Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 30.
Published in final edited form as: Curr Opin Cardiol. 2012 May;27(3):10.1097/HCO.0b013e32835220e3. doi: 10.1097/HCO.0b013e32835220e3

Genetic determinants of response to cardiovascular drugs

Quinn S Wells 1, Jessica T Delaney 1, Dan M Roden 1
PMCID: PMC3874723  NIHMSID: NIHMS524622  PMID: 22382501

Abstract

Purpose of review

To survey genetic variation contributing to variable responsiveness and toxicity to important cardiovascular drugs and highlight recent developments in the field of cardiovascular pharmacogenomics and personalized medicine.

Recent findings

Previously recognized pharmacogenomic associations with drug efficacy have been further validated (e.g. with clopidogrel and warfarin) and shown to influence clinically important outcomes. The clinical significance of variants modulating toxicity (e.g. SLCO1B1 with simvastatin) has also been confirmed. The genetic contribution to variable efficacy and toxicity of other important classes of cardiovascular drugs, such as beta-blockers, is becoming increasingly recognized. Prospective trials testing whether the use of genomic information improves clinical care are underway. Guidance based on the most well-established pharmacogenomic findings has appeared in prescribing labeling and is in the early stages of being implemented into routine clinical care.

Summary

Clinically validated gene variants that modulate responsiveness to cardiovascular drugs continue to be discovered and validated. Early steps are underway to translate these discoveries into clinical care.

Keywords: personalized medicine, pharmacogenomics, variable drug response

Introduction

Pharmacogenomics seeks to determine the contribution of genetic variants to drug efficacy and toxicity. Cardiac pharmacogenomics is a rapidly growing field, offering the potential for improved treatment outcomes and prevention of adverse drug events. Variants common in the population have been shown to modify drug metabolism, drug transport, and drug targets, and could be utilized to predict an individual's treatment response. We discuss recent findings in cardiac pharmacogenetics and their translation into clinical practice for those variants that have been comprehensively studied. Examples of genetic variants influencing response to cardiovascular drugs and the strength of evidence supporting their translation into clinical practice are listed in Table 1.

Table 1. Examples of genetic variants influencing response to cardiovascular drugs.

Gene SNP Chr Position Reported effect on function Possible implication for clinical practice
Tier 1 – Strong level of evidence for clinical translation
CYP2C19*2 rs4244285 10 96531606 Reduced enzyme function→ Resistance to clopidogrel For carriers of 1 or 2 copies of *2 allele, consider alternate antiplatelet agent when undergoing PCI or after ACS. Consider higher maintenance dose in heterozygotes
VKORC1 rs9923231 16 31107689 Increased sensitivity to warfarin For carriers of 1 or 2 copies of the A allele, consider a lower initial dose of warfarin compared with carriers of the G allele (use in conjunction with CYP2C9 genotype information)
CYP2C9*2 rs1799853 10 96702047 Increased sensitivity to warfarin For carriers of 1 or 2 copies of the *2 allele, consider a lower initial dose of warfarin (use in conjunction with CYP2C9 genotype information)
CYP2C9*3 rs1057910 10 96741053 Increased sensitivity to warfarin For carriers of 1 or 2 copies of the *3 allele, consider a lower initial dose of warfarin (use in conjunction with VKORC1 genotype information)
SLCO1B1 rs4149056 12 21331549 Increased likelihood of simvastatin-related myotoxicity For carriers of this variant consider an alternate lipid lowering therapy
Tier 2 – Weaker evidence, clinical implementation premature
ABCB1-C3435T rs1045642 7 86976581 Resistance to clopidogrel If genotype known, may consider alternate antiplatelet agent
CYP2C19*17 rs12248560 10 96511647 Increased clopidogrel efficacy Concern for bleeding with clopidogrel, monitor patients closely if genotype known
CYP2C19*3 rs4986893 10 96540410 Resistance to clopidogrel If genotype known, may consider alternate antiplatelet agent
ADRB1-R389G rs1801253 10 115805056 Increased response to beta-blockers May consider alternate therapeutic class (e.g. maximize ACEi over beta-blocker in heart failure or calcium channel antagonist for rate control in atrial fibrillation)
ADRB1-R389G rs1801253 10 115805056 Associated with successful rate control strategy for AF May assist in selection of atrial fibrillation treatment strategy (i.e. rate versus rhythm control)
4q25 (intergenic near PITX2) rs10033464 4 136232206 Associated with successful rhythm control strategy for AF May assist in selection of atrial fibrillation treatment strategy (i.e. rate versus rhythm control)
KCNE1-D85N rs1805128 21 35821680 Increased risk of drug-induced TdP Assist in assessment of risk for drug-related TdP

ACEi, angiotensin converting enzyme inhibitor; AF, atrial fibrillation; SNP, single-nucleotide polymorphism; TdP, torsades de pointes

There are several types of common genetic variation relevant to pharmacogenomics. The most common form, termed a single-nucleotide polymorphism (SNP), is the change of a single nucleotide within the genome. SNPs may occur in regions that do not encode proteins (noncoding variants). However, SNPs also occur in protein-coding regions of the genome (coding variants), in which they do not necessarily cause an alteration in protein (synonymous variant), but may also lead to the incorporation of a different amino acid (nonsynonymous variant). Additionally, a SNP may introduce a premature stop codon into the DNA sequence and thereby cause production of a truncated protein product (nonsense variant). Other common types of variation include the addition or removal of nucleotides, termed insertions and deletions, respectively. Similarly, a genomic region may be copied multiple times, referred to as duplication.

In general, identification of genomic variation can proceed by focused studies of genes or genomic regions suspected of being related to the disease or phenotype of interest or through unbiased, genome-wide approaches that seek association between genomic variation and selected traits or conditions. The candidate gene approach builds on known or predicted biologic functions, but findings have frequently been difficult to validate [1]. Genome-wide association studies (GWAS) are not limited by biological models and have the potential to discover associations outside current understanding. The GWAS paradigm frequently results in identification of common variants of relatively small effect size; however, a recurrent finding is that pharmacogenetic effect sizes are often significantly higher than those seen in complex diseases [2].

It is interesting to note that trials demonstrating differential treatment outcomes by genotype have generally included a placebo arm, as is seen in the case of β-blockers [3]. One interpretation is that carriers of low-risk alleles receive less benefit from active therapy and treatment of all patients has the effects of making the treated high-risk genotypes more similar to the low-risk genotype (regardless of treatment) and therefore ‘equalizing’ the outcomes. Future pharmacogenomics studies will need to take this observation into account.

Clopidogrel

The thienopyridine antiplatelet agent, clopidogrel, primarily used for the prevention of adverse cardiac events including stent thrombosis, has become one of the most widely prescribed medications [4]. A two-step oxidative process catalyzed by enzymes in the cytochrome P450 system converts clopidogrel to the active thiol metabolite [5]. Response to clopidogrel is variable and this response is in part dependent on an individual's genotype for CYP2C19, a cytochrome P450 enzyme central to clopidogrel bioactivation. Carriers of CYP2C19*2 and CYP2C19*3 have reduced CYP2C19 enzyme activity and are called poor metabolizers, whereas carriers of CYP2C19*17 have increased CYP2C19 enzyme activity and are called ultra-rapid metabolizers. Recent findings have found the CYP2C19*2 polymorphism, which is present in approximately 15% of Caucasians and Africans and approximately 30% of Asians, to be associated with reduced clopidogrel efficacy [512]. This has prompted a Food and Drug Administration (FDA) black box warning for cautious use among carriers of the reduced function alleles [13,14].

Among patients treated with clopidogrel for acute coronary syndrome (ACS) and/or percutaneous coronary intervention (PCI), studies have consistently found carriers of at least one loss-of-function allele to have a significantly increased risk of adverse cardiac events, particularly stent thrombosis [11, 12, 1518, 19•21•]. The few studies that did not find an associated risk among carriers of reduced function allele had lower rates of stent placement, suggesting the greatest risk for adverse events occurs among those undergoing stent placement [22,23]. A meta-analysis including nine studies and 9685 patients reported a hazard ratio of 1.57 [95% confidence interval (CI) 1.13–2.16, P = 0.006] for the composite endpoint of cardiovascular death, myocardial infarction, and ischemic stroke among carsriers of either one or two copies of the reduced function allele [19•]. Additionally, a gene dose effect was observed, with carriers of two reduced function alleles having an even greater risk (hazard ratio 1.76, 95% CI 1.24–2.50, P = 0.002) and the risk of stent thrombosis was further increased in carriers of the reduced function allele (hazard ratio 2.81, 95% CI 1.81–4.37). Apart from CYP2C19*2, the polymorphisms *3 and *17 have had limited study, with *3 being associated with reduced clopidogrel response and *17 being associated with increased bleeding and possibly improved efficacy while on clopidogrel therapy [9,17,24,25]. Notably, *3 is almost exclusively found in people of Asian descent.

Genome-wide association studies of platelet response to clopidogrel exposure has shown that approximately 12% variability in the trait is attributable to CYP2C19*2 carrier status [11]. The common C3435T polymorphism in ABCB1, encoding p-glycoprotein, a drug efflux transporter, has also been associated with reduced clopidogrel efficacy and increased adverse cardiac events while on clopidogrel therapy, but this has not been a consistent finding [22,2527]. Other candidate genes for clopidogrel efficacy have included CYP1A2, CYP3A4, CYP3A5, CYP2B6, PON1, and P2YR12, all with inconclusive or negative results [2730, 31•, 3239].

The Clinical Pharmacogenetics Implementation Consortium has presented a treatment algorithm for antiplatelet agents incorporating genetic information [40••]. For those undergoing PCI or with an ACS, these guidelines recommend placing those who are carriers of one or two copies of the variant allele to be treated with an alternative antiplatelet agent, including prasugrel or the recently approved agent ticagrelor [41, 42, 43•, 4446]. Neither agent requires extensive bioactivation by the CYP2C19 enzyme and therefore both can be given to CYP2C19-poor metabolizers without reduced efficacy. It is unclear whether increasing the dose of clopidogrel can over-ride resistance, with a recent study finding that heterozygotes can have an adequate platelet response with clopidogrel 225–300 mg daily measured in vitro; however, whether this strategy will reduce clinical events is unknown [4749]. Ongoing trials to test these treatment options include the Genotyping Infarct patients to Adjust and Normalize Thienopyridine treatment study with physicianguided therapy after CYP2C19 genotyping among patients post-PCI for ST-segment elevation myocardial infarction (STEMI), and the RAPID STEMI study evaluating treatment with prasugrel or high-dose clopidogrel for carriers of CYP2C19*2 and ABCB1 3435 TT.

Beta-Blockers

Beta-adrenergic receptor antagonists (β-blockers) are an important class of cardiovascular drugs used for a range of conditions including cardiac arrhythmias, ACS, stable angina, hypertension, and heart failure. Beta-blockers antagonize endogenous catecholamines at beta-adrenergic receptors, of which two subtypes, β1 and β2, are most important for cardiovascular pharmacology [50]. Important autoregulatory mechanisms include G-protein-coupled receptor kinases (GRKs), enzymes that moderate signaling through phosphorylation of activated b-receptors, and presynaptic α2C-adrenergic receptors (ADRA2C), which regulate norepinephrine release via a negative feedback pathway [51].

Two commonly used β-blockers, metoprolol and carvedilol, are metabolized through the CYP2D6 metabolic pathway and carry FDA labeling that polymorphisms in CYP2D6 alter plasma drug concentrations [52]. CYP2D6 variants with a range of enzymatic activity have been described, and the expected pharmacokinetic effects have been demonstrated repeatedly. For example, CYP2D6 genotype has been shown to correlate with a several-fold difference in metoprolol plasma concentrations during both short-term and long-term therapy [53,54]. Clinical implications of CYP2D6 have been shown for a wide range of noncardiovascular classes of drugs [5557]. However, in regard to cardiovascular agents, there are few studies examining differences in response, adverse events, or survival. Some studies have suggested differences by CYP2D6 genotype; however, the data have not been consistent [3,5861].

There are two common nonsynonymous SNPs in the β1 receptor gene, ADRB1, resulting in Ser49Gly and Arg389Gly [62,63]. The Arg389 form, more common in those of European descent, demonstrates enhanced G-protein-coupled signaling [64], and provides the theoretical rationale for increased β-blocker responsiveness in individuals possessing this polymorphism. Indeed, among individuals homozygous for this variant (∼50% of Caucasians), there has been greater b-blocker-related improvements in ventricular function have been seen in patients with heart failure [6567], though this observation has not been universal [66,68]. Clinical outcome studies in patients with heart failure have also been mixed [3,6973].

Polymorphisms in the β2 adrenergic receptor (ADRB2) have also been shown to have functional significance. Two nonsynonymous SNPs, Arg16Gly and Gln27Glu, appear to associate with augmented agonist-mediated down-regulation [74]. However, studies investigating variants in ADRB2 have generally showed no association with improvements in left ventricular ejection fraction in response to β-blockers [67,68].

Functional polymorphisms in the α2C-adrenergic receptor, ADRA2C, can theoretically modulate response to β-blockers. The common deletion polymorphism, ADRA2C Del322–325, resulting in the loss of four amino acids, shows decreased activity in transfected cells, suggesting the potential for enhanced norepinephrine release, and therefore β-blocker responsiveness [75]. Further, patients with heart failure who carry the ADRA2C Del322–325 genotype have been found to have worse disease-related outcomes [76]. One study indicates that individuals carrying the ADRA2C Del322–325 genotype had a greater improvement in left-ventricular function when given metoprolol. On the contrary, no effect was seen with bucindolol in the Beta-Blocker Evaluation of Survival Trial [77,78].

A gain-of-function Gln41Leu variant has been described in GRK5. This polymorphism is seen much more frequently in those of African descent and was found to protect against catecholamine-induced cardiomyopathy in a transgenic mouse model [51,79]. African Americans with the GRK5 Gln41Gln genotype receiving β-blocker therapy have shown a significant benefit and, interestingly, have a survival similar to untreated Leu41 carriers; suggesting that Leu carriers may not benefit from β-blocker therapy [79]. Enrichment of the US population, relative to European populations, with genotypes unfavorable to β-blocker responsiveness has been proposed as a cause for apparent differences in outcome for US patients in heart failure trials [80•].

Warfarin

Vitamin K antagonists (warfarin in the US) are anticoagulants commonly used for the treatment and prevention of arterial and venous thromboembolism. Although effective, use is complicated by wide inter-individual variation in drug response and a narrow therapeutic index. Response to warfarin is affected by several patient factors, such as age, weight, and diet; as well, a large portion of the variation is related to known common variants in pharmacokinetic and pharmacodynamic genes, CYP2C9 and VKORC1, respectively.

CYP2C9 metabolizes S-warfarin, the more active enantiomer of warfarin, to an inactive form. Two loss-of-function alleles, CYP2C9*2 and CYP2C9*3, have been associated with decreased warfarin dose requirements and increased risk of hemorrhagic complications [8184]. These findings have been confirmed in a large meta-analysis in which, for example, maintenance doses in the *2 and *3 homozygotes were reduced by 36 and 78.1%, respectively [85]. There is a considerable impact of race, with these variants being significantly less common in those of African descent, and having a correspondingly smaller effect at a population level [86].

Vitamin K epoxide reductase, coded by VKORC1, is critical to normal vitamin K metabolism (and hence synthesis of vitamin K-dependent clotting factors) and represents the pharmacologic target of warfarin. Multiple polymorphisms in the promoter region of VKORC1 associate with variable gene expression and warfarin dose requirements in people of European and African descent [82,87]. GWAS have identified VKORC1 and CYP2C9 as the primary contributors to differential warfarin responsiveness, together accounting for approximately 40% of variation, with other genes contributing only small effects [88,89].

The epoxide form of vitamin K may also be metabolized to an inactive form by the enzyme vitamin K1 oxidase (CYP4F2). Polymorphisms in CYP4F2 have been associated with variable warfarin dose requirements [90,91]; however, findings have been inconsistent [92]. Additional genes have been identified that affect warfarin responsiveness, including APOE, glutamyl carboxylase (GGCX), calumenin (CALU), CYP4F2, epoxide hydrolase 1 (EPHX1), and factor VII (F7) [63]; however, their collective contribution to warfarin dosing variability is less than that of CYP2C9 and VKORC1. When clinical information such as age, sex, and concomitant drug therapy are combined with VKORC1 and CYP2C9 polymorphisms, 50–60% of variation in warfarin dose can be explained [93]. Given these compelling data, the FDA has included the range of expected therapeutic warfarin doses based on CYP2C9 and VKORC1 genotypes in the warfarin labeling [94]. Prospective trials are underway to determine whether a genotype-guided dosing strategy improves patient care [95••, 96, 97].

Two novel oral anticoagulants have recently received FDA approval. Dabigatran, a reversible direct thrombin inhibitor, was approved in 2010 for the indication stroke prevention in atrial fibrillation, and rivaroxaban, a factor Xa inhibitor, was approved for deep venous thrombosis prophylaxis after orthopedic surgery in 2011. Dabigatran is administered as a pro-drug that is rapidly biotrans-formed after absorption. Both dabigatran and rivaroxaban are substrates for the drug efflux pump P-glycoprotein, coded by the ABCB1 gene, and dabigatran (but not rivaroxaban) undergoes metabolism by CYP450 enzymes, predominantly CYP3A4 and CYP2J2 [98100]. To date, no data regarding pharmacogenomic studies of these agents have been published and it is currently unknown whether genetic variants modulate their effectiveness.

Statins

3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) catalyzes the rate-limiting step of cholesterol biosynthesis. HMGCR inhibitors, or statins, lower serum cholesterol by blocking endogenous cholesterol synthesis, with subsequent up-regulation of the low-density lipoprotein receptor (LDLR). Statins are very effective for the treatment and prevention of cardiovascular disease. However, they are also associated with muscle toxicity that is commonly mild, but can rarely present as rhabdomyolysis.

Genetic variants have also been shown to modulate statin dosing and efficacy. Atorvastatin, simvastatin, and lovastatin are inactivated via CYP3A4 and common polymorphisms in this gene are believed to affect dosing requirements through pharmacokinetic effects [101]. Also, certain haplotypes, combinations of alleles (or SNPs) at adjacent locations on the chromosome used to identify other polymorphic sites in the region, have been associated with statin responsiveness. The H7 haplotype of HMGCR, defined by the presence of three intronic SNPs, rs17244841, rs3846662, and rs17238540, leads to an 11–19% reduction in statin sensitivity, and is thought to involve variable production of an HMGCR isoform through alternate splicing [102106]. In individuals of African descent, the L5 hap-lotype of LDLR, defined by six SNPs within the LDLR 3′ untranslated region, is associated with reduced statin responsiveness, possibly related to attenuated induction of LDLR surface expression [101,107]. Statin efficacy for those who carry both the H7 HMGCR haplotype and L5 LDLR haplotypeis further reduced. Other associations between genetic variants and statin efficacy are less clear. CLMN, encoding calmin, a protein of unknown function, has been identified through combined GWA analysis from three clinical trials [108], but the finding requires further study. Additionally, some studies have found an association between the Trp719Arg variant in kinesin-like protein 6 (KIF6) and coronary artery disease (CAD) as well as an increased protective effect from events by statins [109,110]. However, the association with CAD was not seen in a recent large replication study [111], casting uncertainty on the relationship.

Important genetic associations have also been made with statin myotoxicity. A GWAS including only 85 individuals with myotoxicity while taking high-dose simvastatin was nevertheless successful in identifying the V174A variant in the drug uptake transporter encoded by SLCO1B1 as a risk allele. Strikingly, this variant carried an odds ratio (OR) of 16.9 (95% CI 4.7–61.1) for myopathy in homozy-gotes and 4.5 (95% CI 2.6–7.7) in heterozygotes and accounted for approximately 60% of the risk in this population [112]. The SLCO1B1 variant has been independently validated [113,114]. SLCO1B1 genotype information may be integrated into clinical care to inform providers regarding patient risk of myotoxicity and help guide therapeutic decisions [115].

Angiotensin-Converting Enzyme Inhibitors

Most data regarding genomic modulators of angiotensin-converting enzyme (ACE) inhibitor response refer to a common insertion/deletion (I/D) polymorphism in the ACE gene that is strongly correlated with plasma enzyme levels [116,117]. In an observational study, carriers of the DD genotype were found to have significantly higher mortality than those with the I/I genotype, with the risk for heterozygotes being intermediate [118]. However, prospective trials have failed to validate these findings [119,120] and, at present, there is no strong evidence that this variant influences response to ACE inhibitors. A number of other candidates in the renin–angiotensin–aldosterone pathway have been analyzed, including angiotensinogen (AGT) and angiotensin II receptor types I and II (AGTR1 and AGTR2). Signals for higher risk of adverse cardiovascular outcomes while taking ACE inhibitors have emerged with variants in AGT[63]; however, the data are not conclusive.

Recently the Perindopril Genetic Association study (PERGENE) [121•] evaluated 12 genes from the pharmacodynamic pathway of ACE inhibitors in 8907 patients with stable CAD treated with perindopril or placebo. Two polymorphisms in AGTR1 and a third in the bradykinin type I receptor were significantly associated with the combined primary outcome of cardiovascular mortality, nonfatal myocardial infarction, and resuscitated cardiac arrest during 4.2 years of follow-up. A pharmacogenetic score combining these SNPs demonstrated a stepwise decrease in treatment benefit of perindopril with an increasing score and identified a subgroup of 26.5% of the population that did not benefit from therapy [121•]. This finding requires independent replication, but represents a potential advance toward understanding variable response to ACE inhibitors.

Arrhythmia-Related Drugs

Cardiac arrhythmias are a heterogeneous and complex collection of clinical entities associated with substantial morbidity and mortality. Atrial fibrillation, for example, is the commonest sustained arrhythmia encountered in clinical practice and affects more than 2 million Americans [122]. Treatment of atrial fibrillation may be directed at maintenance of normal sinus rhythm or focus on control of ventricular rate. In either case, management is characterized by variable drug response and high recurrence rates. Functional variants influencing drug response in atrial fibrillation have recently been described. The ADRB1 Arg389Gly polymorphism has been associated with improved rate control in patients with atrial fibrillation [123]. A SNP (rs10033464) at the 4q25 locus near the PITX2 gene, which has been associated with increased risk of atrial fibrillation, appears to modulate response to antiarrhythmic drugs and predicts successful rhythm control [124].

Life-threatening arrhythmias, such as Torsade de Pointes (TdP), may also arise as a complication of drug treatment. This adverse event occurs in association with an exaggerated prolongation of the QT interval, most commonly, though not exclusively, due to certain antiarrhythmic agents [125127]. Small cohort studies in which known long-QT disease genes were genotyped identified variants in these ion channel genes in up to 15% of affected patients [128,129]. Using a candidate approach in which nearly 1500 SNPs in 18 key genes related to cardiac electrical properties were genotyped, the nonsynonymous SNP KCNE1-D85N (rs1805128) was significantly associated with increased risk of TdP with an OR of 9.0 [130••]. This SNP, which codes a potassium channel known to modulate an important cardiac current, had also been identified in prior candidate gene studies and is relatively common in the population: minor allele frequency 1–2% [128,131].

Conclusion

Despite improvements in the efficacy and safety of pharmacologic therapies for cardiovascular disease, patients vary considerably in their benefit from therapy and propensity for adverse drug events. There is an increasing appreciation that genetic variation is an important contributor to this variability, and understanding these pharmacogenomic relationships has provided key insights into mechanisms of disease and drug response. The field is in an exciting stage of rapid discovery, and the most robust findings are undergoing translation into clinical practice. Understanding the genomic basis of variable drug response is a key step in making personalized medicine a reality.

Key Points.

  • Genetic variation within individuals and populations contributes to the variable efficacy and toxicity of many important cardiovascular drugs.

  • A number of pharmacogenomic associations have shown clinical relevance and are included in FDA labels.

  • Prospective trials evaluating genomic data care are underway.

  • Translation of pharmacogenomic findings into routine clinical care will be challenging, but early efforts have begun.

Acknowledgments

The authors would like to acknowledge the assistance and contributions of Drs Andrea Ramirez, Jonathan Mosley, Sara Van Driest and Peter Weeke. Q.S.W. is supported, in part, by NIH grant T32HL105334. J.T.D. is supported, in part, by NIH grant GM007569. D.M.R. is supported, in part, by the NIH Pharmacogenetics Research Network (Grant number U01 HL65962).

Footnotes

Conflicts of interest: D.M.R.: Consultation (current): Astellas, Sanofi. Consultation (prior): Merck, Novartis, Warner-Chilcott, Pfizer, Avanir, Vltal Pharmaceutical, ARCA, Sankyo Daiichi. Patents/Royalties: U.S. Letters Patents No. 6456542, issued October 1, 2002 for ‘Method of Screening for Susceptibility to Drug-Induced Cardiac Arrhyth-mia’. The other authors have no conflicts of interest.

References and Recommended Reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest

•• of outstanding interest

Additional references related to this topic can also be found in the Current World Literature section in this issue (p. 322).

  • 1.Ioannidis JP, Ntzani EE, Trikalinos TA, Contopoulos-Ioannidis DG. Replication validity of genetic association studies. Nat Genet. 2001;29:306–309. doi: 10.1038/ng749. [DOI] [PubMed] [Google Scholar]
  • 2.Wilke RA, Dolan ME. Genetics and variable drug response. JAMA. 2011;306:306–307. doi: 10.1001/jama.2011.998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.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;52:644–651. doi: 10.1016/j.jacc.2008.05.022. [DOI] [PubMed] [Google Scholar]
  • 4.Bartholow M Top 200 Prescription Drugs of 2009. [Accessed 19 November 2011]; http://www.pharmacytimes.com/issue/pharmacy/2010/May2010/RxFocusTopDrugs.
  • 5.Kazui M, Nishiya Y, Ishizuka T, et al. Identification of the human cytochrome P450 enzymes involved in the two oxidative steps in the bioactivation of clopidogrel to its pharmacologically active metabolite. Drug Metab Dispos. 2010;38:92–99. doi: 10.1124/dmd.109.029132. [DOI] [PubMed] [Google Scholar]
  • 6.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;108:2244–2247. doi: 10.1182/blood-2006-04-013052. [DOI] [PubMed] [Google Scholar]
  • 7.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;5:2429–2436. doi: 10.1111/j.1538-7836.2007.02775.x. [DOI] [PubMed] [Google Scholar]
  • 8.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;51:1925–1934. doi: 10.1016/j.jacc.2007.12.056. [DOI] [PubMed] [Google Scholar]
  • 9.Harmsze A, van Werkum JW, Bouman HJ, et al. Besides CYP2C19*2, the variant allele CYP2C9*3 is associated with higher on-clopidogrel platelet reactivity in patients on dual antiplatelet therapy undergoing elective coronary stent implantation. Pharmacogenet Genomics. 2010;20:18–25. doi: 10.1097/FPC.0b013e328333dafe. [DOI] [PubMed] [Google Scholar]
  • 10.Hochholzer W, Trenk D, Fromm MF, et al. Impact of cytochrome P450 2C19 loss-of-function polymorphism and of major demographic characteristics on residual platelet function after loading and maintenance treatment with clopidogrel in patients undergoing elective coronary stent placement. J Am Coll Cardiol. 2010;55:2427–2434. doi: 10.1016/j.jacc.2010.02.031. [DOI] [PubMed] [Google Scholar]
  • 11.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;302:849–857. doi: 10.1001/jama.2009.1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 polymorphisms and response to clopidogrel. N Engl J Med. 2009;360:354–362. doi: 10.1056/NEJMoa0809171. [DOI] [PubMed] [Google Scholar]
  • 13.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;107:2908–2913. doi: 10.1161/01.CIR.0000072771.11429.83. [DOI] [PubMed] [Google Scholar]
  • 14.Ellis KJ, Stouffer GA, McLeod HL, Lee CR. Clopidogrel pharmacogenomics and risk of inadequate platelet inhibition: US FDA recommendations. Pharmacogenomics. 2009;10:1799–1817. doi: 10.2217/pgs.09.143. [DOI] [PubMed] [Google Scholar]
  • 15.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;373:309–317. doi: 10.1016/S0140-6736(08)61845-0. [DOI] [PubMed] [Google Scholar]
  • 16.Sibbing D, Stegherr J, Latz W, et al. Cytochrome P450 2C19 loss-of-function polymorphism and stent thrombosis following percutaneous coronary intervention. Eur Heart J. 2009;30:916–922. doi: 10.1093/eurheartj/ehp041. [DOI] [PubMed] [Google Scholar]
  • 17.Harmsze AM, van Werkum JW, Ten Berg JM, et al. CYP2C19*2 and CYP2C9*3 alleles are associated with stent thrombosis: a case-control study. Eur Heart J. 2010;31:3046–3053. doi: 10.1093/eurheartj/ehq321. [DOI] [PubMed] [Google Scholar]
  • 18.Sofi F, Giusti B, Marcucci R, et al. Cytochrome P450 2C19 *2 polymorphism and cardiovascular recurrences in patients taking clopidogrel: a meta-analysis. Pharmacogenomics J. 2011;11:199–206. doi: 10.1038/tpj.2010.21. [DOI] [PubMed] [Google Scholar]
  • 19•.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;304:1821–1830. doi: 10.1001/jama.2010.1543. Finds that, among patients treated with clopidogrel for percutaneous coronary intervention, carriage of even one reduced-function CYP2C19 allele appears to be associated with a significantly increased risk of major adverse cardiovascular events, particularly stent thrombosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20•.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;56:134–143. doi: 10.1016/j.jacc.2009.12.071. A global meta-analysis on the association between the loss-of-function cytochrome P450 2C19 (CYP2C19)*2 variant or the use of proton pump inhibitors (PPIs) and ischemic outcomes in patients treated with clopidogrel. [DOI] [PubMed] [Google Scholar]
  • 21•.Delaney JT, Ramirez AH, Bowton E, et al. Predicting clopidogrel response using DNA samples linked to an electronic health record. Clin Pharmacol Ther. 2012;91:257–263. doi: 10.1038/clpt.2011.221. Data do not show an association between PON1 and recurrent cardiovascular events. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.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;376:1320–1328. doi: 10.1016/S0140-6736(10)61274-3. [DOI] [PubMed] [Google Scholar]
  • 23.Pare G, Mehta SR, Yusuf S, et al. Effects of CYP2C19 genotype on outcomes of clopidogrel treatment. N Engl J Med. 2010;363:1704–1714. doi: 10.1056/NEJMoa1008410. [DOI] [PubMed] [Google Scholar]
  • 24.Sibbing D, Koch W, Gebhard D, et al. Cytochrome 2C19*17 allelic variant, platelet aggregation, bleeding events, and stent thrombosis in clopidogreltreated patients with coronary stent placement. Circulation. 2010;121:512–518. doi: 10.1161/CIRCULATIONAHA.109.885194. [DOI] [PubMed] [Google Scholar]
  • 25.Tiroch KA, Sibbing D, Koch W, et al. Protective effect of the CYP2C19*17 polymorphism with increased activation of clopidogrel on cardiovascular events. Am Heart J. 2010;160:506–512. doi: 10.1016/j.ahj.2010.06.039. [DOI] [PubMed] [Google Scholar]
  • 26.Taubert D, von Beckerath N, Grimberg G, et al. Impact of P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther. 2006;80:486–501. doi: 10.1016/j.clpt.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 27.Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med. 2009;360:363–375. doi: 10.1056/NEJMoa0808227. [DOI] [PubMed] [Google Scholar]
  • 28.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;90:287–295. doi: 10.1038/clpt.2011.127. [DOI] [PubMed] [Google Scholar]
  • 29.Kassimis G, Davlouros P, Xanthopoulou I, et al. CYP2C19*2 and other genetic variants affecting platelet response to clopidogrel in patients undergoing percutaneous coronary intervention. Thromb Res. 2011 doi: 10.1016/j.thromres.2011.07.022. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 30.Bouman HJ, Schomig E, van Werkum JW, et al. Paraoxonase-1 is a major determinant of clopidogrel efficacy. Nat Med. 2011;17:110–116. doi: 10.1038/nm.2281. [DOI] [PubMed] [Google Scholar]
  • 31•.Hulot JS, Collet JP, Cayla G, et al. CYP2C19 but not PON1 genetic variants influence clopidogrel pharmacokinetics, pharmacodynamics, and clinical efficacy in post-myocardial infarction patients. Circ Cardiovasc Interv. 2011;4:422–428. doi: 10.1161/CIRCINTERVENTIONS.111.963025. This study does not confirm that PON1 Q192R or L55M can influence clopidogrel pharmacokinetics or pharmacodynamics in post-MI patients. [DOI] [PubMed] [Google Scholar]
  • 32.Lewis JP, Fisch AS, Ryan K, et al. Paraoxonase 1 (PON1) gene variants are not associated with clopidogrel response. Clin Pharmacol Ther. 2011;90:568–574. doi: 10.1038/clpt.2011.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sibbing D, Koch W, Massberg S, et al. No association of paraoxonase-1 Q192R genotypes with platelet response to clopidogrel and risk of stent thrombosis after coronary stenting. Eur Heart J. 2011;32:1605–1613. doi: 10.1093/eurheartj/ehr155. [DOI] [PubMed] [Google Scholar]
  • 34.Simon T, Steg PG, Becquemont L, et al. Effect of paraoxonase-1 polymorphism on clinical outcomes in patients treated with clopidogrel after an acute myocardial infarction. Clin Pharmacol Ther. 2011;90:561–567. doi: 10.1038/clpt.2011.193. [DOI] [PubMed] [Google Scholar]
  • 35.Trenk D, Hochholzer W, Fromm MF, et al. Paraoxonase-1 Q192R polymorphism and antiplatelet effects of clopidogrel in patients undergoing elective coronary stent placement. Circ Cardiovasc Genet. 2011;4:429–436. doi: 10.1161/CIRCGENETICS.111.960112. [DOI] [PubMed] [Google Scholar]
  • 36.Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, et al. Lack of association between the P2Y12 receptor gene polymorphism and platelet response to clopidogrel in patients with coronary artery disease. Thromb Res. 2005;116:491–497. doi: 10.1016/j.thromres.2005.03.001. [DOI] [PubMed] [Google Scholar]
  • 37.von Beckerath N, von Beckerath O, Koch W, et al. P2Y12 gene H2 haplotype is not associated with increased adenosine diphosphate-induced platelet aggregation after initiation of clopidogrel therapy with a high loading dose. Blood Coagul Fibrinolysis. 2005;16:199–204. doi: 10.1097/01.mbc.0000164429.21040.0a. [DOI] [PubMed] [Google Scholar]
  • 38.Fontana P, Dupont A, Gandrille S, et al. Adenosine diphosphate-induced platelet aggregation is associated with P2Y12 gene sequence variations in healthy subjects. Circulation. 2003;108:989–995. doi: 10.1161/01.CIR.0000085073.69189.88. [DOI] [PubMed] [Google Scholar]
  • 39.Staritz P, Kurz K, Stoll M, et al. Platelet reactivity and clopidogrel resistance are associated with the H2 haplotype of the P2Y12-ADP receptor gene. Int J Cardiol. 2009;133:341–345. doi: 10.1016/j.ijcard.2007.12.118. [DOI] [PubMed] [Google Scholar]
  • 40••.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;90:328–332. doi: 10.1038/clpt.2011.132. Provides evidence from published literature and guidelines for CYPC19 genotypedirected antiplatelet therapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wiviott SD, Braunwald E, McCabe CH, et al. Intensive oral antiplatelet therapy for reduction of ischaemic events including stent thrombosis in patients with acute coronary syndromes treated with percutaneous coronary intervention and stenting in the TRITON-TIMI 38 trial: a subanalysis of a randomised trial. Lancet. 2008;371:1353–1363. doi: 10.1016/S0140-6736(08)60422-5. [DOI] [PubMed] [Google Scholar]
  • 42.Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482. [DOI] [PubMed] [Google Scholar]
  • 43•.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 pharmacogenetic analysis. Lancet. 2010;376:1312–1319. doi: 10.1016/S0140-6736(10)61273-1. Aimed to assess the effect of ABCB1 polymorphism by itself and alongside variants in CYP2C19 on cardiovascular outcomes in patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045–1057. doi: 10.1056/NEJMoa0904327. [DOI] [PubMed] [Google Scholar]
  • 45.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: the ONSET/OFFSET and RESPOND genotype studies. Circ Cardiovasc Genet. 2010;3:556–566. doi: 10.1161/CIRCGENETICS.110.958561. [DOI] [PubMed] [Google Scholar]
  • 46.Mega JL, Close SL, Wiviott SD, et al. Cytochrome P450 genetic polymorphisms and the response to prasugrel: relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Circulation. 2009;119:2553–2560. doi: 10.1161/CIRCULATIONAHA.109.851949. [DOI] [PubMed] [Google Scholar]
  • 47.Cuisset T, Quilici J, Cohen W, et al. Usefulness of high clopidogrel maintenance dose according to CYP2C19 genotypes in clopidogrel low responders undergoing coronary stenting for non ST elevation acute coronary syndrome. Am J Cardiol. 2011;108:760–765. doi: 10.1016/j.amjcard.2011.05.045. [DOI] [PubMed] [Google Scholar]
  • 48.Jeong YH, Kim IS, Park Y, et al. Carriage of cytochrome 2C19 polymorphism is associated with risk of high posttreatment 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. JACC Cardiovasc Interv. 2010;3:731–741. doi: 10.1016/j.jcin.2010.05.007. [DOI] [PubMed] [Google Scholar]
  • 49.Mega JL, Hochholzer W, Frelinger AL, et al. Dosing clopidogrel based on CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovascular disease. JAMA. 2011;306:2221–2228. doi: 10.1001/jama.2011.1703. [DOI] [PubMed] [Google Scholar]
  • 50.Brodde OE, Bruck H, Leineweber K, Seyfarth T. Presence, distribution and physiological function of adrenergic and muscarinic receptor subtypes in the human heart. Basic Res Cardiol. 2001;96:528–538. doi: 10.1007/s003950170003. [DOI] [PubMed] [Google Scholar]
  • 51.Reiter E, Lefkowitz RJ. GRKs and beta-arrestins: roles in receptor silencing, trafficking and signaling. Trends Endocrinol Metab. 2006;17:159–165. doi: 10.1016/j.tem.2006.03.008. [DOI] [PubMed] [Google Scholar]
  • 52.FDA. [Accessed 31 October 2011];Table of pharmacogenomic biomarkers in drug labels. 2011 http://www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm.
  • 53.Ismail R, Teh LK. The relevance of CYP2D6 genetic polymorphism on chronic metoprolol therapy in cardiovascular patients. J Clin Pharm Ther. 2006;31:99–109. doi: 10.1111/j.1365-2710.2006.00699.x. [DOI] [PubMed] [Google Scholar]
  • 54.Rau T, Heide R, Bergmann K, et al. Effect of the CYP2D6 genotype on metoprolol metabolism persists during long-term treatment. Pharmacogenetics. 2002;12:465–472. doi: 10.1097/00008571-200208000-00007. [DOI] [PubMed] [Google Scholar]
  • 55.Roden DM, Stein CM. Clopidogrel and the concept of high-risk pharmacokinetics. Circulation. 2009;119:2127–2130. doi: 10.1161/CIRCULATIONAHA.109.865907. [DOI] [PubMed] [Google Scholar]
  • 56.Zhou SF. Polymorphism of human cytochrome P450 2D6 and its clinical significance: part I. Clin Pharmacokinet. 2009;48:689–723. doi: 10.2165/11318030-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 57.Zhou SF. Polymorphism of human cytochrome P450 2D6 and its clinical significance: part II. Clin Pharmacokinet. 2009;48:761–804. doi: 10.2165/11318070-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 58.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;77:127–137. doi: 10.1016/j.clpt.2004.10.006. [DOI] [PubMed] [Google Scholar]
  • 59.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;76:536–544. doi: 10.1016/j.clpt.2004.08.020. [DOI] [PubMed] [Google Scholar]
  • 60.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;78:378–387. doi: 10.1016/j.clpt.2005.07.004. [DOI] [PubMed] [Google Scholar]
  • 61.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;72:429–437. doi: 10.1067/mcp.2002.127111. [DOI] [PubMed] [Google Scholar]
  • 62.Muthumala A, Drenos F, Elliott PM, Humphries SE. Role of beta adrenergic receptor polymorphisms in heart failure: systematic review and meta-analysis. Eur J Heart Fail. 2008;10:3–13. doi: 10.1016/j.ejheart.2007.11.008. [DOI] [PubMed] [Google Scholar]
  • 63.Verschuren JJ, Trompet S, Wessels JA, et al. A systematic review on pharmacogenetics in cardiovascular disease: is it ready for clinical application? Eur Heart J. 2011;33:165–175. doi: 10.1093/eurheartj/ehr239. [DOI] [PubMed] [Google Scholar]
  • 64.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;274:12670–12674. doi: 10.1074/jbc.274.18.12670. [DOI] [PubMed] [Google Scholar]
  • 65.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;9:1300–1305. doi: 10.1038/nm930. [DOI] [PubMed] [Google Scholar]
  • 66.Terra SG, Hamilton KK, Pauly DF, et al. Beta1-adrenergic receptor polymorphisms and left ventricular remodeling changes in response to betablocker therapy. Pharmacogenet Genomics. 2005;15:227–234. doi: 10.1097/01213011-200504000-00006. [DOI] [PubMed] [Google Scholar]
  • 67.Chen L, Meyers D, Javorsky G, et al. Arg389Gly-beta1-adrenergic receptors determine improvement in left ventricular systolic function in nonischemic cardiomyopathy patients with heart failure after chronic treatment with carvedilol. Pharmacogenet Genomics. 2007;17:941–949. doi: 10.1097/FPC.0b013e3282ef7354. [DOI] [PubMed] [Google Scholar]
  • 68.de Groote P, Helbecque N, Lamblin N, et al. Association between beta-1 and beta-2 adrenergic receptor gene polymorphisms and the response to betablockade in patients with stable congestive heart failure. Pharmacogenet Genomics. 2005;15:137–142. doi: 10.1097/01213011-200503000-00001. [DOI] [PubMed] [Google Scholar]
  • 69.Biolo A, Clausell N, Santos KG, et al. Impact of beta1-adrenergic receptor polymorphisms on susceptibility to heart failure, arrhythmogenesis, prognosis, and response to beta-blocker therapy. Am J Cardiol. 2008;102:726–732. doi: 10.1016/j.amjcard.2008.04.070. [DOI] [PubMed] [Google Scholar]
  • 70.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 U S A. 2006;103:11288–11293. doi: 10.1073/pnas.0509937103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Magnusson Y, Levin MC, Eggertsen R, et al. Ser49Gly of beta1-adrenergic receptor is associated with effective beta-blocker dose in dilated cardiomyopathy. Clin Pharmacol Ther. 2005;78:221–231. doi: 10.1016/j.clpt.2005.06.004. [DOI] [PubMed] [Google Scholar]
  • 72.Cresci S, Kelly RJ, Cappola TP, et al. Clinical and genetic modifiers of longterm survival in heart failure. J Am Coll Cardiol. 2009;54:432–444. doi: 10.1016/j.jacc.2009.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.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;99:250–255. doi: 10.1016/j.amjcard.2006.08.020. [DOI] [PubMed] [Google Scholar]
  • 74.Green SA, Turki J, Innis M, Liggett SB. Amino-terminal polymorphisms of the human beta 2-adrenergic receptor impart distinct agonist-promoted regulatory properties. Biochemistry. 1994;33:9414–9419. doi: 10.1021/bi00198a006. [DOI] [PubMed] [Google Scholar]
  • 75.Small KM, Forbes SL, Rahman FF, et al. A four amino acid deletion polymorphism in the third intracellular loop of the human alpha 2C-adrenergic receptor confers impaired coupling to multiple effectors. J Biol Chem. 2000;275:23059–23064. doi: 10.1074/jbc.M000796200. [DOI] [PubMed] [Google Scholar]
  • 76.Kardia SL, Kelly RJ, Keddache MA, et al. Multiple interactions between the alpha 2C- and beta1-adrenergic receptors influence heart failure survival. BMC Med Genet. 2008;9:93. doi: 10.1186/1471-2350-9-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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;3:21–28. doi: 10.1161/CIRCHEARTFAILURE.109.885962. [DOI] [PubMed] [Google Scholar]
  • 78.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;17:277–282. doi: 10.1097/FPC.0b013e3280105245. [DOI] [PubMed] [Google Scholar]
  • 79.Liggett SB, Cresci S, Kelly RJ, et al. A GRK5 polymorphism that inhibits betaadrenergic receptor signaling is protective in heart failure. Nat Med. 2008;14:510–517. doi: 10.1038/nm1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80•.O'Connor CM, Fiuzat M, Swedberg K, et al. Influence of global region on outcomes in heart failure beta-blocker trials. J Am Coll Cardiol. 2011;58:915–922. doi: 10.1016/j.jacc.2011.03.057. Looks at geographical difference in treatment response in b-blockade. [DOI] [PubMed] [Google Scholar]
  • 81.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;353:717–719. doi: 10.1016/S0140-6736(98)04474-2. [DOI] [PubMed] [Google Scholar]
  • 82.Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med. 2005;352:2285–2293. doi: 10.1056/NEJMoa044503. [DOI] [PubMed] [Google Scholar]
  • 83.Wadelius M, Chen LY, Eriksson N, et al. Association of warfarin dose with genes involved in its action and metabolism. Hum Genet. 2007;121:23–34. doi: 10.1007/s00439-006-0260-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.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;83:312–321. doi: 10.1038/sj.clpt.6100290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lindh JD, Holm L, Andersson ML, Rane A. Influence of CYP2C9 genotype on warfarin dose requirements–a systematic review and meta-analysis. Eur J Clin Pharmacol. 2009;65:365–375. doi: 10.1007/s00228-008-0584-5. [DOI] [PubMed] [Google Scholar]
  • 86.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;9:511–526. doi: 10.2217/14622416.9.5.511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ramirez AH, Shi Y, Schildcrout JS, et al. Predicting warfarin dosage in European and African Americans using DNA samples linked to an electronic health record. Pharmacogenomics. doi: 10.2217/pgs.11.164. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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. doi: 10.1371/journal.pgen.1000433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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;112:1022–1027. doi: 10.1182/blood-2008-01-134247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Caldwell MD, Awad T, Johnson JA, et al. CYP4F2 genetic variant alters required warfarin dose. Blood. 2008;111:4106–4112. doi: 10.1182/blood-2007-11-122010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.McDonald MG, Rieder MJ, Nakano M, et al. CYP4F2 is a vitamin K1 oxidase: an explanation for altered warfarin dose in carriers of the V433M variant. Mol Pharmacol. 2009;75:1337–1346. doi: 10.1124/mol.109.054833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kringen MK, Haug KB, Grimholt RM, et al. Genetic variation of VKORC1 and CYP4F2 genes related to warfarin maintenance dose in patients with myocardial infarction. J Biomed Biotechnol. 2011;2011:739751. doi: 10.1155/2011/739751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wadelius M, Chen LY, Lindh JD, et al. The largest prospective warfarintreated cohort supports genetic forecasting. Blood. 2009;113:784–792. doi: 10.1182/blood-2008-04-149070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.FDA. [accessed 16 November 2011];Warfarin label information. 2011 Updated 22 January 2010. [Google Scholar]
  • 95••.French B, Joo J, Geller NL, et al. Statistical design of personalized medicine interventions: the Clarification of Optimal Anticoagulation through Genetics (COAG) trial. Trials. 2010;11:108. doi: 10.1186/1745-6215-11-108. The COAG trial serves as an illustrative example of a personalized medicine intervention that uses each individual's genotype information. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.van Schie RM, Wadelius MI, Kamali F, et al. Genotype-guided dosing of coumarin derivatives: the European pharmacogenetics of anticoagulant therapy (EU-PACT) trial design. Pharmacogenomics. 2009;10:1687–1695. doi: 10.2217/pgs.09.125. [DOI] [PubMed] [Google Scholar]
  • 97.Gage BF. [Accessed 26 December 2011];Genetics Informatics Trial (GIFT) of warfarin to prevent DVT 2011. doi: 10.1038/tpj.2011.18. http://www.clinicaltrials.gov. [DOI] [PMC free article] [PubMed]
  • 98.Gnoth MJ, Buetehorn U, Muenster U, et al. In vitro and in vivo P-glycoprotein transport characteristics of rivaroxaban. J Pharmacol Exp Ther. 2011;338:372–380. doi: 10.1124/jpet.111.180240. [DOI] [PubMed] [Google Scholar]
  • 99.Blech S, Ebner T, Ludwig-Schwellinger E, et al. The metabolism and disposition of the oral direct thrombin inhibitor, dabigatran, in humans. Drug Metab Dispos. 2008;36:386–399. doi: 10.1124/dmd.107.019083. [DOI] [PubMed] [Google Scholar]
  • 100.Stangier J, Clemens A. Pharmacology, pharmacokinetics, and pharmacodynamics of dabigatran etexilate, an oral direct thrombin inhibitor. Clin Appl Thromb Hemost. 2009;15(Suppl 1):9S–16S. doi: 10.1177/1076029609343004. [DOI] [PubMed] [Google Scholar]
  • 101.Roden DM, Johnson JA, Kimmel SE, et al. Cardiovascular pharmacogenomics. Circ Res. 2011;109:807–820. doi: 10.1161/CIRCRESAHA.110.230995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Medina MW, Gao F, Ruan W, et al. Alternative splicing of 3-hydroxy-3-methylglutaryl coenzyme A reductase is associated with plasma low-density lipoprotein cholesterol response to simvastatin. Circulation. 2008;118:355–362. doi: 10.1161/CIRCULATIONAHA.108.773267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Chasman DI, Posada D, Subrahmanyan L, et al. Pharmacogenetic study of statin therapy and cholesterol reduction. JAMA. 2004;291:2821–2827. doi: 10.1001/jama.291.23.2821. [DOI] [PubMed] [Google Scholar]
  • 104.Donnelly LA, Doney AS, Dannfald J, et al. A paucimorphic variant in the HMGCoA reductase gene is associated with lipid-lowering response to statin treatment in diabetes: a GoDARTS study. Pharmacogenet Genomics. 2008;18:1021–1026. doi: 10.1097/FPC.0b013e3283106071. [DOI] [PubMed] [Google Scholar]
  • 105.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;117:1537–1544. doi: 10.1161/CIRCULATIONAHA.107.708388. [DOI] [PubMed] [Google Scholar]
  • 106.Poduri A, Khullar M, Bahl A, et al. Common variants of HMGCR, CETP, APOAI, ABCB1, CYP3A4, and CYP7A1 genes as predictors of lipid-lowering response to atorvastatin therapy. DNA Cell Biol. 2010;29:629–637. doi: 10.1089/dna.2009.1008. [DOI] [PubMed] [Google Scholar]
  • 107.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;30:1485–1492. doi: 10.1161/ATVBAHA.110.203273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Barber MJ, Mangravite LM, Hyde CL, et al. Genome-wide association of lipidlowering response to statins in combined study populations. PLoS One. 2010;5:e9763. doi: 10.1371/journal.pone.0009763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Iakoubova OA, Robertson M, Tong CH, et al. KIF6 Trp719Arg polymorphism and the effect of statin therapy in elderly patients: results from the PROSPER study. Eur J Cardiovasc Prev Rehabil. 2010;17:455–461. doi: 10.1097/HJR.0b013e328336a0dd. [DOI] [PubMed] [Google Scholar]
  • 110.Li Y, Iakoubova OA, Shiffman D, et al. KIF6 polymorphism as a predictor of risk of coronary events and of clinical event reduction by statin therapy. Am J Cardiol. 2010;106:994–998. doi: 10.1016/j.amjcard.2010.05.033. [DOI] [PubMed] [Google Scholar]
  • 111.Assimes TL, Holm 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;56:1552–1563. doi: 10.1016/j.jacc.2010.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Link E, Parish S, Armitage J, et al. SLCO1B1 variants and statin-induced myopathy: a genomewide study. N Engl J Med. 2008;359:789–799. doi: 10.1056/NEJMoa0801936. [DOI] [PubMed] [Google Scholar]
  • 113.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;54:1609–1616. doi: 10.1016/j.jacc.2009.04.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.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;89:210–216. doi: 10.1038/clpt.2010.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Pulley JM, Denny JC, Peterson JF, et al. Operational implementation of prospective genotyping for personalized medicine:The design of the Vanderbilt PREDICT project. Clin Pharmacol Ther. doi: 10.1038/clpt.2011.371. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Rigat B, Hubert C, Alhenc-Gelas F, et al. An insertion/deletion polymorphism in the angiotensin I-converting enzyme gene accounting for half the variance of serum enzyme levels. J Clin Invest. 1990;86:1343–1346. doi: 10.1172/JCI114844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Tiret L, Rigat B, Visvikis S, et al. Evidence, from combined segregation and linkage analysis, that a variant of the angiotensin I-converting enzyme (ACE) gene controls plasma ACE levels. Am J Hum Genet. 1992;51:197–205. [PMC free article] [PubMed] [Google Scholar]
  • 118.Bleumink GS, Schut AF, Sturkenboom MC, et al. Mortality in patients with hypertension on angiotensin-I converting enzyme (ACE)-inhibitor treatment is influenced by the ACE insertion/deletion polymorphism. Pharmacogenet Genomics. 2005;15:75–81. doi: 10.1097/01213011-200502000-00003. [DOI] [PubMed] [Google Scholar]
  • 119.Arnett DK, Boerwinkle E, Davis BR, et al. Pharmacogenetic approaches to hypertension therapy: design and rationale for the Genetics of Hypertension Associated Treatment (GenHAT) study. Pharmacogenomics J. 2002;2:309–317. doi: 10.1038/sj.tpj.6500113. [DOI] [PubMed] [Google Scholar]
  • 120.Harrap SB, Tzourio C, Cambien F, et al. The ACE gene I/D polymorphism is not associated with the blood pressure and cardiovascular benefits of ACE inhibition. Hypertension. 2003;42:297–303. doi: 10.1161/01.HYP.0000088322.85804.96. [DOI] [PubMed] [Google Scholar]
  • 121•.Brugts JJ, Isaacs A, Boersma E, et al. Genetic determinants of treatment benefit of the angiotensin-converting enzyme-inhibitor perindopril in patients with stable coronary artery disease. Eur Heart J. 2010;31:1854–1864. doi: 10.1093/eurheartj/ehq169. First to identify genetic determinants of treatment benefit of ACE-inhibitor therapy. [DOI] [PubMed] [Google Scholar]
  • 122.Feinberg WM, Blackshear JL, Laupacis A, et al. Prevalence, age distribution, and gender of patients with atrial fibrillation. Analysis and implications Arch Intern Med. 1995;155:469–473. [PubMed] [Google Scholar]
  • 123.Parvez B, Chopra N, Rowan S, et al. A common β1-adrenergic receptor polymorphism predicts favorable response to rate-control therapy in atrial fibrillation. J Am Coll Cardiol. 2012;59:49–56. doi: 10.1016/j.jacc.2011.08.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Parvez B, Vaglio J, Rowan S, et al. Symptomatic response to antiarrhythmic drug therapy is modulated by a common single nucleotide polymorphism in atrial fibrillation. J Am Coll Cardiol. doi: 10.1016/j.jacc.2012.01.070. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Fenichel RR, Malik M, Antzelevitch C, et al. Drug-induced torsades de pointes and implications for drug development. J Cardiovasc Electrophysiol. 2004;15:475–495. doi: 10.1046/j.1540-8167.2004.03534.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Haverkamp W, Breithardt G, Camm AJ, et al. The potential for QT prolongation and proarrhythmia by nonantiarrhythmic drugs:clinical and regulatory implications Report on a policy conference of the European Society of Cardiology. Eur Heart J. 2000;21:1216–1231. doi: 10.1053/euhj.2000.2249. [DOI] [PubMed] [Google Scholar]
  • 127.Roden DM. Drug-induced prolongation of the QT interval. N Engl J Med. 2004;350:1013–1022. doi: 10.1056/NEJMra032426. [DOI] [PubMed] [Google Scholar]
  • 128.Paulussen AD, Gilissen RA, Armstrong M, et al. Genetic variations of KCNQ1, KCNH2, SCN5A, KCNE1, and KCNE2 in drug-induced long QT syndrome patients. J Mol Med (Berl) 2004;82:182–188. doi: 10.1007/s00109-003-0522-z. [DOI] [PubMed] [Google Scholar]
  • 129.Yang P, Kanki H, Drolet B, et al. Allelic variants in long-QT disease genes in patients with drug-associated torsades de pointes. Circulation. 2002;105:1943–1948. doi: 10.1161/01.cir.0000014448.19052.4c. [DOI] [PubMed] [Google Scholar]
  • 130••.Kaab S, Crawford DC, Sinner MF, et al. A large candidate gene survey identifies the KCNE1 D85N polymorphism as a possible modulator of druginduced torsades de pointes. Circ Cardiovasc Genet. 2011 doi: 10.1161/CIRCGENETICS.111.960930. [Epub ahead of print] Identified a key potassium channel susceptibility allele that may be associated with the rare adverse drug reaction torsades de pointes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Nishio Y, Makiyama T, Itoh H, et al. D85N, a KCNE1 polymorphism, is a disease-causing gene variant in long QT syndrome. J Am Coll Cardiol. 2009;54:812–819. doi: 10.1016/j.jacc.2009.06.005. [DOI] [PubMed] [Google Scholar]

RESOURCES