21–28. doi: 10.1161/CIRCHEARTFAILURE.109.885962. [DOI] [PubMed] [Google Scholar]
  • 67.Lobmeyer MT, Gong Y, Terra SG, et al. Synergistic polymorphisms of beta1 and alpha2C-adrenergic receptors and the influence on left ventricular ejection fraction response to beta-blocker therapy in heart failure. Pharmacogenet Genomics. 2007;17(4):277–282. doi: 10.1097/FPC.0b013e3280105245. [DOI] [PubMed] [Google Scholar]
  • 68.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(5):510–517. doi: 10.1038/nm1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.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(1):44–52. doi: 10.1016/S0009-9236(03)00068-7. [DOI] [PubMed] [Google Scholar]
  • 70.Liu J, Liu ZQ, Tan ZR, et al. Gly389Arg polymorphism of beta1-adrenergic receptor is associated with the cardiovascular response to metoprolol. Clin Pharmacol Ther. 2003;74(4):372–379. doi: 10.1016/S0009-9236(03)00224-8. [DOI] [PubMed] [Google Scholar]
  • 71.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(6):715–721. doi: 10.1038/clpt.2008.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Roden DM, Johnson JA, Kimmel SE, et al. Cardiovascular pharmacogenomics. Circ Res. 2011;109(7):807–820. doi: 10.1161/CIRCRESAHA.110.230995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Terra SG, Pauly DF, Lee CR, et al. beta-adrenergic receptor polymorphisms and responses during titration of metoprolol controlled release/extended release in heart failure. Clin Pharmacol Ther. 2005;77(3):127–137. doi: 10.1016/j.clpt.2004.10.006. [DOI] [PubMed] [Google Scholar]
  • 74.Johnson JA. Pharmacogenomics of antihypertensive drugs: past, present and future. Pharmacogenomics. 2010;11(4):487–491. doi: 10.2217/pgs.10.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Manunta P, Maillard M, Tantardini C, et al. Relationships among endogenous ouabain, alpha-adducin polymorphisms and renal sodium handling in primary hypertension. J Hypertens. 2008;26(5):914–920. doi: 10.1097/HJH.0b013e3282f5315f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sciarrone MT, Stella P, Barlassina C, et al. ACE and alpha-adducin polymorphism as markers of individual response to diuretic therapy. Hypertension. 2003;41(3):398–403. doi: 10.1161/01.HYP.0000057010.27011.2C. [DOI] [PubMed] [Google Scholar]
  • 77▪.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(13):1680–1689. doi: 10.1001/jama.287.13.1680. Showed that in carriers of the adducin variant, diuretic therapy was associated with a lower risk of combined myocardial infarction or stroke than other antihypertensive therapies. Suggests that this large subgroup of the hypertensive population may be especially likely to benefit from low-dose diuretic therapy. [DOI] [PubMed] [Google Scholar]
  • 78.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(2):112–122. doi: 10.1038/sj.tpj.6500395. [DOI] [PubMed] [Google Scholar]
  • 79.Gerhard T, Gong Y, Beitelshees AL, et al. Alpha-adducin polymorphism associated with increased risk of adverse cardiovascular outcomes: results from GENEtic Substudy of the INternational VErapamil SR-trandolapril STudy (INVEST-GENES) Am Heart J. 2008;156(2):397–404. doi: 10.1016/j.ahj.2008.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lanzani C, Citterio L, Glorioso N, et al. Adducin- and ouabain-related gene variants predict the antihypertensive activity of rostafuroxin, part 2: clinical studies. Sci Transl Med. 2010;2(59):59ra87. doi: 10.1126/scitranslmed.3001814. [DOI] [PubMed] [Google Scholar]
  • 81.Citterio L, Lanzani C, Manunta P, Bianchi G. Genetics of primary hypertension: the clinical impact of adducin polymorphisms. Biochim Biophys Acta. 2010;1802(12):1285–1298. doi: 10.1016/j.bbadis.2010.03.014. [DOI] [PubMed] [Google Scholar]
  • 82.Goldstein JL, Brown MS. The LDL receptor. Arterioscler Thromb Vasc Biol. 2009;29(4):431–438. doi: 10.1161/ATVBAHA.108.179564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83▪.Chasman DI, Posada D, Subrahmanyan L, Cook NR, Stanton VP, Jr, Ridker PM. Pharmacogenetic study of statin therapy and cholesterol reduction. JAMA. 2004;291(23):2821–2827. doi: 10.1001/jama.291.23.2821. Showed that individuals heterozygous for a genetic variant in the HMG-CoA reductase gene may experience significantly smaller reductions in cholesterol when treated with pravastatin. [DOI] [PubMed] [Google Scholar]
  • 84.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(4):355–362. doi: 10.1161/CIRCULATIONAHA.108.773267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Donnelly LA, Doney AS, Dannfald J, et al. A paucimorphic variant in the HMG-CoA reductase gene is associated with lipid-lowering response to statin treatment in diabetes: a GoDARTS study. Pharmacogenet Genomics. 2008;18(12):1021–1026. doi: 10.1097/FPC.0b013e3283106071. [DOI] [PubMed] [Google Scholar]
  • 86.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(12):1537–1544. doi: 10.1161/CIRCULATIONAHA.107.708388. [DOI] [PubMed] [Google Scholar]
  • 87.Poduri A, Khullar M, Bahl A, Sehrawat BS, Sharma Y, Talwar KK. Common variants of HMGCR, CETP, APOAI, ABCB1, CYP3A4, and CYP7A1 genes as predictors of lipid-lowering response to atorvastatin therapy. DNA Cell Biol. 2010;29(10):629–637. doi: 10.1089/dna.2009.1008. [DOI] [PubMed] [Google Scholar]
  • 88.Mangravite LM, Medina MW, Cui J, et al. Combined influence of LDLR and HMGCR sequence variation on lipid-lowering response to simvastatin. Arterioscler Thromb Vasc Biol. 2010;30(7):1485–1492. doi: 10.1161/ATVBAHA.110.203273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kajinami K, Brousseau ME, Ordovas JM, Schaefer EJ. CYP3A4 genotypes and plasma lipoprotein levels before and after treatment with atorvastatin in primary hypercholesterolemia. Am J Cardiol. 2004;93(1):104–107. doi: 10.1016/j.amjcard.2003.08.078. [DOI] [PubMed] [Google Scholar]
  • 90.Wang A, Yu BN, Luo CH, et al. Ile118Val genetic polymorphism of CYP3A4 and its effects on lipid-lowering efficacy of simvastatin in Chinese hyperlipidemic patients. Eur J Clin Pharmacol. 2005;60(12):843–848. doi: 10.1007/s00228-004-0848-7. [DOI] [PubMed] [Google Scholar]
  • 91.Link E, Parish S, Armitage J, et al. SLCO1B1 variants and statin-induced myopathy – a genomewide study. N Engl J Med. 2008;359(8):789–799. doi: 10.1056/NEJMoa0801936. [DOI] [PubMed] [Google Scholar]
  • 92.Pasanen MK, Neuvonen M, Neuvonen PJ, Niemi M. SLCO1B1 polymorphism markedly affects the pharmacokinetics of simvastatin acid. Pharmacogenet Genomics. 2006;16(12):873–879. doi: 10.1097/01.fpc.0000230416.82349.90. [DOI] [PubMed] [Google Scholar]
  • 93.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(17):1609–1616. doi: 10.1016/j.jacc.2009.04.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Donnelly LA, Doney AS, Tavendale R, et al. Common nonsynonymous substitutions in SLCO1B1 predispose to statin intolerance in routinely treated individuals with Type 2 diabetes: a go-DARTS study. Clin Pharmacol Ther. 2011;89(2):210–216. doi: 10.1038/clpt.2010.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Marciante KD, Durda JP, Heckbert SR, et al. Cerivastatin, genetic variants, and the risk of rhabdomyolysis. Pharmacogenet Genomics. 2011;21(5):280–288. doi: 10.1097/FPC.0b013e328343dd7d. [DOI] [PMC free article] [PubMed] [Google Scholar]

Websites

RESOURCES