Abstract
Variability in drug responsiveness is a sine qua non of modern therapeutics, and the contribution of genomic variation is increasingly recognized. Investigating the genomic basis for variable responses to cardiovascular therapies has been a model for pharmacogenomics in general and has established critical pathways and specific loci modulating therapeutic responses to commonly used drugs such as clopidogrel, warfarin, and statins. In addition, genomic approaches have defined mechanisms and genetic variants underlying important toxicities with these and other drugs. These findings have not only resulted in changes to the product labels but also have led to development of initial clinical guidelines that consider how to facilitate incorporating genetic information to the bedside. This review summarizes the state of knowledge in cardiovascular pharmacogenomics and considers how variants described to date might be deployed in clinical decision making.
Keywords: Pharmacogenomics; Polymorphism; Genetics; Clopidogrel; Warfarin; Statin; Beta-blocker, Anti-arrhythmic agents; Cardiovascular disease; Drug responsiveness; Toxicity
Introduction
Pharmacogenomic research aims to identify how genetic variability affects drug responsiveness and toxicity. Substantial progress has been made over the past decade in improving our understanding of genetic determinants influencing response to cardiovascular drugs such as warfarin, clopidogrel, and simvastatin. These advances have fueled a hope for “personalized medicine” which includes tailoring of diagnostic and treatment strategies to the needs and characteristics of individual patients – including genomic variation as well as other features such as personal preferences or access to care – aiming to improve drug responsiveness and lessen risk of toxicity. This article describes recent findings in the realm of cardiovascular pharmacogenomics and discusses the potential for clinical implication.
Clopidogrel
Clopidogrel is a thienopyridine derivative commonly used to prevent thromboembolic events including myocardial infarction (MI) and stent thrombosis among patients with cardiovascular disease. The antiplatelet activity of clopidogrel is characterized by considerable inter-individual responsiveness with certain genotypes causing attenuated platelet inhibition and ultimately more adverse cardiovascular events [1]. Clopidogrel is a prodrug that requires oxidation by cytochrome P-450 (CYP) iso-enzymes to generate the pharmacologically active thiol-containing metabolite that inhibits the platelet adenosine diphosphate receptor, P2RY12 [2].
Polymorphisms in CYP2C19, the major CYP isoform mediating clopidogrel bioactivation, have been associated with both decreased and increased enzymatic function. CYP2C19*2 (rs4244285) is the most common loss-of-function allele and is associated with impaired clopidogrel activity, i.e., decreased platelet inhibition [3-6]. Homozygous carriers for the loss-of-function allele (*2/*2; “poor metabolizers”) have decreased platelet inhibition compared to individuals who carry the reference alleles (*1/*1; “extensive metabolizers”), and heterozygotes (*1/*2; “intermediate metabolizers”) [4, 7, 8]. The *1/*2 genotype is found in ~25 % of Caucasians, and ~2 % are *2/*2 [9••] Other loss-of-function alleles that mimic the effects of CYP2C19*2 have also been identified: *3 (rs498693), *4 (rs28399504), *5 (rs72552267). These are rare (minor allele frequency [MAF] <1 %) in Caucasian populations, but *3 are common among Asians (MAF: 2-9 %). In contrast to these loss-of-function alleles, *17 (rs3758581) is a gain-of-function allele and has been associated with increased enzymatic activity and improved platelet inhibition [10]. Carriers of the *17 variant have been termed ultra-rapid metabolizers.
In acute coronary syndrome (ACS) and percutaneous intervention (PCI), the CYP2C19*2 loss-of-function variant has been associated with increased risk of adverse cardiovascular events including stent thrombosis during clopidogrel treatment (Table 1) [7, 10-12, 13••, 14•]. Table 1 lists pharmacogenomic targets where the level of evidence of clinical implementation is strong. Data from a meta-analysis on 9685 clopidogrel treated patients identified a gene dose–response relationship between the number of loss-of-function variants and the composite end-point of cardiovascular death, myocardial infarction, and stroke. Compared to non-carriers, carriers of one loss-of-function allele had increased risk of a cardiovascular event (hazard ratio (HR) = 1.55, 95 % confidence interval (CI): 1.11-2.17) and carriers of two loss-of-function variants had an even greater risk (HR = 1.76, CI: 1.24-2.50) (Table 1) [13••]. Notably, the risk of stent thrombosis was particularly high among carriers of one loss-of-function variant (HR = 2.81, CI: 1.81-4.37), and even higher in homozygous (*2/*2) patients (HR = 3.97, CI: 1.75-9.02) (Table 2) [13••]. This pharmacogenomic interaction between clopidogrel and carriers of a CYP2C19*2 loss-of-function variant was not been replicated in other analyses [15, 16]; one possible explanation is that the interaction is especially important in ACS/PCI compared to other indications for clopidogrel. The Food and Drug Administration (FDA) has issued a black-box warning that loss-of-function carriers may exhibit lower capacity to metabolize clopidogrel with subsequent elevated risk of thromboembolic complications [17]. Conversely, carriers of the gain-of-function variant allele *17 have been reported to display increased risk of bleeding [18] and increased protection from ischemic events [16]; these data have not yet been widely replicated (Table 2). Table 2 lists the pharmacogenomic targets where current level of evidence for clinical application is weak.
Table 1.
Cardiovascular pharmacogenomic targets with strong level of evidence for clinical implementation
| Gene | Variant | Chr | Position | Reported allele frequency of the risk-allele (%) |
Drug | Drug response associated with risk allele |
Possible clinical implications |
||
|---|---|---|---|---|---|---|---|---|---|
| Caucasians | Africans | Asians | |||||||
| CYP2C19 | *2 (rs4244285) | 10 | 96531606 | 16 | 14 | 27 | Clopidogrel | Reduced active metabolite concentration. Impaired platelet inhibition. |
Carriers of the risk allele who are treated for ACS or PCI have a graded risk of cardiovascular complications with 0,1, or 2 alleles including MI, stroke, and stent thrombosis |
| VKORC1 | −1639G>A (rs9923231) | 16 | 31107689 | 60 | 98 | 2 | Warfarin | Carriers of the A allele require lower warfarin dosing. Increased time to stable dose |
|
| CYP2C9 | *2 (rs1799853) | 10 | 96702047 | 10 | <1 | <1 | Warfarin | Reduced clearance of the potent S-warfarin |
Stepwise reduction in warfarin dosing required with 0, 1, or 2 alleles. Increased risk of bleeding |
| CYP2C9 | *3 (rs1057910) | 10 | 96741053 | 6 | <1 | 4 | Warfarin | Reduced clearance of the potent S-warfarin |
Stepwise reduction in warfarin dosing required with 0, 1, or 2 alleles. Increased risk of bleeding |
| SLC01B1 | rs4149056 | 12 | 21331549 | 15 | <1 | 13 | Simvastatin | Impaired transporter function leading to increased simvastatin levels |
Simvastatin induced myopathy, particularly among patients on high dose simvastatin |
ACS acute coronary syndrome; PCI percutaneous coronary intervention; MI myocardial infarction
Table 2.
Cardiovascular pharmacogenomic targets with weaker level of evidence for clinical implementation
| Gene | Variant | Chr | Position | Reported allele frequency of the risk-allele (%) |
Drug | Drug response associated with risk allele |
Possible clinical implications | ||
|---|---|---|---|---|---|---|---|---|---|
| Caucasians | Africans | Asians | |||||||
| ABCB1 | C3435T (rs1045642) | 7 | 89676581 | 57 | 11 | 46 | Clopidogrel | Reduced bioavailability of substrate drug |
Reduced platelet inhibition |
| CYP2C19 | *17 (rs12248560) | 10 | 96511647 | 5 | 10 | 6 | Clopidogrel | Increased active metabolite concentration. Increased platelet inhibition |
Increased risk of bleeding |
| CYP2C19 | *3 (rs4986893) | 10 | 96540410 | <1 | 2-9 | Clopidogrel | Reduced active metabolite concentration. Impaired platelet inhibition. |
Carriers of the risk allele have increased risk of stent thrombosis following PCI |
|
| ADBR1 | Ser49Gly (rs1801252) | 10 | 115804036 | 20 | <1 | <1 | Metoprolol | Impaired down-regulation of ADBR1 leading to higher signal transduction |
Carriers of risk allele have greater reductions in BP and HR |
| ADBR1 | Arg389Gly (rsl801253) | 10 | 115805056 | 31 | 41 | 20 | Metoprolol | Impaired down-regulation of ADBR1 leading to higher signal transduction |
Carriers of risk allele have greater reductions in BP and HR as well as improved LVEF |
| CYP2D6 | *4 (rs3892097) and many others |
22 | 42524947 | 18 | 23 | Metoprolol | Impaired metabolism | Increased plasma metoprolol concentrations with reduction in BP and HR. Increased risk of ADR especially among vulnerable HF patients |
|
| KCNE1 | D85N (Rsl805128) | 21 | 35821680 | 5 | 4 | 2 | Multiple | Modulates important electronic currents of the heart |
Attenuated risk of TdP |
| HMGCR | H7 haplotype (defined by three intronic SNPs: rs17244841, rs3846662, and rsl7238540) |
5 | 74642855, 74651084, 74655498 |
3 | 6 | Simvastatin, pravastatin |
Alternatively spliced MHGCR transcript that is less sensitive to therapy |
Impaired LDLc lowering | |
| APOE |
ε2, ε3, ε4 (defined by rs7412 and rs429358) |
19 | 45412079, 45411941 |
Statins | Major binding protein for VLDL/IDL cholesterol |
Impaired LDLc reduction | |||
PCI percutaneous coronary intervention; BP blood pressure; HR heart rate; LVEF left ventricular ejection fraction; ADR adverse drug reaction; TdP Torsade de Pointes
ABCB1 encodes P-glycoprotein, an ATP-dependent drug efflux transporter. Carriers of a common polymorphism C3435T in ABCB1 (homozygous carriers in particular) have been shown to have reduced bioavailability of substrate drugs such as clopidogrel [19] and have higher rates of adverse cardiovascular outcomes [15, 20, 21••], although this finding has not been consistent (Table 2) [22]. Patients treated with prasugrel or ticagrelor are not seemingly affected by common variants at ABCB1 or CYP2C19 [15, 23].
Common variants in P2YR12 (rs6798347, rs6787801, rs9859552, rs6801273, rs9848789, and rs2046934), a G-protein-coupled ADP receptor involved in regulation of platelet aggregation, have been associated with improved platelet inhibition in a candidate-gene study [24]. However, this finding has not been consistently replicated [20]. One report found that the common Q192R (rs662) variant in Paraoxonase-1 (PON1), thought to be involved in the bioactivation of clopidogrel, had greater risk of stent thrombosis [25], although this finding has not been replicated in multiple other datasets [7, 14•, 26]. A common variant in another CYP (CYP2C9*3; rs1057910) showed an association with reduced platelet inhibition in initial reports [4], but has not been validated [7, 10].
Clinical Implications
An ACC/AHA task force did not recommend routine CYP2C19 testing [27]. The Clinical Pharmacogenetics Implementation Consortium (CPIC) poses a different question: if the genotype were available, what action, if any should be considered [9••]? In this context, CPIC recommends that patients with ACS or PCI who carry one or two copies of the loss-of-function allele should receive alternative antiplatelet agents (i.e., prasugrel or ticagrelor) in order to alleviate the risk of cardiovascular adverse events as these do not undergo extensive bioactivation by the CYP2C19 enzyme. Investigators at Vanderbilt University have already described a program implementing preemptive CYP2C19 genotyping in patients likely to receive clopidogrel [28•].
Warfarin
Vitamin K antagonists (warfarin and others) are effective and widely used for treatment and prevention of venous and arterial thromboembolic events. Warfarin targets blood coagulation by inhibiting the vitamin K epoxide reductase multiprotein complex, subunit 1 (VKORC1)[29] and inhibits the activation of vitamin K dependent clotting factors II, VII, IX, and X [30]. However, wide inter-individual dosing variability and a narrow therapeutic index necessitates frequent monitoring to balance the risks of over anticoagulation and bleeding with those of under anticoagulation and clotting and this monitoring adds to the complexity of warfarin use [31, 32]. Warfarin-related bleeding complications are one of the most common reasons for emergency room visits [33]. Variables contributing to steady state warfarin dose include clinical factors such as age, weight, diet, and interacting drugs (notably amiodarone) and genetic factors. Polymorphisms at three loci have been associated with warfarin dose: CYP2C9, VKORC1, and CYP4F2.
CYP2C9 is the predominant enzyme responsible for metabolism (to inactive metabolites) of the potent warfarin S-enantiomer [29]. Common polymorphisms CYP2C9*2 (rs1799853) and CYP2C9*3 (rs1057910) have been identified as the loci most commonly associated with reduced enzyme activity (~30-40 % and ~80-90 %, respectively) [34]. As compared with non-carriers, heterozygous carriers of the CYP2C9*2 or CYP2C9*3 reduced-function alleles require ~19 % and ~33 % reduction in dosing, respectively, and homozygous carriers require even greater reductions (~36 % and ~78 %, respectively) [35]. Carriers of CYP2C9*2 or CYP2C9*3 reduced-function alleles have also been reported to be at increased risk of hemorrhagic complications during warfarin treatment (Table 1) [36, 37].
VKORC1 acts by converting the oxidized, inactive form of vitamin K to the active form and represents the pharmacological target of warfarin. Multiple common promoter polymorphisms influencing VKORC1 gene expression and dosing requirements include −1173T>C (rs9934438), −3730G>A (rs7294), and −1639G>A (rs9923231) [32, 38]. The −1639G>A variant reduces protein expression [39] which results in lower warfarin maintenance dose requirement compared to non-carriers (Table 1) [32]. Multiple rare non-synonymous variants in VKORC1 leading to warfarin resistance and higher dose requirements have also been identified [40].
Genome-wide association studies have identified variants in CYP4F2, a vitamin K(1) oxidase associated with reduced capacity to metabolize vitamin K1 and higher warfarin dose requirements[41], although findings have been inconsistent [42].
Clinical Application
Collectively, ~50-60 % of warfarin variability can be accounted for if patient related factors are evaluated in concert with genetic factors [43, 44]. In order to facilitate the clinical implementation of genetic information guidelines incorporating genetic information have been published [45••]. The pharmacogenetic interaction relating to warfarin has also been recognized by the FDA [46]. Ongoing clinical trials aimed to evaluate the benefit of incorporating genetic information into the warfarin dosing algorithms will provide more insight [47, 48•]. The extent to which genotype-guided therapy will be implemented in practice will also be influenced by uptake of newer oral anticoagulants such as dabigatran or rivaroxaban.
Statins
3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) inhibitors have in large clinical trials been shown to reduce the risk of cardiovascular events and are considered to be first-line therapy in conjunction with life-style changes for prevention of cardiovascular disease. Competitive HMGCR inhibitors, or statins, lower low-density lipoprotein cholesterol (LDLc) by attenuating endogenous cholesterol production, an early rate limiting step in cholesterol synthesis; they cause up-regulation of the liver LDLc receptor (LDLR) expression and increased LDLc clearance. However, the wide inter-individual variability in the extent of LDLc lowering by statins [49], attributable to environmental and genomic factors, means that some patients are at risk of cardiovascular events despite multiple dose adjustments [50]. In addition, concomitant drug therapy and genetic heterogeneity have been associated with statin-induced myopathy.
Responsiveness and Cardiovascular Events
The H7 haplotype of HMGCR (defined by three intronic SNPs: rs17244841, rs3846662, and rs17238540) has been associated with impaired reduction in LDLc lowering (5-20 %) following therapy with pravastatin [51] or simvastatin [52]. The attenuated LDLc response associated with the minor H7 haplotype has been attributed to an alternatively spliced HMGCR transcript that is less sensitive to simvastatin inhibition [53]. However, this finding has not been confirmed for all types of statins (Table 2) [54]. The H2 haplotype (defined by the intronic SNP rs3846662) and the LDLRL5 haplotype (defined by six SNPs within the LDLR3 un-translated region) have also been associated with attenuated LDLc response following statin treatment, particularly in blacks [52].
Genetic variation in apolipoprotein E (APOE) (two variants: rs429358 and rs7412 define the haplotypes ε2, ε3, and ε4) has previously been associated with variability in cholesterol levels across multiple populations [55] including the first statin response genome-wide association study (GWAS) [54]. Carriers of the ε2 haplotype have the greatest attenuation in lipid response followed by ε3 and ε4, respectively [56]. However, the validity of these findings been questioned by the lack of replication in a recent large meta-analysis (Table 2) [57].
The kinesin-like-protein 6 (KIF6) is involved in intracellular transport of key molecules including mRNA. Carriers of a missense SNP (Trp719Arg, rs20455) in KIF6 have been associated with increased risk of coronary events [58]. Interestingly, studies have found carriers of the risk allele to also have a greater benefit from statin therapy compared to non-carriers independent from LDLc, triglycerides and C-reactive protein [59, 60]. However, the association between KIF6 and cardiovascular disease was not replicated in a recent meta-analysis [61] which is in accordance with previous study findings that are unable to couple KIF6 to a differential statin response [62].
The drug pump P-glycoprotein serves to reduce bioavailability by promoting drug efflux from hepatocytes and enterocytes. The tri-allelic G2677T/A variant in ABCB1 (encoding the transporter) has been associated with impaired lowering of LDLc [63], whereas others have reported improved LDLc lowering [64].
The function and location of the ATP-binding cassette, subfamily G, member 2 (ABCG2), is similar to that of ABCB1. The common variant (rs2231142) reduces its transport capacity and rosuvastatin treated carriers of the A allele compared to non-carriers experience 5-7 % greater reduction in LDLc [65] although inconsistently [54].
Statin Induced Adverse Events
Genetic variation in the hepatic drug uptake transporter (polypeptide organic anion transporter P1B (OATP1B1)) encoded by SLCO1B1 is involved in the uptake of statins and has been coupled with altered risk of adverse drug reactions (ADRs). The predominant ADRs associated with statin therapy are myopathy and rhabdomyolysis (11.0 and 3.4 per 100,000 person-years, respectively) with the latter having a mortality rate of 10 % [66]. The pharmacokinetic properties of statin are altered via two common variants in SLCO1B1 (rs4149056 and rs2306283) that interferes with the transporter function [67] and hepatocyte localization of the transporter leading to higher plasma statin levels [68]. Findings from a GWAS saw a strong association between the common variant rs4363657 (rs4149056 and rs4363657 are in near-perfect disequilibrium, R2=0.97) and statin-induced myopathy in patients on high-dose simvastatin [69]. Notably, heterozygous carriers of the risk allele had odds ratio of 4.7 for developing myopathy per allele compared to non-carriers; the odds ratio was 17.4 in homozygotes (Table 1). Fifteen percent of the population were carriers of the risk allele (rs4149056) and 60 % of patients who developed myopathy were carriers of the risk allele [69]. These findings have subsequently been replicated [70].
Clinical Application
Using genetic information to determine the dose needed to acquire an adequate drop in LDLc is currently not implemented for several reasons. First, reasonably forecasting an expected LDLc drop is usually not a problem using only non-genetic patient information. Further, a smaller than expected LDLc reduction is not an acute problem and is often amenable to dose adjustments. Second, the genetic effect of genetic markers identified thus far is small. Third, variability in study designs and endpoints make interpretation of results difficult. Recent approaches modeling expected maximal statin effect on the basis of LDLc response to >1 dose may prove to be clinically useful [71].
Evidence of statin-induced ADRs has prompted CPIC Guidelines to generate guidelines with clinical recommendations for Simvastatin-Induced Myopathy (Table 1) [72••]. The extent to which these or other markers might also be useful for myopathy with other statins remains under study.
Beta-Blockers
Beta-adrenergic antagonists (β-blockers) are used for the management of cardiac arrhythmias, angina pectoris, myocardial infarction and hypertension. β -blockers competitively antagonize endogenous catecholamine at the beta-1-adrenergic receptor, encoded by ADBR1, and improve survival, remodeling and left ventricular ejection fraction after MI. However, inter-individual variability in response to β-blocker treatment has raised the question of genetic components influencing β-blocker response. Genes associated with inter-individual β-blocker response include CYP2D6 (pharmacokinetic; some agents), ADBR1, ADBR2, and GRK5 (pharmacodynamic).
Two common variants in ADBR1, resulting in Ser49Gly (rs1801252) and Arg389Gly (rs1801253), have previously been associated with impaired down-regulation [73], higher signal transduction [74], and altered biological function invitro [75]. Studies have shown that homozygous carriers of the Arg389 haplotype respond better to β-blocker treatment translating into improved left ventricular ejection fraction compared to carriers of the risk allele Gly389 [76-78], although such findings have not been consistent (Table 2) [79]. Similarly, reports of improved β-blocker response on blood pressure [80] and heart rate have also been inconsistent [81].
Two common variants in the beta-2-adrenergic receptor (ADBR2), resulting in Arg16Gly (rs1042713) and Gln27Glu (rs1042714), have been coupled with stimulation of adenylyl cyclase activity translating into increased agonistpromoted down-regulation of ADBR2 [82]. The majority of studies performed did not find the variants to be associated with improved clinical outcomes such as enhanced left ventricular ejection fraction [76, 83] although positive associations for Gln27Glu have been reported in a small study [84].
Many β-blockers, including propranolol, timolol, and metoprolol, are metabolized by CYP2D6, and loss of function variants (of which there are dozens) are very common. Poor metabolizers, 5-10 % of Caucasian and African subjects, carry two loss-of-function alleles, and generate unusually high plasma metoprolol concentrations with pronounced effects on blood pressure and heart rate lowering [85, 86] with the potential for ADRs (Table 2) [86]. In spite of only few studies providing evidence of a pharmacogenomic interaction, dose adjustments should be considered among patients prone to complications [87] which has also been acknowledged by the FDA [88]. Carvedilol is also a CYP2D6 substrate, but the poor metabolizer trait does not translate into variable clinical effects. Other β-blockers such as atenolol and nadolol do not require CYP2D6 to be metabolized.
A gain-of-function polymorphism (Glu41Leu, rs17098707) in the G protein-coupled receptor kinase 5 (GRK5) has been coupled with a beta-blocker-like phenotype, and appears to protect against catecholamine induced cardiomyopathy in mice [89]. The variant is more common among individuals of African descent and studies of metoprolol treated carriers of the Glu41Leu polymorphism experienced improved survival similar to untreated non-carriers (Leu41).
Variants in the α2c-adrenergic receptor (ADRA2C) have been associated with variable β-blocker responsiveness. A common four amino-acid deletion (Del322-325) has shown reduced ADRA2C activity in transfected cells [90] and has been associated with adverse outcomes in heart failure patients [91] although no effect was seen in the Beta-Blocker Evaluation of Survival Trial among patients on bucindolol (BEST) [78].
Clinical Implication
The findings of loss-of-function polymorphisms in CYP2D6 and the associated β-blocker response is equivocal and highlighted by the added FDA label. Thus, caution should be exerted among vulnerable heart failure patients who carry the loss-of-function in order to avoid ADRs. Moreover, there is some evidence of a pharmacogenomic interaction between β-blockers and the Arg389Gly polymorphism in ADBR1 although variable study findings make clinical interpretation difficult. Combining multiple risk alleles may provide more information [92].
Angiotensin-Converting Enzyme Inhibitors
Angiotensin-converting-enzyme inhibitors (ACE-I) are predominantly used to treat hypertension and have been associated with improved cardiovascular outcomes. A common insertion/deletion (I/D) polymorphism in the ACE gene (rs4646994) has been shown to influence ACE plasma concentrations [93]. An observational study reported that carriers of the DD genotype had a greater 10-year mortality risk compared to II carriers [94••]. However, these findings have not been replicated in prospective trials [95] or a large meta-analysis examining the influence of the variant on outcomes during ACE-I [96]. Thus, the proposed pharmacogenomic interaction between the I/D polymorphism in the ACE gene and ACE-I does not seem to be real and publication bias could have distorted any association [96]. Other candidates in the renin-angiotensin-aldosterone pathway, including the angiotensinogen (AGT) and angiotensin-II receptor types I and II (AGTR1 and AGTR2), have been interrogated and no definite pharmacogenomic associations have been identified.
The Perindopril Genetic Association study (PERGENE) sought to predict the treatment benefit of ACE-I and optimize ACE-I therapy by developing a genetic profile in 8907 CAD patients using 52 haplotype tagging polymorphisms across 12 genes [94••]. Two polymorphisms in the AGTR1 gene and one in the bradykinin type 1 receptor gene were significantly associated with improved treatment benefit during 4.2 years of follow-up (The primary composite endpoint was reduction in cardiovascular mortality, non-fatal myocardial infarction, and resuscitated cardiac arrest). Moreover, combining the three previously mentioned polymorphisms into a pharmacogenetic score produced a stepwise decrease in treatment benefit of perindopril with increasing scores [94••] which was replicated using a subset of patients from the Perindopril Protection Against Recurrent Stroke Study (PROGRESS) [95].
Clinical Implications
As of yet no definite pharmacogenomic targets have been identified for ACE-I responsiveness that merits routine genetic testing. However, the findings made in the PERGENE study could represent a true target if further validated.
Anti-Arrhythmic Drugs
Antiarrhythmic drugs that block the repolarizing potassium current IKr are among the commonest causes of a prolonged QT interval and are associated with increased risk of developing the malignant arrhythmia Torsade de Pointes (TdP). Importantly, the ability to produce a prolonged QT interval is not limited to cardiovascular drugs only [97]. Multiple polymorphisms in known long-QT genes have been reported among affected patients [98•]. Thus far, the evidence for causation is strongest for the D85N non-synonymous polymorphism (rs1805128) in KCNE1, encoding a potassium channel subunit; in a study of 176 cases of TdP, this variant conferred an odds ratio of 9.0 for developing TdP (Table 2) [99••].
Prioritized Pharmacogenomic Testing
While Clinical Laboratory Improvement Amendments (CLIA) approved laboratories do provide pharmacogenomic testing it is not always clear to clinicians which tests should be ordered, how to interpret the results, or if the results are even clinically actionable, particularly if test are ordered at the time of drug prescription. Given the rapid drop in genotyping costs, the CPIC approaches these issues from a different perspective, assuming that the data are already available from previous “preemptive” genotyping. CPIC provides a three-tier system (A, B, and C) to evaluate the level of evidence associated with a drug-gene interaction. “A” holds the strongest level of evidence and is assigned to pharmacogenetic signals where the “evidence includes consistent results from well-designed, well-conducted studies”. Pharmacogenomic signals that are assigned to “A” evidence level include: CYP2C19*2, CYP2C19*17, VKORC1: −1639G>A, CYP2C9*2, CYP2C9*3, and SLCO1B1: rs4149056. “B” is assigned where the evidence is sufficient to determine the effects, but the strength of the “evidence is limited by the number, quality, or consistency of the individual studies, by the inability to generalize to routine practice, or by the indirect nature of the evidence”. “C” is assigned where the evidence is insufficient to evaluate the effects on health and is not clinically actionable [100].
Conclusion
Fueled by the success of pharmacogenomic research the promise of tailoring diagnostic and treatment strategies to the needs of the individual patient may be within reach. Highlighting this exciting stage of rapid discovery is the development of clinical guidelines that incorporate genetic information to guide warfarin, clopidogrel, and statin therapy to reduce the risk of toxicity. Implementing these on a case by case and patient by patient basis is cumbersome and costly. With the plummeting cost of multiplexed genotyping, the idea of preemptively embedding DNA variant data in patient records, to be acted on when a target drug is prescribed, is beginning to be explored. Evaluation of the mechanics and healthcare outcomes in such systems, as well as ongoing clinical trials in pharmacogenomics and continuing genotype-phenotype association discovery, will determine how this personalized approach can be adopted.
Acknowledgment
Peter Weeke is funded by an unrestrictive grant from the Tryg Foundation (J.nr. 7343–09, TrygFonden, Denmark). Supported in part by U19 HL065962. Dan M. Roden has received grant support from NIH.
Clinical Trial Acronyms
- BEST
Beta-Blocker Evaluation of Survival Trial Among Patients on Bucindolol
- PERGENE
Perindopril Genetic Association Study
- PROGRESS
Perindopril Protection Against Recurrent Stroke Study
Footnotes
Compliance with Ethics Guidelines
Conflict of Interest Peter Weeke declares that he has no conflict of interest.
Dan M. Roden has received the following patents/royalties: U.S. Letters Patents No. 6456542, issued October 1, 2002 for ‘Method of Screening for Susceptibility to Drug-Induced Cardiac Arrhythmia.’
Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.
References
Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1.Breet NJ, van Werkum JW, Bouman HJ, et al. Comparison of platelet function tests in predicting clinical outcome in patients undergoing coronary stent implantation. JAMA. 2010;303(8):754–62. doi: 10.1001/jama.2010.181. [DOI] [PubMed] [Google Scholar]
- 2.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(1):92–9. doi: 10.1124/dmd.109.029132. [DOI] [PubMed] [Google Scholar]
- 3.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):2244–7. doi: 10.1182/blood-2006-04-013052. [DOI] [PubMed] [Google Scholar]
- 4.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):2429–36. doi: 10.1111/j.1538-7836.2007.02775.x. [DOI] [PubMed] [Google Scholar]
- 5.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 implantaion. Pharmacogenet Genomics. 2010;20(1):18–25. doi: 10.1097/FPC.0b013e328333dafe. [DOI] [PubMed] [Google Scholar]
- 6.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 bareetal stents. J Am Coll Cardiol. 2008;51(20):1925–34. doi: 10.1016/j.jacc.2007.12.056. [DOI] [PubMed] [Google Scholar]
- 7.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):849–57. doi: 10.1001/jama.2009.1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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(2):287–95. doi: 10.1038/clpt.2011.127. [DOI] [PubMed] [Google Scholar]
- 9••.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):328–32. doi: 10.1038/clpt.2011.132. Guidelines incorporating genetic variation in CYP2C19 on clopidogrel directed therapy.
- 10.Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 polymorphisms and response to clopidogrel. N Engl J Med. 2009;360(4):354–62. doi: 10.1056/NEJMoa0809171. [DOI] [PubMed] [Google Scholar]
- 11.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):309–17. doi: 10.1016/S0140-6736(08)61845-0. [DOI] [PubMed] [Google Scholar]
- 12.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):134–43. doi: 10.1016/j.jacc.2009.12.071. [DOI] [PubMed] [Google Scholar]
- 13••.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):1821–30. doi: 10.1001/jama.2010.1543. Carriers of the CYP2C19*2 risk allele who received clopidogrel post-PCI had a significantly increased risk of a major cardiovascuar event, most notably stent thrombosis.
- 14•.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(2):257–63. doi: 10.1038/clpt.2011.221. Confirmed findings on clopidogrel resistance in ABCB1 and CYP2C19 using a real-world population identified from electronic health records coupled with genetic information. No evidence of clopidogrel resistance was found for PON1.
- 15.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(9749):1320–8. doi: 10.1016/S0140-6736(10)61274-3. [DOI] [PubMed] [Google Scholar]
- 16.Pare G, Mehta SR, Yusuf S, et al. Effects of CYP2C19 genotype on outcomes of clopidogrel treatment. N Engl J Med. 2010;363(18):1704–14. doi: 10.1056/NEJMoa1008410. [DOI] [PubMed] [Google Scholar]
- 17.Ellis KJ, Stouffer GA, McLeod HL, Lee CR. Clopidogrel pharmacogenomics and risk of inadequate platelet inhibition: US FDA recommendations. Pharmacogenomics. 2009;10(11):1799–817. doi: 10.2217/pgs.09.143. [DOI] [PubMed] [Google Scholar]
- 18.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):512–8. doi: 10.1161/CIRCULATIONAHA.109.885194. [DOI] [PubMed] [Google Scholar]
- 19.Taubert D, von Beckerath N, Grimberg G, et al. Impact of P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther. 2006;80(5):486–501. doi: 10.1016/j.clpt.2006.07.007. [DOI] [PubMed] [Google Scholar]
- 20.Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopidogrel and cardiovascular events. N ngl J Med. 2009;360(4):363–75. doi: 10.1056/NEJMoa0808227. [DOI] [PubMed] [Google Scholar]
- 21••.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(9749):1312–9. doi: 10.1016/S0140-6736(10)61273-1. ABCB1 associated with adverse cardiovascular events independent of genetic variation in CYP2C19 among clopidogrel treated ACS patients.
- 22.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(3):506–12. doi: 10.1016/j.ahj.2010.06.039. [DOI] [PubMed] [Google Scholar]
- 23.Mega JL, Close SL, Wiviott SD, et al. Cytochrome P450 genetic polymorphisms and the response to prasugrel: relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Cirulation. 2009;119(19):2553–60. doi: 10.1161/CIRCULATIONAHA.109.851949. [DOI] [PubMed] [Google Scholar]
- 24.Rudez G, Bouman HJ, van Werkum JW, et al. Common variation in the platelet receptor P2RY12 gene is associated with residual on-clopidogrel platelet reactivity in patients undergoing elective percutaneous coronary interventions. Circ Cardiovasc Genet. 2009;2(5):515–21. doi: 10.1161/CIRCGENETICS.109.861799. [DOI] [PubMed] [Google Scholar]
- 25.Bouman HJ, Schomig E, van Werkum JW, et al. Paraoxonase-1 is a major determinant of clopidogrel efficacy. Nat Med. 2011;17(1):110–6. doi: 10.1038/nm.2281. [DOI] [PubMed] [Google Scholar]
- 26.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(13):1605–13. doi: 10.1093/eurheartj/ehr155. [DOI] [PubMed] [Google Scholar]
- 27.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):537–57. doi: 10.1161/CIR.0b013e3181ee08ed. [DOI] [PubMed] [Google Scholar]
- 28•.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. 2012;92(1):87–95. doi: 10.1038/clpt.2011.371. Description of the Vanderbilt PREDICT project aimed at implementing and evaluating strategies for personlized medicine.
- 29.Rost S, Fregin A, Ivaskevicius V, et al. Mutations in VKORC1 cause warfarin resistance and multiple coagulation factor deficiency type 2. Nature. 2004;427(6974):537–41. doi: 10.1038/nature02214. [DOI] [PubMed] [Google Scholar]
- 30.Schalekamp T, de Boer A. Pharmacogenetics of oral anticoaguant therapy. Curr Pharm Des. 2010;16(2):187–203. doi: 10.2174/138161210790112737. [DOI] [PubMed] [Google Scholar]
- 31.Higashi MK, Veenstra DL, Kondo LM, et al. Association between CYP2C9 genetic variants and anticoagulation-related outomes during warfarin therapy. JAMA. 2002;287(13):1690–8. doi: 10.1001/jama.287.13.1690. [DOI] [PubMed] [Google Scholar]
- 32.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):2285–93. doi: 10.1056/NEJMoa044503. [DOI] [PubMed] [Google Scholar]
- 33.Shehab N, Sperling LS, Kegler SR, Budnitz DS. National estimates of emergency department visits for hemorrhage-related adverse events from clopidogrel plus aspirin and from warfarin. Arch Intern Med. 2010;170(21):1926–33. doi: 10.1001/archinternmed.2010.407. [DOI] [PubMed] [Google Scholar]
- 34.Lee CR, Goldstein JA, Pieper JA. Cytochrome P450 2C9 polymorphisms: a comprehensive review of the in-vitro and human data. Pharmacogenetics. 2002;12(3):251–63. doi: 10.1097/00008571-200204000-00010. [DOI] [PubMed] [Google Scholar]
- 35.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(4):365–75. doi: 10.1007/s00228-008-0584-5. [DOI] [PubMed] [Google Scholar]
- 36.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]
- 37.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):717–9. doi: 10.1016/S0140-6736(98)04474-2. [DOI] [PubMed] [Google Scholar]
- 38.Yang L, Ge W, Yu F, Zhu H. Impact of VKORC1 gene polymorphism on interindividual and interethnic warfarin dosage requirement–a systematic review and meta analysis. Thromb Res. 2010;125(4):e159–66. doi: 10.1016/j.thromres.2009.10.017. [DOI] [PubMed] [Google Scholar]
- 39.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):1013–21. doi: 10.1182/blood-2008-03-144899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Scott SA, Edelmann L, Kornreich R, Desnick RJ. Warfarin pharmacogenetics: CYP2C9 and VKORC1 genotypes predict different sensitivity and resistance frequencies in the Ashkenazi and Sephardi Jewish populations. Am J Hum Genet. 2008;82(2):495–500. doi: 10.1016/j.ajhg.2007.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.McDonald MG, Rieder MJ, Nakano M, Hsia CK, Rettie AE. CYP4F2 is a vitamin K1 oxidase: an explanation for altered warfarin dose in carriers of the V433M variant. Mol Pharmacol. 2009;75(6):1337–46. doi: 10.1124/mol.109.054833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.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]
- 43.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):326–31. doi: 10.1038/clpt.2008.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wadelius M, Chen LY, Lindh JD, et al. The largest prospective warfarin-treated cohort supports genetic forecasting. Blood. 2009;113(4):784–92. doi: 10.1182/blood-2008-04-149070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45••.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):625–9. doi: 10.1038/clpt.2011.185. Guidelines incorporating genetic variation in CYP2C9 and VKORC1 in warfarin directed therapy.
- 46.Warfarin label information [Updated January 22, 2010].
- 47.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(10):1687–95. doi: 10.2217/pgs.09.125. [DOI] [PubMed] [Google Scholar]
- 48•.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. Study utilizing genetic information in each individual to guide warfarin therapy.
- 49.Simon JA, Lin F, Hulley SB, et al. Phenotypic predictors of response to simvastatin therapy among African-Americans and Caucasians: the Cholesterol and Pharmacogenetics (CAP) Study. Am J Cardiol. 2006;97(6):843–50. doi: 10.1016/j.amjcard.2005.09.134. [DOI] [PubMed] [Google Scholar]
- 50.Mangravite LM, Thorn CF, Krauss RM. Clinical implications of pharmacogenomics of statin treatment. Pharmacogenomics J. 2006;6(6):360–74. doi: 10.1038/sj.tpj.6500384. [DOI] [PubMed] [Google Scholar]
- 51.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):2821–7. doi: 10.1001/jama.291.23.2821. [DOI] [PubMed] [Google Scholar]
- 52.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):1537–44. doi: 10.1161/CIRCULATIONAHA.107.708388. [DOI] [PubMed] [Google Scholar]
- 53.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):355–62. doi: 10.1161/CIRCULATIONAHA.108.773267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Thompson JF, Hyde CL, Wood LS, et al. Comprehensive whole-genome and candidate gene analysis for response to statin therapy in the Treating to New Targets (TNT) cohort. Circ Cardiovasc Genet. 2009;2(2):173–81. doi: 10.1161/CIRCGENETICS.108.818062. [DOI] [PubMed] [Google Scholar]
- 55.Utermann G. Apolipoprotein E polymorphism in health and disease. Am Heart J. 1987;113(2 Pt 2):433–40. doi: 10.1016/0002-8703(87)90610-7. [DOI] [PubMed] [Google Scholar]
- 56.Voora D, Shah SH, Reed CR, et al. Pharmacogenetic predictors of statin-mediated low-density lipoprotein cholesterol reduction and dose response. Circ Cardiovasc Genet. 2008;1(2):100–6. doi: 10.1161/CIRCGENETICS.108.795013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zintzaras E, Kitsios GD, Triposkiadis F, Lau J, Raman G. APOE gene polymorphisms and response to statin therapy. Pharm J. 2009;9(4):248–57. doi: 10.1038/tpj.2009.25. [DOI] [PubMed] [Google Scholar]
- 58.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(4):455–61. doi: 10.1097/HJR.0b013e328336a0dd. [DOI] [PubMed] [Google Scholar]
- 59.Iakoubova OA, Sabatine MS, Rowland CM, et al. Polymorphism in KIF6 gene and benefit from statins after acute coronary syndromes: results from the PROVE IT-TIMI 22 study. J Am Coll Cardiol. 2008;51(4):449–55. doi: 10.1016/j.jacc.2007.10.017. [DOI] [PubMed] [Google Scholar]
- 60.Iakoubova OA, Tong CH, Rowland CM, et al. Association of the Trp719Arg polymorphism in kinesin-like protein 6 with myocardial infarction and coronary heart disease in 2 prospective trials: the CARE and WOSCOPS trials. J Am Coll Cardiol. 2008;51(4):435–43. doi: 10.1016/j.jacc.2007.05.057. [DOI] [PubMed] [Google Scholar]
- 61.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(19):1552–63. doi: 10.1016/j.jacc.2010.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ridker PM, MacFadyen JG, Glynn RJ, Chasman DI. Kinesin-like protein 6 (KIF6) polymorphism and the efficacy of rosuvastatin in primary prevention. Circ Cardiovasc Genet. 2011;4(3):312–7. doi: 10.1161/CIRCGENETICS.110.959353. [DOI] [PubMed] [Google Scholar]
- 63.Mega JL, Morrow DA, Brown A, Cannon CP, Sabatine MS. Identification of genetic variants associated with response to statin therapy. Arterioscler Thromb Vasc Biol. 2009;29(9):1310–5. doi: 10.1161/ATVBAHA.109.188474. [DOI] [PubMed] [Google Scholar]
- 64.Kajinami K, Brousseau ME, Ordovas JM, Schaefer EJ. Polymorphisms in the multidrug resistance-1 (MDR1) gene influence the response to atorvastatin treatment in a gender-specific manner. Am J Cardiol. 2004;93(8):1046–50. doi: 10.1016/j.amjcard.2004.01.014. [DOI] [PubMed] [Google Scholar]
- 65.Kondo C, Suzuki H, Itoda M, et al. Functional analysis of SNPs variants of BCRP/ABCG2. Pharm Res. 2004;21(10):1895–903. doi: 10.1023/b:pham.0000045245.21637.d4. [DOI] [PubMed] [Google Scholar]
- 66.Law M, Rudnicka AR. Statin safety: a systematic review. Am J Cardiol. 2006;97(8A):52C–60C. doi: 10.1016/j.amjcard.2005.12.010. [DOI] [PubMed] [Google Scholar]
- 67.Tirona RG, Leake BF, Merino G, Kim RB. Polymorphisms in OATP-C: identification of multiple allelic variants associated with altered transport activity among European- and African-Americans. J Biol Chem. 2001;276(38):35669–75. doi: 10.1074/jbc.M103792200. [DOI] [PubMed] [Google Scholar]
- 68.Pasanen MK, Neuvonen M, Neuvonen PJ, Niemi M. SLCO1B1 polymorphism markedly affects the pharmacokinetics of simvatatin acid. Pharmacogenet Genomics. 2006;16(12):873–9. doi: 10.1097/01.fpc.0000230416.82349.90. [DOI] [PubMed] [Google Scholar]
- 69.Link E, Parish S, Armitage J, et al. SLCO1B1 variants and statin-induced myopathy-a genomewide study. N Engl J Med. 2008;359(8):789–99. doi: 10.1056/NEJMoa0801936. [DOI] [PubMed] [Google Scholar]
- 70.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):1609–16. doi: 10.1016/j.jacc.2009.04.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wilke RA, Berg RL, Linneman JG, Zhao C, McCarty CA, Krauss RM. Characterization of low-density lipoprotein cholesterol-lowering efficacy for atorvastatin in a population-based DNA biorepository. Basic Clin Pharmacol Toxicol. 2008;103(4):354–9. doi: 10.1111/j.1742-7843.2008.00291.x. [DOI] [PubMed] [Google Scholar]
- 72••.Wilke RA, Ramsey LB, Johnson SG, et al. The clinical pharmacogenomics implementation consortium: CPIC guideline for SLCO1B1 and simvastatin-induced myopathy. Clin Pharmacol Ther. 2012;92(1):112–7. doi: 10.1038/clpt.2012.57. Guidelines incorporating genetic variation in SLCO1B1 to reduce statin related myopathy.
- 73.Rathz DA, Brown KM, Kramer LA, Liggett SB. Amino acid 49 polymorphisms of the human beta1-adrenergic receptor affect agonist-promoted trafficking. J Cardiovasc Pharmacol. 2002;39(2):155–60. doi: 10.1097/00005344-200202000-00001. [DOI] [PubMed] [Google Scholar]
- 74.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(18):12670–4. doi: 10.1074/jbc.274.18.12670. [DOI] [PubMed] [Google Scholar]
- 75.Brodde OE. Beta1- and beta2-adrenoceptor polymorphisms and cardiovascular diseases. Fundam Clin Pharmacol. 2008;22(2):107–25. doi: 10.1111/j.1472-8206.2007.00557.x. [DOI] [PubMed] [Google Scholar]
- 76.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(11):941–9. doi: 10.1097/FPC.0b013e3282ef7354. [DOI] [PubMed] [Google Scholar]
- 77.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):277–82. doi: 10.1097/FPC.0b013e3280105245. [DOI] [PubMed] [Google Scholar]
- 78.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):21–8. doi: 10.1161/CIRCHEARTFAILURE.109.885962. [DOI] [PubMed] [Google Scholar]
- 79.White HL, de Boer RA, Maqbool A, et al. An evaluation of the beta-1 adrenergic receptor Arg389Gly polymorphism in individuals with heart failure: a MERIT-HF sub-study. Eur J Heart Fail. 2003;5(4):463–8. doi: 10.1016/s1388-9842(03)00044-8. [DOI] [PubMed] [Google Scholar]
- 80.Liu J, Liu ZQ, Yu BN, et al. beta1-Adrenergic receptor polymorphisms influence the response to metoprolol monotherapy in patients with essential hypertension. Clin Pharmacol Ther. 2006;80(1):23–32. doi: 10.1016/j.clpt.2006.03.004. [DOI] [PubMed] [Google Scholar]
- 81.Karlsson J, Lind L, Hallberg P, et al. Beta1-adrenergic receptor gene polymorphisms and response to beta1-adrenergic receptor blockade in patients with essential hypertension. Clin Cardiol. 2004;27(6):347–50. doi: 10.1002/clc.4960270610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.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(32):9414–9. doi: 10.1021/bi00198a006. [DOI] [PubMed] [Google Scholar]
- 83.de Groote P, Helbecque N, Lamblin N, et al. Association between beta-1 and beta-2 adrenergic receptor gene polymorphisms and the response to beta-blockade in patients with stable congestive heart failure. Pharmacogenet Genomics. 2005;15(3):137–42. doi: 10.1097/01213011-200503000-00001. [DOI] [PubMed] [Google Scholar]
- 84.Troncoso R, Moraga F, Chiong M, et al. Gln(27)->Glubeta(2)-adrenergic receptor polymorphism in heart failure patients: differential clinical and oxidative response to carvedilol. Basic Clin Pharmacol Toxicol. 2009;104(5):374–8. doi: 10.1111/j.1742-7843.2008.00370.x. [DOI] [PubMed] [Google Scholar]
- 85.Rau T, Wuttke H, Michels LM, et al. Impact of the CYP2D6 genotype on the clinical effects of metoprolol: a prospective longitudinal study. Clin Pharmacol Ther. 2009;85(3):269–72. doi: 10.1038/clpt.2008.218. [DOI] [PubMed] [Google Scholar]
- 86.Bijl MJ, Visser LE, van Schaik RH, et al. Genetic variation in the CYP2D6 gene is associated with a lower heart rate and blood pressure in beta-blocker users. Clin Pharmacol Ther. 2009;85(1):45–50. doi: 10.1038/clpt.2008.172. [DOI] [PubMed] [Google Scholar]
- 87.Swen JJ, Wilting I, de Goede AL, et al. Pharmacogenetics: from bench to byte. Clin Pharmacol Ther. 2008;83(5):781–7. doi: 10.1038/sj.clpt.6100507. [DOI] [PubMed] [Google Scholar]
- 88.FDA Table of pharmacogenomic biomarkers in drug labels. 2012 http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm236819.htm.
- 89.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):510–7. doi: 10.1038/nm1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Small KM, Forbes SL, Rahman FF, Bridges KM, Liggett SB. 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(30):23059–64. doi: 10.1074/jbc.M000796200. [DOI] [PubMed] [Google Scholar]
- 91.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]
- 92.Petersen M, Andersen JT, Hjelvang BR, et al. Association of beta-adrenergic receptor polymorphisms and mortality in carvedilol-treated chronic heart-failure patients. Br J Clin Pharmacol. 2011;71(4):556–65. doi: 10.1111/j.1365-2125.2010.03868.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.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(1):197–205. [PMC free article] [PubMed] [Google Scholar]
- 94••.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(15):1854–64. doi: 10.1093/eurheartj/ehq169. First study to identify genetic determinants influencing ACE-I therapy.
- 95.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(3):297–303. doi: 10.1161/01.HYP.0000088322.85804.96. [DOI] [PubMed] [Google Scholar]
- 96.Agema WR, Jukema JW, Zwinderman AH, van der Wall EE. A meta-analysis of the angiotensin-converting enzyme gene polymorphism and restenosis after percutaneous transluminal coronary revascularization: evidence for publication bias. Am Heart J. 2002;144(5):760–8. doi: 10.1067/mhj.2002.125509. [DOI] [PubMed] [Google Scholar]
- 97.Roden DM. Drug-induced prolongation of the QT interval. N Engl J Med. 2004;350(10):1013–22. doi: 10.1056/NEJMra032426. [DOI] [PubMed] [Google Scholar]
- 98•.Ramirez AH, Shaffer CM, Delaney JT, et al. Novel rare variants in congenital cardiac arrhythmia genes are frequent in drug-induced torsades de pointes. Pharmacogenomics J. 2012 doi: 10.1038/tpj.2012.14. doi:10.1038/tpj.2012.14. Directed genotyping demonstrate how patients developing TdP frequently have rare mutations in genes known to associate cardiac arrhythmia.
- 99••.Kaab S, Crawford DC, Sinner MF, et al. A large candidate gene survey identifies the KCNE1 D85N polymorphism as a possible modulator of drug-induced torsades de pointes. Circ Cardiovasc Genet. 2012;5(1):91–9. doi: 10.1161/CIRCGENETICS.111.960930. Polymorphism (D85N) in important potassium channel (KCNE1) associated with TdP.
- 100.Relling MV, Klein TE. CPIC: clinical pharmacogenetics implementation consortium of the pharmacogenomics research network. Clin Pharmacol Ther. 2011;89(3):464–7. doi: 10.1038/clpt.2010.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
