Abstract
Cardiovascular pharmacogenomics is the study and identification of genomic markers that are associated with variability in cardiovascular drug response, cardiovascular drug related outcomes, or cardiovascular drug related adverse events. This overview presents an introduction and historical background to cardiovascular pharmacogenomics, and a protocol for designing a cardiovascular pharmacogenomics study. Important considerations are also included for constructing a cardiovascular pharmacogenomics phenotype, designing the replication or validation strategy, common statistical approaches, and how to put the results in context with the cardiovascular drug or cardiovascular disease under investigation.
Basic Protocol 1: Designing a Cardiovascular Pharmacogenomics Study
Keywords: Pharmacogenomics, Cardiovascular, Complex diseases, Antiplatelet therapy, Antihypertensive therapy
INTRODUCTION:
Pharmacogenomics (PGx) aims to identify genomic markers, most commonly single nucleotide polymorphisms (SNPs), that are associated with variability in drug response, drug related outcomes, or adverse events (Meyer 2004, Wang, et al. 2011). Cardiovascular disease has been one therapeutic area that has seen many high impact PGx associations (Gage, et al. 2008, Mega, et al. 2010, Shuldiner, et al. 2009, Turner, et al. 2013a, Wilke, et al. 2012). However, the translation of these findings into clinical practice has not been straightforward (Cavallari, et al. 2017, Luzum, et al. 2017, Manolio, et al. 2015, Roden, et al. 2018). The purpose of this article is to give a general overview and methodology for designing a cardiovascular PGx study.
The first studies in cardiovascular PGx were focused on candidate genes known to be involved in the pharmacokinetics (PK) and pharmacodynamics (PD) of the drug. PK is often referred to as the study of what the body does to the drug, and in terms of PGx, relates to studying the variation in genes that encode drug metabolism enzymes, or cytochrome P450 enzymes (CYP enzymes), that metabolize the drug being studied (Meyer 2004). PD is often referred to as the study of what the drug does to the body, and in terms of PGx, focuses on studying variation in genes that encode drug targets (Meyer 2004). Finally, the study of variation in genes that encode drug transporters can impact both PK and PD. Taken together, genetic variability in the genes that code for drug metabolizing enzymes, drug transporters, and drug targets can impact the variability in the PK and PD of a drug, which impacts variability in the efficacy (response) and toxicity (adverse outcomes) of a drug.
While some candidate gene studies were successful in cardiovascular PGx (Rettie, et al. 1994, Rieder, et al. 2005), there were also candidate genes that failed to replicate (Arnett, et al. 2005, Maitland-van der Zee, et al. 2011, Schelleman, et al. 2008), and additional unexplained variability in cardiovascular drug response. Through genome-wide association studies (GWAS), cardiovascular PGx candidate genes were confirmed (Cooper, et al. 2008, Takeuchi, et al. 2009), and novel associations were identified (Link, et al. 2008, Shuldiner, et al. 2009). Currently, the landscape in cardiovascular PGx stretches from candidate gene studies to GWAS to GWAS meta-analyses to studies utilizing other ‘omic technologies (e.g., RNAseq, proteomics, metabolomics) to implementation. While there are some cardiovascular drug-gene pairs being implemented clinically (Cavallari, et al. 2017, Luzum, et al. 2017, Weitzel, et al. 2016), there still remains much additional discovery work to be done.
A detailed protocol for designing a cardiovascular PGx study is described below. The factors to consider during the study design process are also provided. Additional information on example cardiovascular PGx applications, and common analysis methods are also given below.
STRATEGIC PLANNING
Designing a cardiovascular PGx study is a multi-stage process. While considering the many different steps, and the pros and cons in study design, the two most important factors to consider are the trait or disease state, and the drug response. Prior to designing the entire cardiovascular PGx study, additional time and planning should be spent on the phenotyping methods that will be used to determine these two factors.
-
1The Trait or Disease State. The definition of the trait or disease state is one half of the cardiovascular PGx phenotype. When determining what the overall cardiovascular PGx phenotype will be, there are considerations that should be taken for the trait or disease state. Some common questions to ask include:
- How will the disease be reported (e.g., patient reported versus clinically confirmed)?
- Are there ways to clinically confirm the disease or trait under study (e.g., vitals, laboratory tests, clinical procedures, diagnoses, physician examination)?
- Is the drug currently, or has it been previously, approved for multiple indications or used off-label (e.g., the use of clopidogrel post stroke versus post percutaneous coronary intervention)?
- What stage of the disease state would be most informative to study (e.g., mild to moderate hypertension versus severe hypertension)?
- Do patients need to be newly diagnosed? Is the trait something that is measured routinely in healthy patients?
- Can patients have other comorbidities? What are the influences on the disease or trait by age, sex, and ancestry?
- Are there environmental influences on the disease or trait that need to be considered?
If the investigator designing the cardiovascular PGx study is not an expert in the trait or disease state being considered, it is recommended they identify a collaborator with this expertise.
-
2The Drug Response. The definition of the drug response is the other half of the cardiovascular PGx phenotype. The most common drug responses are either an efficacy outcome or a safety outcome. When investigating these outcomes, some common questions to consider are:
- Does a patient need to be drug naïve or washed-out from drug before treatment?
- How is the drug response calculated (e.g., blood pressure response = blood pressure post-treatment – blood pressure pre-treatment)?
- How many response phenotypes need to be studied (e.g., systolic blood pressure and diastolic blood pressure)?
- Does medication dose information need to be collected?
- Does the drug need to be titrated for maximum efficacy?
- What is the half-life of the drug?
- Are there known drug-drug interactions with the drug under study?
- Are there known environmental-drug interactions with the drug under study?
- Can patients be treated with other drugs during the study?
- How are cardiovascular outcomes reported?
- How are cardiovascular outcomes adjudicated?
- Do laboratory values need to be collected?
- Do patients need to be fasting when laboratory values are collected?
BASIC PROTOCOL 1
DESIGNING A CARDIOVASCULAR PHARMACOGENOMICS STUDY
Selecting the cardiovascular PGx phenotype to be studied is the main factor in designing a cardiovascular PGx study. However, there are many additional considerations, and factors that may influence how the cardiovascular PGx phenotype is determined. This protocol describes the procedures for designing a cardiovascular PGx study. A summary of the elements to consider during the study design process is provided in Table 1.
Table 1.
Study Design Element | Consideration for PGx Study |
---|---|
Phenotype | 1. Cardiovascular trait or disease 2. Cardiovascular drug 3. Drug response |
Type of Study | 1. Existing Data or New Study 2. Retrospective or Prospective 3. Observational or Clinical Trial 4. Candidate gene or GWAS |
DNA | 1. Existing DNA samples 2. Need to collect DNA samples |
Study Population | 1. Race and ancestry populations 2. Other comorbidities 3. Other exposures |
Power | 1. Sample size 2. Effect size 3. Minor allele frequency |
Replication | 1. Direct replication 2. Validation in similar drug 3. Validation in similar disease state 4. Validation in another race group |
Statistical Analysis | 1. Logistic regression 2. Linear regression 3. Interaction analysis 4. Single study or Meta-analysis |
PGx: Pharmacogenomics
GWAS: Genome-wide association study
DNA: Deoxyribonucleic acid
Materials
- Cardiovascular Pharmacogenomic Phenotype.
- The phenotype determination is the first step of the protocol; however, it helps to have an idea of some possible phenotypes before designing the study. Examples of cardiovascular PGx phenotypes are shown in Table 2.
- Existing SNP or GWAS data OR a DNA collection and genotyping plan.
- To conduct a cardiovascular PGx study, there must be genetic data of some kind.
- Demographics and Characteristics of the Study Population.
- This may be existing data or data that needs to be collected.
-
Software for a Power Calculation
Note: Examples of programs that can be used are listed in Table 3.
-
Software for Analyses.
Note: Examples of common programs are listed in Table 3.
- Databases used:
- For published literature: PubMed (https://pubmed.ncbi.nlm.nih.gov/)
- For pharmacogenomic knowledge: PharmGKB (https://www.pharmgkb.org/)
- For clinical pharmacogenomic guidelines: CPIC (https://cpicpgx.org/)
- For drugs with biomarker information in the label (FDA): FDA Biomarker List (https://www.fda.gov/drugs/science-and-research-drugs/table-pharmacogenomic-biomarkers-drug-labeling)
Table 2.
Cardiovascular PGx Phenotype | Trait or Disease State | Cardiovascular Drug | Drug Response |
---|---|---|---|
Blood Pressure Response to Thiazide Diuretics | Change in Blood Pressure in Hypertension patients | Thiazide Diuretic | Post-treatment Blood Pressure - Pre-Treatment Blood Pressure |
Bleeding Outcomes after treatment with Warfarin | Atrial Fibrillation | Warfarin | Bleeding events after treatment with Warfarin |
Myopathy after treatment with Simvastatin | Hypercholesterolemia | Simvastatin | Myopathy after treatment with Simvastatin |
Cardiovascular Outcomes after treatment with Clopidogrel | Percutaneous Coronary Intervention | Clopidogrel | Cardiovascular Outcomes after treatment with Clopidogrel |
Elevated Fasting Glucose after treatment with Thiazide Diuretics | Hypertension | Thiazide Diuretic | Post-treatment Fasting Glucose - Pre-treatment Fasting Glucose |
Cardiovascular Outcomes after treatment with Calcium Channel Blockers | Hypertension | Calcium Channel Blocker | Difference in Cardiovascular Outcomes in patients treated with Calcium Channel Blockers compared to those not treated with Calcium Channel Blockers |
PGx: Pharmacogenomics
Table 3.
Program | Input Data Type | Functions | Remarks | Website |
---|---|---|---|---|
PLINK | Directly Typed genotypes and Imputed genotypes | Linear Regression Logistic Regression Interaction Term |
Allows for data management, Principal Component Analysis, Linkage Disequilibrium calculations, Genotypic means for quantitative traits | https://www.cog-genomics.org/plink/2.0/ |
ProbABEL | Imputed genotypes | Linear Regression Logistic Regression Interaction Term |
Allows for Cox proportional hazards models | https://github.com/GenABEL-Project/ProbABEL |
EPACTS | Sequence-based genotypes from sequencing or imputation | Linear Regression Logistic Regression EMMAX |
Allows single variant and gene-wise or group-wise tests | https://genome.sph.umich.edu/wiki/EPACTS |
METAL | Summary results from GWAS or top SNP signals | Meta-analysis | Allows for both inverse-variance weighting and weighting by sample size Includes options for genomic control correction |
https://genome.sph.umich.edu/wiki/METAL_Documentation |
SAS | Single SNP genotypes Demographics and Characteristics |
Linear Regression Logistic Regression Interaction Term Descriptive Statistics |
Used for PGx analyses by genotype or by drug for top SNPs | https://www.sas.com/en_us/home.html |
R | Directly Typed genotypes and Imputed genotypes Single SNP genotypes Demographics and Characteristics |
Linear Regression Logistic Regression Interaction Term Descriptive Statistics |
Additional R Packages for GWAS analyses Used for PGx analyses by genotype or by drug for top SNPs |
https://www.r-project.org/ |
Quanto | Study Characteristics | Power Calculation | Windows application | https://preventivemedicine.usc.edu/download-quanto/ |
G*Power | Study Characteristics | Power Calculation | Mac and Windows applications | https://stats.idre.ucla.edu/other/gpower/ |
Protocols steps and annotations
-
1
Select the cardiovascular pharmacogenomic phenotype.
A cardiovascular PGx phenotype includes a cardiovascular trait or disease, a cardiovascular drug, and a drug response. The phenotype can be dichotomous or continuous, where the coded phenotype is often the drug response. The PGx element of the phenotype can also be incorporated through an interaction analysis, by adding a SNPxDrug interaction term to the regression model. Examples of cardiovascular PGx phenotypes are shown in Table 2.
Clearly define the cardiovascular trait or disease state by diagnosis codes, prescription medications, or laboratory values. Construct a list of inclusion criteria and exclusion criteria for how the trait or disease state will be defined.
Determine what cardiovascular drug will be studied. Determine if an entire drug class will be included, or if a specific drug is being studied. Determine if a specific formulation is required, or all brand name and generics will be included.
- Define how the drug response will be measured or collected. Determine when the drug response will be measured or collected. Determine if the study participants need to be drug naïve or washed out from the drug before the study.
-
2Determine the type of study design.
-
2
There are factors in the study design that determine how data will be collected, how study participants will be recruited, and the number of SNPs or genes being studied.
Determine if the study will use existing data or is a new study. There are many cardiovascular PGx studies that are sub-studies of larger cardiovascular randomized controlled trials (RCTs) or other cardiovascular clinical trials. These studies have taken advantage of the larger design of the clinical trial, while also collecting DNA and any additional measures to construct the cardiovascular PGx phenotype under investigation. Other prior studies in cardiovascular PGx have been smaller, specifically designed PGx clinical trials, or prospective, observational studies. Additionally, prior cardiovascular PGx studies have been designed using longitudinal cohort studies, and biobanks that have collected DNA. By using an existing data source, often all data elements have already been collected, and DNA has also been collected. However, the phenotype needs to be designed considering what variables were collected and at what time points. By designing a new study, there is more flexibility in what variables will be collected and when they will be collected, but there is much more time and money needed for the overall study.
Determine the timeframe of the study. Will the study be retrospective, taking a timepoint to select study participants and looking back into time? Or will the study be prospective, recruiting study participants and then moving forward into time? Additionally, the study could be cross-sectional with information just at a single timepoint. However, often cardiovascular PGx are more difficult to design in a cross-sectional manner, as it is much harder to construct a drug-response phenotype from data from a single timepoint.
Determine if the study will be observational or a clinical trial. Do study participants need to be randomized to drug, or receive a specific intervention? Or can data be collected on drug response by observing study participants who are already, or were already, prescribed the drug being studied?
- Determine the number of SNPs or genes that will be studied. PGx candidate gene or candidate SNP studies are based on prior knowledge or the PK and PD of the drug. Whereas a GWAS interrogates SNPs across the genome based off of a genome-wide SNP panel.
-
3Determine how DNA will be collected, or if DNA has already been collected.
-
3
In study designs that are using existing data or retrospective, often DNA has already been collected, and in many studies, DNA has already been isolated and GWAS data may already exist. In study designs that are prospective or new studies, the collection of DNA must be included in the study design.
-
4
Consider the study population.
The study population often includes study participants from different race and ancestry backgrounds, study participants with other comorbidities, and study participants with other exposures.
Establish how race and ancestry will be determined and accounted for in the study. While many of the well validated PGx variants are important in all race and ancestry backgrounds, during the discovery phase it is important to conduct analyses separately in each race and ancestry group due to the differences in linkage disequilibrium across the genome.
Collect information on any additional comorbidities, traits, demographics, or vitals that may influence the cardiovascular PGx phenotype. These variables could be included as covariates in the statistical model(s). Additionally, these variables could be used for sensitivity analyses or sub-group analyses.
- Determine if there are other exposures that may influence the cardiovascular PGx phenotype. Is the drug under study metabolized by a CYP enzyme that has known inducers or inhibitors? Can smoking or caffeine influence the metabolism of the drug being studied? These additional variables should also be collected.
-
5Conduct a power calculation.
-
5
A power calculation is necessary when using an existing dataset to determine what effect size can be detected at different minor allele frequencies. A power calculation will also determine the ideal sample size for a new study.
Determine the sample size. If the study is using existing data, then the sample size is known. If the study is a new study, then use a power calculation to determine the sample size.
Determine the effect size. The effect size can be measured as an Odds Ratio in a case-control study design, or as a β-coefficient if the study is examining a continuous PGx phenotypes. Use prior validated or replicated associations in the literature to gauge what effect size is reasonable to detect. It has been shown that PGx variants have larger effect sizes when compared to other complex traits (Maranville and Cox 2016).
- Select the minor allele frequency. If the study is a candidate gene or candidate SNP study, then the minor allele frequencies of the SNPs is known. If the study is a GWAS, then examine power across minor allele frequencies from 5% to 45%.
-
6Determine a replication plan.
-
6
Replication can be more difficult in PGx studies. It is often difficult to find a study with the exact same study design and phenotyping. In cardiovascular PGx studies, there are often four types of replications or validations that are used.
Directly replicate the top SNPs examining the same PGx phenotype with the same drug in the same population.
Validate the top SNPs in a similar drug or a drug in the same drug class, with the same PGx phenotype and the same population.
Validate the top SNPs in a similar disease state, or related PGx phenotype, with the same drug and in the same population.
- Validate the top SNPs in another race or ancestry group, with the same PGx phenotype and the same drug.
-
7Determine the analysis plan.
-
7
The statistical analysis plan directly relates to the type of cardiovascular PGx phenotype, and the data sources for the study. Common analyses include logistic regression models in case-control based PGx studies, and linear regression models in continuous response or dose based PGx studies. Interaction analyses are also used to examine a phenotype or outcome by a certain drug treatment, adding a SNPxdrug interaction term into the regression model. Finally, there are both single study cardiovascular PGx designs, as well as meta-analysis cardiovascular PGx designs.
COMMENTARY
BACKGROUND INFORMATION
There have been many successful discoveries through cardiovascular PGx studies in multiple therapeutic areas. Several studies have further elucidated the PK/PD of the drug being investigated, by identifying significant associations with SNPs in and near genes that encode for drug metabolism enzymes, drug transporters, and drug targets. Further examination of these studies highlights important elements in cardiovascular PGx study design.
One of the first cardiovascular therapeutic areas that saw success through both candidate gene approaches and GWAS approaches was oral anticoagulants, specifically, the drug warfarin. Through multiple candidate gene studies SNPs in and near CYP2C9 and VKORC1 were associated with stable warfarin dose (D’Andrea, et al. 2005, Rettie, et al. 1994, Rieder, et al. 2005, Thijssen, et al. 2001). Next, the association between a SNP in CYP4F2 and stable warfarin dose was identified using a drug metabolism and drug transporter SNP panel (Caldwell, et al. 2008). These associations were further confirmed through GWAS (Cooper, et al. 2008, Takeuchi, et al. 2009). In 2007, the Food and Drug Administration (FDA) added a genetic table to the warfarin label (Gage and Lesko 2008). Additionally, warfarin dosing algorithms have also been constructed to take both clinical and genetic information under consideration to calculate a starting warfarin dose for a patient (Finkelman, et al. 2011, Klein, et al. 2009). However, the translational of these results into the clinic has been difficult for multiple reasons including mixed results from randomized controlled clinical trials (Anderson, et al. 2012, Gage, et al. 2017, Kimmel, et al. 2013, Pirmohamed, et al. 2013), ancestry specific variants influencing stable warfarin dose (Asiimwe, et al. 2020, Daneshjou, et al. 2014, Scott, et al. 2009), and the use of direct oral anticoagulants or DOACs (Burn and Pirmohamed 2018). These studies highlight the importance of race, and ancestry specific variants that can impact drug response.
Clopidogrel, an oral antiplatelet medication, is cardiovascular drug with a significant association discovered through GWAS. The association between CYP2C19*2 and diminished clopidogrel response was discovered through a GWAS for platelet activation in healthy individuals after taking clopidogrel for seven days (Shuldiner, et al. 2009). This finding was further validated through the association of CYP2C19*2 and adverse cardiovascular events in clopidogrel treated patients undergoing percutaneous coronary intervention (PCI) (Mega, et al. 2009, Mega, et al. 2010, Simon, et al. 2009). This led to the FDA issuing a “Black Box” warning for the clopidogrel label in March 2010 (Ford and Taubert 2013), and the implementation of CYP2C19 genotyping before prescribing antiplatelet treatment after PCI at some medical centers (Cavallari, et al. 2017, Empey, et al. 2018, Pulley, et al. 2012). In the setting of clopidogrel therapy after stroke, the association between CYP2C19*2 and adverse cardiovascular outcomes has been mixed (Hoh, et al. 2016, Jia, et al. 2013, McDonough, et al. 2015). Additionally, while the results from real-world and pragmatic data support genotyping in the setting of PCI (Cavallari, et al. 2018), data from clinical trials are less clear (Claassens, et al. 2019, Pereira, et al. 2020). Further, with the introduction of newer antiplatelet agents, there have also been studies discussing using CYP2C19 genotyping to de-escalate patients from a newer antiplatelet agent to clopidogrel (Angiolillo, et al. 2019, Cavallari and Lee 2019, Martin, et al. 2020, Sibbing, et al. 2017). Overall, the story of clopidogrel pharmacogenomics highlights the importance of therapeutic area, and the need to understand the changing use of the drug in the clinical setting.
Additional cardiovascular therapeutic areas that have seen various degrees of success in discovery PGx are hypercholesterolemia (statins) and hypertension (antihypertensives). In a GWAS of patients treated with high dose simvastatin (80 mg per day) of 85 cases of incident myopathy compared to 90 controls, a SNP in SLCO1B1 was identified at the genome-wide significant level. The SNP is in linkage disequilibrium with rs4149056 (*5), a nonsynonymous SNP in SLCO1B1, which showed an ~5 times increased risk for statin-induced myopathy per C allele (Link, et al. 2008). Despite the small sample size, the association was replicated (Link, et al. 2008), and guidelines have been created for simvastatin dosing based on SLCO1B1 phenotype (Ramsey, et al. 2014). This illustrates that even with a small sample size, significant SNPs can still be identified in cardiovascular PGx studies if the effect size is large. While there are currently no clinical pharmacogenomic guidelines in hypertension, there have been significant SNPs identified and replicated with blood pressure response and adverse cardiovascular outcomes after treatment with antihypertensive drugs. SNPs in and near NEDD4L, PRKCA, and YEATS4 have been significantly associated with blood pressure response to thiazide diuretics, and successfully replicated (Duarte, et al. 2012, McDonough, et al. 2013a, Turner, et al. 2013b). A meta-analysis of five randomized controlled trials identified a missense SNP in BST1, rs28404156, that was significantly associated with blood pressure response to β-blockers (Singh, et al. 2019). Additionally, SNPs in the following genes have been significantly associated with adverse cardiovascular outcomes after treatment with antihypertensive drugs: ADRB1 with β-blockers (Pacanowski, et al. 2008), NEDD4L with thiazide diuretics (McDonough, et al. 2013a, Svensson-Färbom, et al. 2011), CACNA1C and CACNB2 with calcium channel blockers (Beitelshees, et al. 2009, Niu, et al. 2010), FGB with ACE-inhibitors (Lynch, et al. 2009), and SIGLEC12, A1BG, and F5 with β-blockers and calcium channel blockers (McDonough, et al. 2013c). In hypertension, while there have been many studies there are few well replicated PGx SNPs. This illustrates the difficulty in replication, and the value in meta-analyses to increase sample size and power.
There is still great need for further study in each of these therapeutic areas to identify additional genetic variants that influence cardiovascular drug response. There is also great need to study the identified variants in other racial and ancestry groups, and to conduct additional studies in racial and ethnic minorities. Prior research has shown both the existence and importance of racial specific PGx variants (Asiimwe, et al. 2020, Cavallari and Perera 2012, Daneshjou, et al. 2014), and the need to include diverse populations in research so they may also benefit from any future implementation.
CRITICAL PARAMETERS AND TROUBLESHOOTING
One of the biggest issues in cardiovascular PGx is replication. Many cardiovascular PGx studies are sub-studies of larger, cardiovascular RCTs. While these studies are advantageous due to the higher quality of data and multiple time points for data collection, the exact same clinical trial will never be conducted twice, making replication harder. Other common data sources for cardiovascular PGx studies are longitudinal cohort studies with prescription drug information. However, this information is often patient reported and only recorded once a year, or once or twice over the study period. This methodology also makes it difficult to construct many cardiovascular PGx phenotypes. With these common themes in cardiovascular PGx studies, replication of top SNPs from one study in another study with the exact same PGx phenotype, drug, and population is very difficult.
Nevertheless, replication and validation in cardiovascular PGx studies is not impossible. The strategy for replication may require additional considerations. First, slight changes in the drug may be considered. Either a drug in the same class (e.g., the thiazide diuretic hydrochlorothiazide and the thiazide-like diuretic chlorthalidone), or a different dose of the same drug (e.g., 80 mg vs 40 mg of simvastatin). Second, the PGx phenotype maybe slightly different. A couple examples from prior cardiovascular PGx studies include a discovery GWAS of antiplatelet response to clopidogrel and a replication of adverse cardiovascular events after treatment with clopidogrel in patients undergoing PCI (Shuldiner, et al. 2009), and plasma renin activity in hypertension patients and validation with corresponding blood pressure response to antihypertensive therapy (McDonough, et al. 2018). Third, the study population may be different. There are multiple examples where a trans-ethnic or trans-racial validation strategy was used (Fontana, et al. 2014, Gong, et al. 2015, McDonough, et al. 2013b). Overall, considerations for a replication plan should occur early in the study design and should be flexible.
STATISTICAL ANALYSIS:
The most common statistical methods used for analysis of cardiovascular PGx studies are very similar to those used in other genetic studies and GWAS. If the cardiovascular PGx phenotype is continuous a linear regression model is often used, and if the cardiovascular PGx phenotype is binary a logistic regression model is often used. In consideration for additional comorbidities and exposures that could influence drug response, multiple covariates are often added to the regression model. Commonly these include age, sex, principal components for ancestry, other drug exposures, other comorbidities known to influence the drug response or outcome, and randomization arm if the data is originating from a RCT. Some cardiovascular PGx studies include a drug interaction term, investigating the interaction between SNP genotype and drug exposure. Many software programs that are used for GWAS analysis allow for the inclusion of an interaction term (e.g., SNPxdrug). A list of commonly used software programs in cardiovascular PGx studies is shown in Table 3.
For GWAS analysis, some of the common programs used in cardiovascular PGx include PLINK, ProbABEL, and EPACTS (Table 3). Additionally, there are also R packages that many investigators utilize for cardiovascular PGx GWAS analyses. For cardiovascular PGx meta-analysis studies, METAL is commonly used (Table 3). Another feature in cardiovascular PGx studies is interpreting the results of the top SNP(s) in context of the drug response, drug outcome, or drug interaction. In order to conduct analyses that are focused on the drug response or drug outcome, investigators may conduct genotypic means analyses for quantitative traits or look at the outcome by genotype and by drug for binary traits. Finally, in all cardiovascular PGx studies, it is important to conduct a power calculation (Table 3).
UNDERSTANDING RESULTS:
After completing the statistical analyses, the replication and validation plans, and compiling a list of top SNPs; the next step is to put them in context. During this phase, it is important to remember the PK and PD of the drug being studied, as well as the physiology of the disease process. There are many examples in prior cardiovascular PGx studies of associations with SNPs in and near genes that encode for drug metabolism enzymes or drug transporters. There are also examples of associations that connect back to the disease phenotype under study (e.g., adverse cardiovascular outcomes) and the findings suggest new mechanisms of action of how a drug may act to reduce risk for an outcome.
FUTURE DIRECTIONS
The field of cardiovascular PGx has seen many exciting discoveries emerge from candidate gene studies and GWAS. The PGx variants associated with warfarin, clopidogrel, and simvastatin have led to FDA label changes (Gage and Lesko 2008), clinical PGx implementation programs (Cavallari, et al. 2017, Empey, et al. 2018), and clinical PGx guidelines (Ramsey, et al. 2014, Relling and Klein 2011). As cardiovascular PGx expands, the use of other ‘omic technologies in discovery studies is also growing. Additionally, the results from cardiovascular PGx discovery GWAS and other –omic studies provide further insight into the pathways of these drugs, and targets for future drug discovery.
SIGNIFICANCE STATEMENT:
Cardiovascular pharmacogenomics is the study and identification of genomic markers that are associated with variability in cardiovascular drug response, cardiovascular drug related outcomes, or cardiovascular drug related adverse events. This overview presents an introduction and historical background to cardiovascular pharmacogenomics, and a protocol for designing a cardiovascular pharmacogenomics study.
ACKNOWLEDGEMENTS:
CWM is supported by National Institutes of Health (NIH) grant K01 HL141690.
LITERATURE CITED:
- Anderson JL, Horne BD, Stevens SM, Woller SC, Samuelson KM, Mansfield JW, Robinson M, Barton S, Brunisholz K, Mower CP, Huntinghouse JA, Rollo JS, Siler D, Bair TL, Knight S, Muhlestein JB, Carlquist JF (2012) A randomized and clinical effectiveness trial comparing two pharmacogenetic algorithms and standard care for individualizing warfarin dosing (CoumaGen-II). Circulation 125: 1997–2005 doi: 10.1161/CIRCULATIONAHA.111.070920 [DOI] [PubMed] [Google Scholar]
- Angiolillo DJ, Patti G, Chan KT, Han Y, Huang WC, Yakovlev A, Paek D, Del Aguila M, Girotra S, Sibbing D (2019) De-escalation from ticagrelor to clopidogrel in acute coronary syndrome patients: a systematic review and meta-analysis. J Thromb Thrombolysis 48: 1–10 doi: 10.1007/s11239-019-01860-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnett DK, Davis BR, Ford CE, Boerwinkle E, Leiendecker-Foster C, Miller MB, Black H, Eckfeldt JH (2005) Pharmacogenetic association of the angiotensin-converting enzyme insertion/deletion polymorphism on blood pressure and cardiovascular risk in relation to antihypertensive treatment: the Genetics of Hypertension-Associated Treatment (GenHAT) study. Circulation 111: 3374–3383 doi: CIRCULATIONAHA.104.504639 [pii]10.1161/CIRCULATIONAHA.104.504639 [DOI] [PubMed] [Google Scholar]
- Asiimwe IG, Zhang EJ, Osanlou R, Krause A, Dillon C, Suarez-Kurtz G, Zhang H, Perini JA, Renta JY, Duconge J, Cavallari LH, Marcatto LR, Beasly MT, Perera MA, Limdi NA, Santos PCJL, Kimmel SE, Lubitz SA, Scott SA, Kawai VK, Jorgensen AL, Pirmohamed M (2020) Genetic Factors Influencing Warfarin Dose in Black-African Patients: A Systematic Review and Meta-Analysis. Clin Pharmacol Ther 107: 1420–1433 doi: 10.1002/cpt.1755 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beitelshees AL, Navare H, Wang D, Gong Y, Wessel J, Moss JI, Langaee TY, Cooper-DeHoff RM, Sadee W, Pepine CJ, Schork NJ, Johnson JA (2009) CACNA1C gene polymorphisms, cardiovascular disease outcomes, and treatment response. Circ Cardiovasc Genet 2: 362–370 doi: CIRCGENETICS.109.857839 [pii] 10.1161/CIRCGENETICS.109.857839 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burn J, Pirmohamed M (2018) Direct oral anticoagulants versus warfarin: is new always better than the old? Open Heart 5: e000712 doi: 10.1136/openhrt-2017-000712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caldwell MD, Awad T, Johnson JA, Gage BF, Falkowski M, Gardina P, Hubbard J, Turpaz Y, Langaee TY, Eby C, King CR, Brower A, Schmelzer JR, Glurich I, Vidaillet HJ, Yale SH, Qi Zhang K, Berg RL, Burmester JK (2008) CYP4F2 genetic variant alters required warfarin dose. Blood 111: 4106–4112 doi: blood-2007–11-122010 [pii] 10.1182/blood-2007-11-122010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavallari LH, Lee CR (2019) A case for genotype-guided de-escalation of antiplatelet therapy after percutaneous coronary angioplasty. Future Cardiol 15: 251–254 doi: 10.2217/fca-2019-0017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavallari LH, Lee CR, Beitelshees AL, Cooper-DeHoff RM, Duarte JD, Voora D, Kimmel SE, McDonough CW, Gong Y, Dave CV, Pratt VM, Alestock TD, Anderson RD, Alsip J, Ardati AK, Brott BC, Brown L, Chumnumwat S, Clare-Salzler MJ, Coons JC, Denny JC, Dillon C, Elsey AR, Hamadeh IS, Harada S, Hillegass WB, Hines L, Horenstein RB, Howell LA, Jeng LJB, Kelemen MD, Lee YM, Magvanjav O, Montasser M, Nelson DR, Nutescu EA, Nwaba DC, Pakyz RE, Palmer K, Peterson JF, Pollin TI, Quinn AH, Robinson SW, Schub J, Skaar TC, Smith DM, Sriramoju VB, Starostik P, Stys TP, Stevenson JM, Varunok N, Vesely MR, Wake DT, Weck KE, Weitzel KW, Wilke RA, Willig J, Zhao RY, Kreutz RP, Stouffer GA, Empey PE, Limdi NA, Shuldiner AR, Winterstein AG, Johnson JA, Network I (2018) Multisite Investigation of Outcomes With Implementation of CYP2C19 Genotype-Guided Antiplatelet Therapy After Percutaneous Coronary Intervention. JACC Cardiovasc Interv 11: 181–191 doi: 10.1016/j.jcin.2017.07.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavallari LH, Perera MA (2012) The future of warfarin pharmacogenetics in under-represented minority groups. Future Cardiol 8: 563–576 doi: 10.2217/fca.12.31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavallari LH, Weitzel KW, Elsey AR, Liu X, Mosley SA, Smith DM, Staley BJ, Winterstein AG, Mathews CA, Franchi F, Rollini F, Angiolillo DJ, Starostik P, Clare-Salzler MJ, Nelson DR, Johnson JA (2017) Institutional profile: University of Florida Health Personalized Medicine Program. Pharmacogenomics 18: 421–426 doi: 10.2217/pgs-2017-0028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Claassens DMF, Vos GJA, Bergmeijer TO, Hermanides RS, van ‘t Hof AWJ, van der Harst P, Barbato E, Morisco C, Tjon Joe Gin RM, Asselbergs FW, Mosterd A, Herrman JR, Dewilde WJM, Janssen PWA, Kelder JC, Postma MJ, de Boer A, Boersma C, Deneer VHM, Ten Berg JM (2019) A Genotype-Guided Strategy for Oral P2Y. N Engl J Med 381: 1621–1631 doi: 10.1056/NEJMoa1907096 [DOI] [PubMed] [Google Scholar]
- Cooper GM, Johnson JA, Langaee TY, Feng H, Stanaway IB, Schwarz UI, Ritchie MD, Stein CM, Roden DM, Smith JD, Veenstra DL, Rettie AE, Rieder MJ (2008) A genome-wide scan for common genetic variants with a large influence on warfarin maintenance dose. Blood 112: 1022–1027 doi: 10.1182/blood-2008-01-134247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Andrea G, D’Ambrosio RL, Di Perna P, Chetta M, Santacroce R, Brancaccio V, Grandone E, Margaglione M (2005) A polymorphism in the VKORC1 gene is associated with an interindividual variability in the dose-anticoagulant effect of warfarin. Blood 105: 645–649 doi: DOI 10.1182/blood-2004-06-2111 [DOI] [PubMed] [Google Scholar]
- Daneshjou R, Gamazon ER, Burkley B, Cavallari LH, Johnson JA, Klein TE, Limdi N, Hillenmeyer S, Percha B, Karczewski KJ, Langaee T, Patel SR, Bustamante CD, Altman RB, Perera MA (2014) Genetic variant in folate homeostasis is associated with lower warfarin dose in African Americans. Blood 124: 2298–2305 doi: 10.1182/blood-2014-04-568436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duarte JD, Turner ST, Tran B, Chapman AB, Bailey KR, Gong Y, Gums JG, Langaee TY, Beitelshees AL, Cooper-Dehoff RM, Boerwinkle E, Johnson JA (2012) Association of chromosome 12 locus with antihypertensive response to hydrochlorothiazide may involve differential YEATS4 expression. Pharmacogenomics J tpj20124 [pii] 10.1038/tpj.2012.4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Empey PE, Stevenson JM, Tuteja S, Weitzel KW, Angiolillo DJ, Beitelshees AL, Coons JC, Duarte JD, Franchi F, Jeng LJB, Johnson JA, Kreutz RP, Limdi NA, Maloney KA, Owusu Obeng A, Peterson JF, Petry N, Pratt VM, Rollini F, Scott SA, Skaar TC, Vesely MR, Stouffer GA, Wilke RA, Cavallari LH, Lee CR, Network I (2018) Multisite Investigation of Strategies for the Implementation of CYP2C19 Genotype-Guided Antiplatelet Therapy. Clin Pharmacol Ther 104: 664–674 doi: 10.1002/cpt.1006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Finkelman BS, Gage BF, Johnson JA, Brensinger CM, Kimmel SE (2011) Genetic warfarin dosing: tables versus algorithms. J Am Coll Cardiol 57: 612–618 doi: 10.1016/j.jacc.2010.08.643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fontana V, McDonough CW, Gong Y, El Rouby NM, Sa AC, Taylor KD, Chen YD, Gums JG, Chapman AB, Turner ST, Pepine CJ, Johnson JA, Cooper-DeHoff RM (2014) Large-scale gene-centric analysis identifies polymorphisms for resistant hypertension. J Am Heart Assoc 3: e001398 doi: 10.1161/jaha.114.001398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ford NF, Taubert D (2013) Clopidogrel, CYP2C19, and a Black Box. J Clin Pharmacol 53: 241–248 doi: 10.1002/jcph.17 [DOI] [PubMed] [Google Scholar]
- Gage BF, Bass AR, Lin H, Woller SC, Stevens SM, Al-Hammadi N, Li J, Rodríguez T, Miller JP, McMillin GA, Pendleton RC, Jaffer AK, King CR, Whipple BD, Porche-Sorbet R, Napoli L, Merritt K, Thompson AM, Hyun G, Anderson JL, Hollomon W, Barrack RL, Nunley RM, Moskowitz G, Dávila-Román V, Eby CS (2017) Effect of Genotype-Guided Warfarin Dosing on Clinical Events and Anticoagulation Control Among Patients Undergoing Hip or Knee Arthroplasty: The GIFT Randomized Clinical Trial. JAMA 318: 1115–1124 doi: 10.1001/jama.2017.11469 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gage BF, Eby C, Johnson JA, Deych E, Rieder MJ, Ridker PM, Milligan PE, Grice G, Lenzini P, Rettie AE, Aquilante CL, Grosso L, Marsh S, Langaee T, Farnett LE, Voora D, Veenstra DL, Glynn RJ, Barrett A, McLeod HL (2008) Use of pharmacogenetic and clinical factors to predict the therapeutic dose of warfarin. Clin Pharmacol Ther 84: 326–331 doi: clpt200810 [pii] 10.1038/clpt.2008.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gage BF, Lesko LJ (2008) Pharmacogenetics of warfarin: regulatory, scientific, and clinical issues. J Thromb Thrombolysis 25: 45–51 doi: 10.1007/s11239-007-0104-y [DOI] [PubMed] [Google Scholar]
- Gong Y, McDonough CW, Beitelshees AL, El Rouby N, Hiltunen TP, O’Connell JR, Padmanabhan S, Langaee TY, Hall K, Schmidt SO, Curry RW Jr., Gums JG, Donner KM, Kontula KK, Bailey KR, Boerwinkle E, Takahashi A, Tanaka T, Kubo M, Chapman AB, Turner ST, Pepine CJ, Cooper-DeHoff RM, Johnson JA (2015) PTPRD gene associated with blood pressure response to atenolol and resistant hypertension. J Hypertens 33: 2278–2285 doi: 10.1097/hjh.0000000000000714 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoh BL, Gong Y, McDonough CW, Waters MF, Royster AJ, Sheehan TO, Burkley B, Langaee TY, Mocco J, Zuckerman SL, Mummareddy N, Stephens ML 2nd, Ingram C, Shaffer CM, Denny JC, Brilliant MH, Kitchner TE, Linneman JG, Roden DM, Johnson JA (2016) CYP2C19 and CES1 polymorphisms and efficacy of clopidogrel and aspirin dual antiplatelet therapy in patients with symptomatic intracranial atherosclerotic disease. J Neurosurg 124: 1746–1751 doi: 10.3171/2015.6.jns15795 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jia DM, Chen ZB, Zhang MJ, Yang WJ, Jin JL, Xia YQ, Zhang CL, Shao Y, Chen C, Xu Y (2013) CYP2C19 polymorphisms and antiplatelet effects of clopidogrel in acute ischemic stroke in China. Stroke 44: 1717–1719 doi: 10.1161/STROKEAHA.113.000823 [DOI] [PubMed] [Google Scholar]
- Kimmel SE, French B, Kasner SE, Johnson JA, Anderson JL, Gage BF, Rosenberg YD, Eby CS, Madigan RA, McBane RB, Abdel-Rahman SZ, Stevens SM, Yale S, Mohler ER, Fang MC, Shah V, Horenstein RB, Limdi NA, Muldowney JA, Gujral J, Delafontaine P, Desnick RJ, Ortel TL, Billett HH, Pendleton RC, Geller NL, Halperin JL, Goldhaber SZ, Caldwell MD, Califf RM, Ellenberg JH, Investigators C (2013) A pharmacogenetic versus a clinical algorithm for warfarin dosing. N Engl J Med 369: 2283–2293 doi: 10.1056/NEJMoa1310669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein TE, Altman RB, Eriksson N, Gage BF, Kimmel SE, Lee MTM, Limdi NA, Page D, Roden DM, Wagner MJ, Caldwell MD, Johnson JA, Chen YT, Wen MS, Caraco Y, Achache I, Blotnick S, Muszkat M, Shin JG, Kim HS, Suarez-Kurtz G, Perini JA, Silva-Assuncao E, Andereson JL, Horne BD, Carlquist JF, Caldwell MD, Berg RL, Burmester JK, Goh BC, Lee SC, Kamali F, Sconce E, Daly AK, Wu AHB, Langaee TY, Feng H, Cavallari L, Momary K, Pirmohamed M, Jorgensen A, Toh CH, Williamson P, McLeod H, Evans JP, Weck KE, Brensinger C, Nakamura Y, Mushiroda T, Veenstra D, Meckley L, Rieder MJ, Rettie AE, Wadelius M, Melhus H, Stein CM, Schwartz U, Kurnik D, Deych E, Lenzini P, Eby C, Chen LY, Deloukas P, Motsinger-Reif A, Sagreiya H, Srinivasan BS, Lantz E, Chang T, Ritchie M, Lu LS, Shin JG (2009) Estimation of the Warfarin Dose with Clinical and Pharmacogenetic Data (vol 360, pg 753, 2009). New England Journal of Medicine 361: 1613–1613 doi: [DOI] [PMC free article] [PubMed] [Google Scholar]
- Link E, Parish S, Armitage J, Bowman L, Heath S, Matsuda F, Gut I, Lathrop M, Collins R, Group SC (2008) SLCO1B1 variants and statin-induced myopathy--a genomewide study. N Engl J Med 359: 789–799 doi: 10.1056/NEJMoa0801936 [DOI] [PubMed] [Google Scholar]
- Luzum JA, Pakyz RE, Elsey AR, Haidar CE, Peterson JF, Whirl-Carrillo M, Handelman SK, Palmer K, Pulley JM, Beller M, Schildcrout JS, Field JR, Weitzel KW, Cooper-DeHoff RM, Cavallari LH, O’Donnell PH, Altman RB, Pereira N, Ratain MJ, Roden DM, Embi PJ, Sadee W, Klein TE, Johnson JA, Relling MV, Wang L, Weinshilboum RM, Shuldiner AR, Freimuth RR, Program PRNTP (2017) The Pharmacogenomics Research Network Translational Pharmacogenetics Program: Outcomes and Metrics of Pharmacogenetic Implementations Across Diverse Healthcare Systems. Clin Pharmacol Ther 102: 502–510 doi: 10.1002/cpt.630 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lynch AI, Boerwinkle E, Davis BR, Ford CE, Eckfeldt JH, Leiendecker-Foster C, Arnett DK (2009) Antihypertensive pharmacogenetic effect of fibrinogen-beta variant −455G>A on cardiovascular disease, end-stage renal disease, and mortality: the GenHAT study. Pharmacogenet Genomics 19: 415–421 doi: 10.1097/FPC.0b013e32832a8e81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maitland-van der Zee AH, van Wieren-de Wijer DB, de Boer A, Kroon AA, de Leeuw PW, Schiffers P, Janssen RG, Psaty BM, van Duijn CM, Stricker BH, Klungel OH (2011) Genetic variation in the renin--angiotensin system, use of renin--angiotensin system inhibitors and the risk of myocardial infarction. J Renin Angiotensin Aldosterone Syst 12: 208–214 doi: 10.1177/14703203103918341470320310391834 [pii] [DOI] [PubMed] [Google Scholar]
- Manolio TA, Abramowicz M, Al-Mulla F, Anderson W, Balling R, Berger AC, Bleyl S, Chakravarti A, Chantratita W, Chisholm RL, Dissanayake VH, Dunn M, Dzau VJ, Han BG, Hubbard T, Kolbe A, Korf B, Kubo M, Lasko P, Leego E, Mahasirimongkol S, Majumdar PP, Matthijs G, McLeod HL, Metspalu A, Meulien P, Miyano S, Naparstek Y, O’Rourke PP, Patrinos GP, Rehm HL, Relling MV, Rennert G, Rodriguez LL, Roden DM, Shuldiner AR, Sinha S, Tan P, Ulfendahl M, Ward R, Williams MS, Wong JE, Green ED, Ginsburg GS (2015) Global implementation of genomic medicine: We are not alone. Sci Transl Med 7: 290ps213 doi: 10.1126/scitranslmed.aab0194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maranville JC, Cox NJ (2016) Pharmacogenomic variants have larger effect sizes than genetic variants associated with other dichotomous complex traits. Pharmacogenomics J 16: 388–392 doi: 10.1038/tpj.2015.47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin J, Williams AK, Klein MD, Sriramoju VB, Madan S, Rossi JS, Clarke M, Cicci JD, Cavallari LH, Weck KE, Stouffer GA, Lee CR (2020) Frequency and clinical outcomes of CYP2C19 genotype-guided escalation and de-escalation of antiplatelet therapy in a real-world clinical setting. Genet Med 22: 160–169 doi: 10.1038/s41436-019-0611-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonough CW, Burbage SE, Duarte JD, Gong Y, Langaee TY, Turner ST, Gums JG, Chapman AB, Bailey KR, Beitelshees AL, Boerwinkle E, Pepine CJ, Cooper-DeHoff RM, Johnson JA (2013a) Association of variants in NEDD4L with blood pressure response and adverse cardiovascular outcomes in hypertensive patients treated with thiazide diuretics. J Hypertens 31: 698–704 doi: 10.1097/HJH.0b013e32835e2a71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonough CW, Gillis NK, Alsultan A, Chang SW, Kawaguchi-Suzuki M, Lang JE, Shahin MH, Buford TW, El Rouby NM, Sa AC, Langaee TY, Gums JG, Chapman AB, Cooper-DeHoff RM, Turner ST, Gong Y, Johnson JA (2013b) Atenolol induced HDL-C change in the pharmacogenomic evaluation of antihypertensive responses (PEAR) study. PLoS One 8: e76984 doi: 10.1371/journal.pone.0076984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonough CW, Gong Y, Padmanabhan S, Burkley B, Langaee TY, Melander O, Pepine CJ, Dominiczak AF, Cooper-Dehoff RM, Johnson JA (2013c) Pharmacogenomic Association of Nonsynonymous SNPs in SIGLEC12, A1BG, and the Selectin Region and Cardiovascular Outcomes. Hypertension HYPERTENSIONAHA.111.00823 [pii] 10.1161/HYPERTENSIONAHA.111.00823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonough CW, Magvanjav O, Sá ACC, El Rouby NM, Dave C, Deitchman AN, Kawaguchi-Suzuki M, Mei W, Shen Y, Singh RSP, Solayman M, Bailey KR, Boerwinkle E, Chapman AB, Gums JG, Webb A, Scherer SE, Sadee W, Turner ST, Cooper-DeHoff RM, Gong Y, Johnson JA (2018) Genetic Variants Influencing Plasma Renin Activity in Hypertensive Patients From the PEAR Study (Pharmacogenomic Evaluation of Antihypertensive Responses). Circ Genom Precis Med 11: e001854 doi: 10.1161/CIRCGEN.117.001854 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonough CW, McClure LA, Mitchell BD, Gong Y, Horenstein RB, Lewis JP, Field TS, Talbert RL, Benavente OR, Johnson JA, Shuldiner AR (2015) CYP2C19 metabolizer status and clopidogrel efficacy in the Secondary Prevention of Small Subcortical Strokes (SPS3) study. J Am Heart Assoc 4: e001652 doi: 10.1161/jaha.114.001652 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mega JL, Close SL, Wiviott SD, Shen L, Hockett RD, Brandt JT, Walker JR, Antman EM, Macias W, Braunwald E, Sabatine MS (2009) Cytochrome p-450 polymorphisms and response to clopidogrel. N Engl J Med 360: 354–362 doi: 10.1056/NEJMoa0809171 [DOI] [PubMed] [Google Scholar]
- Mega JL, Simon T, Collet JP, Anderson JL, Antman EM, Bliden K, Cannon CP, Danchin N, Giusti B, Gurbel P, Horne BD, Hulot JS, Kastrati A, Montalescot G, Neumann FJ, Shen L, Sibbing D, Steg PG, Trenk D, Wiviott SD, Sabatine MS (2010) Reduced-function CYP2C19 genotype and risk of adverse clinical outcomes among patients treated with clopidogrel predominantly for PCI: a meta-analysis. JAMA 304: 1821–1830 doi: 10.1001/jama.2010.1543 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer UA (2004) Pharmacogenetics - five decades of therapeutic lessons from genetic diversity. Nat Rev Genet 5: 669–676 doi: 10.1038/nrg1428 [DOI] [PubMed] [Google Scholar]
- Niu Y, Gong Y, Langaee TY, Davis HM, Elewa H, Beitelshees AL, Moss JI, Cooper-Dehoff RM, Pepine CJ, Johnson JA (2010) Genetic variation in the beta2 subunit of the voltage-gated calcium channel and pharmacogenetic association with adverse cardiovascular outcomes in the INternational VErapamil SR-Trandolapril STudy GENEtic Substudy (INVEST-GENES). Circ Cardiovasc Genet 3: 548–555 doi: 3/6/548 [pii] 10.1161/CIRCGENETICS.110.957654 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pacanowski MA, Gong Y, Cooper-Dehoff RM, Schork NJ, Shriver MD, Langaee TY, Pepine CJ, Johnson JA (2008) beta-adrenergic receptor gene polymorphisms and beta-blocker treatment outcomes in hypertension. Clin Pharmacol Ther 84: 715–721 doi: clpt2008139 [pii] 10.1038/clpt.2008.139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pereira NL, Farkouh ME, So D, Lennon R, Geller N, Mathew V, Bell M, Bae JH, Jeong MH, Chavez I, Gordon P, Abbott JD, Cagin C, Baudhuin L, Fu YP, Goodman SG, Hasan A, Iturriaga E, Lerman A, Sidhu M, Tanguay JF, Wang L, Weinshilboum R, Welsh R, Rosenberg Y, Bailey K, Rihal C (2020) Effect of Genotype-Guided Oral P2Y12 Inhibitor Selection vs Conventional Clopidogrel Therapy on Ischemic Outcomes After Percutaneous Coronary Intervention: The TAILOR-PCI Randomized Clinical Trial. JAMA 324: 761–771 doi: 10.1001/jama.2020.12443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pirmohamed M, Burnside G, Eriksson N, Jorgensen AL, Toh CH, Nicholson T, Kesteven P, Christersson C, Wahlström B, Stafberg C, Zhang JE, Leathart JB, Kohnke H, Maitland-van der Zee AH, Williamson PR, Daly AK, Avery P, Kamali F, Wadelius M, Group E-P (2013) A randomized trial of genotype-guided dosing of warfarin. N Engl J Med 369: 2294–2303 doi: 10.1056/NEJMoa1311386 [DOI] [PubMed] [Google Scholar]
- Pulley JM, Denny JC, Peterson JF, Bernard GR, Vnencak-Jones CL, Ramirez AH, Delaney JT, Bowton E, Brothers K, Johnson K, Crawford DC, Schildcrout J, Masys DR, Dilks HH, Wilke RA, Clayton EW, Shultz E, Laposata M, McPherson J, Jirjis JN, Roden DM (2012) Operational implementation of prospective genotyping for personalized medicine: the design of the Vanderbilt PREDICT project. Clin Pharmacol Ther 92: 87–95 doi: 10.1038/clpt.2011.371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramsey LB, Johnson SG, Caudle KE, Haidar CE, Voora D, Wilke RA, Maxwell WD, McLeod HL, Krauss RM, Roden DM, Feng Q, Cooper-DeHoff RM, Gong L, Klein TE, Wadelius M, Niemi M (2014) The clinical pharmacogenetics implementation consortium guideline for SLCO1B1 and simvastatin-induced myopathy: 2014 update. Clin Pharmacol Ther 96: 423–428 doi: 10.1038/clpt.2014.125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Relling MV, Klein TE (2011) CPIC: Clinical Pharmacogenetics Implementation Consortium of the Pharmacogenomics Research Network. Clin Pharmacol Ther 89: 464–467 doi: 10.1038/clpt.2010.279 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rettie AE, Wienkers LC, Gonzalez FJ, Trager WF, Korzekwa KR (1994) Impaired (S)-warfarin metabolism catalysed by the R144C allelic variant of CYP2C9. Pharmacogenetics 4: 39–42 doi: 10.1097/00008571-199402000-00005 [DOI] [PubMed] [Google Scholar]
- Rieder MJ, Reiner AP, Gage BF, Nickerson DA, Eby CS, McLeod HL, Blough DK, Thummel KE, Veenstra DL, Rettie AE (2005) Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med 352: 2285–2293 doi: 10.1056/NEJMoa044503 [DOI] [PubMed] [Google Scholar]
- Roden DM, Van Driest SL, Wells QS, Mosley JD, Denny JC, Peterson JF (2018) Opportunities and Challenges in Cardiovascular Pharmacogenomics: From Discovery to Implementation. Circ Res 122: 1176–1190 doi: 10.1161/CIRCRESAHA.117.310965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schelleman H, Klungel OH, Witteman JC, Breteler MM, Hofman A, van Duijn CM, de Boer A, Stricker BH (2008) Interaction between polymorphisms in the renin-angiotensin-system and angiotensin-converting enzyme inhibitor or beta-blocker use and the risk of myocardial infarction and stroke. Pharmacogenomics J 8: 400–407 doi: 10.1038/sj.tpj.65004936500493 [pii] [DOI] [PubMed] [Google Scholar]
- Scott SA, Jaremko M, Lubitz SA, Kornreich R, Halperin JL, Desnick RJ (2009) CYP2C9*8 is prevalent among African-Americans: implications for pharmacogenetic dosing. Pharmacogenomics 10: 1243–1255 doi: 10.2217/pgs.09.71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shuldiner AR, O’Connell JR, Bliden KP, Gandhi A, Ryan K, Horenstein RB, Damcott CM, Pakyz R, Tantry US, Gibson Q, Pollin TI, Post W, Parsa A, Mitchell BD, Faraday N, Herzog W, Gurbel PA (2009) Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA 302: 849–857 doi: 302/8/849 [pii] 10.1001/jama.2009.1232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sibbing D, Aradi D, Jacobshagen C, Gross L, Trenk D, Geisler T, Orban M, Hadamitzky M, Merkely B, Kiss RG, Komócsi A, Dézsi CA, Holdt L, Felix SB, Parma R, Klopotowski M, Schwinger RHG, Rieber J, Huber K, Neumann FJ, Koltowski L, Mehilli J, Huczek Z, Massberg S, Investigators T-A (2017) Guided de-escalation of antiplatelet treatment in patients with acute coronary syndrome undergoing percutaneous coronary intervention (TROPICAL-ACS): a randomised, open-label, multicentre trial. Lancet 390: 1747–1757 doi: 10.1016/S0140-6736(17)32155-4 [DOI] [PubMed] [Google Scholar]
- Simon T, Verstuyft C, Mary-Krause M, Quteineh L, Drouet E, Meneveau N, Steg PG, Ferrieres J, Danchin N, Becquemont L (2009) Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med 360: 363–375 doi: NEJMoa0808227 [pii] 10.1056/NEJMoa0808227 [DOI] [PubMed] [Google Scholar]
- Singh S, Warren HR, Hiltunen TP, McDonough CW, El Rouby N, Salvi E, Wang Z, Garofalidou T, Fyhrquist F, Kontula KK, Glorioso V, Zaninello R, Glorioso N, Pepine CJ, Munroe PB, Turner ST, Chapman AB, Boerwinkle E, Johnson JA, Gong Y, Cooper-DeHoff RM (2019) Genome-Wide Meta-Analysis of Blood Pressure Response to β. J Am Heart Assoc 8: e013115 doi: 10.1161/JAHA.119.013115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Svensson-Färbom P, Wahlstrand B, Almgren P, Dahlberg J, Fava C, Kjeldsen S, Hedner T, Melander O (2011) A functional variant of the NEDD4L gene is associated with beneficial treatment response with β-blockers and diuretics in hypertensive patients. J Hypertens 29: 388–395 doi: 10.1097/HJH.0b013e3283410390 [DOI] [PubMed] [Google Scholar]
- Takeuchi F, McGinnis R, Bourgeois S, Barnes C, Eriksson N, Soranzo N, Whittaker P, Ranganath V, Kumanduri V, McLaren W, Holm L, Lindh J, Rane A, Wadelius M, Deloukas P (2009) A Genome-Wide Association Study Confirms VKORC1, CYP2C9, and CYP4F2 as Principal Genetic Determinants of Warfarin Dose. Plos Genetics 5: - doi: ARTN e1000433DOI 10.1371/journal.pgen.1000433 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thijssen HHW, Drittij MJ, Vervoort LMT, de Vries-Hanje JC (2001) Altered pharmacokinetics of R- and S-acenocoumarol in a subject heterozygous for CYP2C9*3. Clinical Pharmacology & Therapeutics 70: 292–298 doi: [DOI] [PubMed] [Google Scholar]
- Turner ST, Boerwinkle E, O’Connell JR, Bailey KR, Gong Y, Chapman AB, McDonough CW, Beitelshees AL, Schwartz GL, Gums JG, Padmanabhan S, Hiltunen TP, Citterio L, Donner KM, Hedner T, Lanzani C, Melander O, Saarela J, Ripatti S, Wahlstrand B, Manunta P, Kontula K, Dominiczak AF, Cooper-DeHoff RM, Johnson JA (2013a) Genomic association analysis of common variants influencing antihypertensive response to hydrochlorothiazide. Hypertension 62: 391–397 doi: 10.1161/hypertensionaha.111.00436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner ST, Boerwinkle E, O’Connell JR, Bailey KR, Gong Y, Chapman AB, McDonough CW, Beitelshees AL, Schwartz GL, Gums JG, Padmanabhan S, Hiltunen TP, Citterio L, Donner KM, Hedner T, Lanzani C, Melander O, Saarela J, Ripatti S, Wahlstrand B, Manunta P, Kontula K, Dominiczak AF, Cooper-Dehoff RM, Johnson JA (2013b) Genomic Association Analysis of Common Variants Influencing Antihypertensive Response to Hydrochlorothiazide. Hypertension HYPERTENSIONAHA.111.00436 [pii] 10.1161/HYPERTENSIONAHA.111.00436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L, McLeod HL, Weinshilboum RM (2011) Genomics and drug response. N Engl J Med 364: 1144–1153 doi: 10.1056/NEJMra1010600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weitzel KW, Alexander M, Bernhardt BA, Calman N, Carey DJ, Cavallari LH, Field JR, Hauser D, Junkins HA, Levin PA, Levy K, Madden EB, Manolio TA, Odgis J, Orlando LA, Pyeritz R, Wu RR, Shuldiner AR, Bottinger EP, Denny JC, Dexter PR, Flockhart DA, Horowitz CR, Johnson JA, Kimmel SE, Levy MA, Pollin TI, Ginsburg GS (2016) The IGNITE network: a model for genomic medicine implementation and research. BMC Med Genomics 9: 1 doi: 10.1186/s12920-015-0162-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilke RA, Ramsey LB, Johnson SG, Maxwell WD, McLeod HL, Voora D, Krauss RM, Roden DM, Feng Q, Cooper-Dehoff RM, Gong L, Klein TE, Wadelius M, Niemi M (2012) The clinical pharmacogenomics implementation consortium: CPIC guideline for SLCO1B1 and simvastatin-induced myopathy. Clin Pharmacol Ther 92: 112–117 doi: 10.1038/clpt.2012.57 [DOI] [PMC free article] [PubMed] [Google Scholar]