Hostname: page-component-848d4c4894-p2v8j Total loading time: 0 Render date: 2024-05-01T06:09:03.808Z Has data issue: false hasContentIssue false

Cardiovascular precision medicine – A pharmacogenomic perspective

Published online by Cambridge University Press:  29 June 2023

Sandosh Padmanabhan
Affiliation:
BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
Clea du Toit
Affiliation:
BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
Anna F. Dominiczak*
Affiliation:
BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
*
Corresponding author: Anna F. Dominiczak; Email: Anna.Dominiczak@glasgow.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Precision medicine envisages the integration of an individual’s clinical and biological features obtained from laboratory tests, imaging, high-throughput omics and health records, to drive a personalised approach to diagnosis and treatment with a higher chance of success. As only up to half of patients respond to medication prescribed following the current one-size-fits-all treatment strategy, the need for a more personalised approach is evident. One of the routes to transforming healthcare through precision medicine is pharmacogenomics (PGx). Around 95% of the population is estimated to carry one or more actionable pharmacogenetic variants and over 75% of adults over 50 years old are on a prescription with a known PGx association. Whilst there are compelling examples of pharmacogenomic implementation in clinical practice, the case for cardiovascular PGx is still evolving. In this review, we shall summarise the current status of PGx in cardiovascular diseases and look at the key enablers and barriers to PGx implementation in clinical practice.

Topics structure

Topic(s)

Subtopic(s)

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press

Impact statement

Pharmacogenomics, the study of the effect of inherited or acquired genetic variation on differences in drug response or adverse effects. Around 95% of the population carry one or more actionable pharmacogenetic variants and over 75% of adults over 50 years old are on a prescription with a known PGx association. Pharmacogenomic evidence for cardiovascular drugs is growing along with emerging evidence for efficacy and cost-effectiveness. Successful pharmacogenomic implementation in healthcare requires strong scientific evidence, comprehensive and updated clinical guidelines, clinician champions and stakeholder engagement.

Introduction

An average one-size-fits-all approach is the foundation of the existing general healthcare paradigm of therapeutic, and preventative interventions. Whilst this is a very practical and effective strategy, only 40–50% of patients respond to treatment in this all-comers approach prescribed as per current practice, indicating a large proportion of the population may be facing a deficit in addressing their medical needs (Collins and Varmus, Reference Collins and Varmus2015). This requirement for a transformation in the current paradigm of healthcare has motivated the emergence of precision medicine as a more targeted approach to treatment (Goldberger and Buxton, Reference Goldberger and Buxton2013; Schork, Reference Schork2015). Precision medicine envisages an integration of an individual’s clinical and biological features obtained from laboratory tests, imaging, high-throughput omics and health records, to drive a personalised approach to diagnosis and treatment with a higher chance of success (Collins and Varmus, Reference Collins and Varmus2015). The anticipated benefits of the precision medicine approach for patients are quicker diagnosis and targeted treatment leading to higher treatment success with minimal to no adverse drug reactions (ADRs), with wider benefits in terms of decreased healthcare costs and increased economic productivity.

One of the routes to precision medicine is pharmacogenomics (PGx), the study of the effect of inherited or acquired genetic variation on drug absorption, distribution, metabolism and excretion (pharmacokinetics) or modification of drug target or biological pathways (pharmacodynamics) resulting in variations in drug response or adverse effects. Around 95% of the population carry one or more actionable pharmacogenetic variants and over 75% of adults over 50 years old are on a prescription with a known PGx association (Chanfreau-Coffinier et al., Reference Chanfreau-Coffinier, Hull, Lynch, DuVall, Damrauer, Cunningham, Voight, Matheny, Oslin, Icardi and Tuteja2019; Heise et al., Reference Heise, Gallo, Curry and Woosley2020; Zhou and Lauschke, Reference Zhou and Lauschke2022; Zhou et al., Reference Zhou, Nevosadova, Eliasson and Lauschke2023). The U.S. Food and Drug Administration (FDA) lists around 499 drugs which have PGx biomarkers in the labelling, with around a 100 of them linked to data supporting PGx-guided therapeutic recommendations (FDA, 2023a, 2023b). PharmGKB (PharmGKB, 2023a, 2023b) and the Clinical Pharmacogenetics Implementation Consortium (CPIC) (Relling et al., Reference Relling, Klein, Gammal, Whirl-Carrillo, Hoffman and Caudle2020) publish evidence-based, peer-reviewed guidelines on applying PGx test results into actionable prescribing decisions. PharmGKB Level 1 genes or gene–drug combinations are considered pharmacogenomically significant and are linked to specific prescribing guidance. Similarly, CPIC Levels A and B indicate that genetic information should be considered before prescribing.

CPIC currently reports around 480 gene–drug interactions, including 93 gene–drug pairs (24 genes with 75 drugs) that are annotated with Level A evidence and prescription guidelines (Crews et al., Reference Crews, Gaedigk, Dunnenberger, Leeder, Klein, Caudle, Haidar, Shen, Callaghan, Sadhasivam, Prows, Kharasch, Skaar and Implementation2014; Ramsey et al., Reference Ramsey, Johnson, Caudle, Haidar, Voora, Wilke, Maxwell, McLeod, Krauss, Roden, Feng, Cooper-DeHoff, Gong, Klein, Wadelius and Niemi2014; Hicks et al., Reference Hicks, Bishop, Sangkuhl, Muller, Ji, Leckband, Leeder, Graham, Chiulli, LLerena, Skaar, Scott, Stingl, Klein, Caudle and Gaedigk2015; Bell et al., Reference Bell, Caudle, Whirl-Carrillo, Gordon, Hikino, Prows, Gaedigk, Agundez, Sadhasivam, Klein and Schwab2017; Johnson et al., Reference Johnson, Caudle, Gong, Whirl-Carrillo, Stein, Scott, Lee, Gage, Kimmel, Perera, Anderson, Pirmohamed, Klein, Limdi, Cavallari and Wadelius2017; Amstutz et al., Reference Amstutz, Henricks, Offer, Barbarino, Schellens, Swen, Klein, McLeod, Caudle, Diasio and Schwab2018; Relling et al., Reference Relling, Schwab, Whirl-Carrillo, Suarez-Kurtz, Pui, Stein, Moyer, Evans, Klein, Antillon-Klussmann, Caudle, Kato, Yeoh, Schmiegelow and Yang2019; CPIC, 2022). Although integration of PGx into routine clinical practice is not widespread, the recent PREPARE trial demonstrated both efficacy and feasibility of implementation of a 12 gene pharmacogenomic panel across diverse European health-care system organisations and settings (Swen et al., Reference Swen, van der Wouden, Manson, Abdullah-Koolmees, Blagec, Blagus, Bohringer, Cambon-Thomsen, Cecchin, Cheung, Deneer, Dupui, Ingelman-Sundberg, Jonsson, Joefield-Roka, Just, Karlsson, Konta, Koopmann, Kriek, Lehr, Mitropoulou, Rial-Sebbag, Rollinson, Roncato, Samwald, Schaeffeler, Skokou, Schwab, Steinberger, Stingl, Tremmel, Turner, van Rhenen, Davila Fajardo, Dolzan, Patrinos, Pirmohamed, Sunder-Plassmann, Toffoli and Guchelaar2023) even if only limited to current CPIC Level A drugs (Chanfreau-Coffinier et al., Reference Chanfreau-Coffinier, Hull, Lynch, DuVall, Damrauer, Cunningham, Voight, Matheny, Oslin, Icardi and Tuteja2019; Heise et al., Reference Heise, Gallo, Curry and Woosley2020; Relling et al., Reference Relling, Klein, Gammal, Whirl-Carrillo, Hoffman and Caudle2020; Hicks et al., Reference Hicks, El Rouby, Ong, Schildcrout, Ramsey, Shi, Anne Tang, Aquilante, Beitelshees, Blake, Cimino, Davis, Empey, Kao, Lemkin, Limdi, PL, Rosenman, Skaar, Teal, Tuteja, Wiley, Williams, Winterstein, Van Driest, Cavallari and Peterson2021; Pritchard et al., Reference Pritchard, Patel, Stephens and Mc Leod2022). Only a small subset of the roughly 15% of medications that cite PGx information on their labels have actionable pharmacogenes (Ehmann et al., Reference Ehmann, Caneva, Prasad, Paulmichl, Maliepaard, Llerena, Ingelman-Sundberg and Papaluca-Amati2015; Mehta et al., Reference Mehta, Uber, Ingle, Li, Liu, Thakkar, Ning, Wu, Yang, Harris, Zhou, Xu, Tong, Lesko and Fang2020). Of the approximately 20,000 human genes, only 34 of them are considered clinically actionable with PGx (PharmGKB level 1) (PharmGKB, 2023a, 2023b). The majority of PGx-labelled agents are cancer therapies targeted for somatic mutations, rather than germline variants. Actionable germline PGx variants are present for around 7% of medications with CPIC Level A or B recommendations directing prescribing changes based on genotype (Relling et al., Reference Relling, Klein, Gammal, Whirl-Carrillo, Hoffman and Caudle2020).

Pharmacogenomics

The broad clinical relevance of PGx is evident across the medical spectrum from improving treatment efficacy to avoiding ADRs. CYP2D6 genotype guided optimisation of opioid analgesia resulted in a 30% reduction in pain intensity among 24% of patients (Smith et al., Reference Smith, Weitzel, Elsey, Langaee, Gong, Wake, Duong, Hagen, Harle, Mercado, Nagoshi, Newsom, Wright, Rosenberg, Starostik, Clare-Salzler, Schmidt, Fillingim, Johnson and Cavallari2019). Antidepressant prescribing guided by PGx variants across eight genes (CYP1A2, CYP2C9, CYP2C19, CYP3A4, CYP2B6, CYP2D6, HTR2A, SLC6A4) in the Genomics Used to Improve DEpression Decisions (GUIDED) trial (Greden et al., Reference Greden, Parikh, Rothschild, Thase, Dunlop, DeBattista, Conway, Forester, Mondimore, Shelton, Macaluso, Li, Brown, Gilbert, Burns, Jablonski and Dechairo2019) showed improved response and remission rates in difficult-to-treat depression, but no difference between the study arms for symptom improvement (primary outcome). A trial in a predominantly white human immunodeficiency virus type 1 infected population showed 100% elimination of immunologically confirmed abacavir hypersensitivity syndrome in those randomised to pre-emptive HLA-B*57:01-guided abacavir initiation (Mallal et al., Reference Mallal, Phillips, Carosi, Molina, Workman, Tomazic, Jagel-Guedes, Rugina, Kozyrev, Cid, Hay, Nolan, Hughes, Hughes, Ryan, Fitch, Thorborn and Benbow2008). Similarly, pre-emptive DPYD genotype guided dosing reduced from 73% to 28% the risk of fluoropyrimidine toxicity and completely abolished fluoropyrimidine-related mortality (Deenen et al., Reference Deenen, Meulendijks, Cats, Sechterberger, Severens, Boot, Smits, Rosing, Mandigers, Soesan, Beijnen and Schellens2016). Whilst these examples are compelling, the case for cardiovascular PGx is still evolving. In this review, we shall summarise the current status of PGx in cardiovascular diseases (CVDs) and look at the key enablers and barriers to PGx implementation in clinical practice.

Warfarin

The coumarin derivatives (warfarin, acenocoumarol and phenprocoumon) are a mainstay of CVD therapy due to their crucial role in preventing or treating thromboembolism.

Coumarins inhibit vitamin K epoxide reductase complex subunit 1 (VKORC1) and thence clotting factors II, VII, IX and X to yield its pharmacological anticoagulant effect (Verhoef et al., Reference Verhoef, Redekop, Daly, van Schie, de Boer and Maitland-van der Zee2014). Coumarins are racemic mixtures with one dominant pharmacological enantiomer. For warfarin, S-warfarin is 3–5 times more potent than R-warfarin and is preferentially metabolised by CYP2C9 (Kaminsky and Zhang, Reference Kaminsky and Zhang1997). Warfarin is unique in that, unlike most other drugs, its dose titration is based on coagulation levels in response to treatment. Warfarin has a narrow therapeutic index and exceeding optimal anticoagulation (measured by the international normalised ratio, INR) increases the risk of bleeding, necessitating frequent monitoring and dose titration (Landefeld and Beyth, Reference Landefeld and Beyth1993). One study found hospitalisation due to bleeding and supra-therapeutic INRs was seen in 6–7% of patients prescribed warfarin (Hylek et al., Reference Hylek, Evans-Molina, Shea, Henault and Regan2007; Lau et al., Reference Lau, Li, Wong, Man, Lip, Leung, Siu and Chan2017), while conversely, decreased time in the therapeutic INR range (TTR) was associated with increased ischaemic stroke, other thromboembolic events and mortality (Jones et al., Reference Jones, McEwan, Morgan, Peters, Goodfellow and Currie2005; Cancino et al., Reference Cancino, Hylek, Reisman and Rose2014).

There is substantial interpatient variability in warfarin response, with warfarin doses necessary to attain target INR ranging from <1 mg/day to >10 mg/day (stable dosing after loading dose) (Pokorney et al., Reference Pokorney, Simon, Thomas, Fonarow, Kowey, Chang, Singer, Ansell, Blanco, Gersh, Mahaffey, Hylek, Go, Piccini and Peterson2015). Genetic variation accounts for 55–60% of this dose variability: VKORC1 (∼25%), CYP2C9 (∼15%), CYP4F2*3 (∼1–7%) (Zhou et al., Reference Zhou, Nevosadova, Eliasson and Lauschke2023). Non-genetic factors collectively account for <20%: age, body mass index (BMI), smoking and drug interactions (Rost et al., Reference Rost, Fregin, Ivaskevicius, Conzelmann, Hortnagel, Pelz, Lappegard, Seifried, Scharrer, Tuddenham, Muller, Strom and Oldenburg2004; Wadelius et al., Reference Wadelius, Chen, Lindh, Eriksson, Ghori, Bumpstead, Holm, McGinnis, Rane and Deloukas2009; Verhoef et al., Reference Verhoef, Redekop, Daly, van Schie, de Boer and Maitland-van der Zee2014; Bourgeois et al., Reference Bourgeois, Jorgensen, Zhang, Hanson, Gillman, Bumpstead, Toh, Williamson, Daly, Kamali, Deloukas and Pirmohamed2016).

The CYP2C9*2, *3, *5, *6, *8 and *11 alleles reduce clearance of the more active S-warfarin, thus decreasing dose requirements by 5–7 mg/week in those carrying *2, *8 and *11 alleles, and 14 mg/week reported for the *3 and *5 alleles. Consequently, these variants are also associated with increased risk of over-anticoagulation. The *2 and *3 alleles are common among Europeans, while the *5, *6, *8 and *11 alleles occur almost exclusively in African ancestry populations (Johnson et al., Reference Johnson, Caudle, Gong, Whirl-Carrillo, Stein, Scott, Lee, Gage, Kimmel, Perera, Anderson, Pirmohamed, Klein, Limdi, Cavallari and Wadelius2017; Zhou et al., Reference Zhou, Nevosadova, Eliasson and Lauschke2023).

VKORC1 regulatory variant c.−1639G>A (rs9923231) is associated with reduced VKORC1 expression and lower warfarin dose requirements, with the −1,639 AA (high sensitivity) genotype more common among Asians and the −1,639 GG (reduced sensitivity) genotype more common among Africans (Limdi et al., Reference Limdi, Wadelius, Cavallari, Eriksson, Crawford, Lee, Chen, Motsinger-Reif, Sagreiya, Liu, Wu, Gage, Jorgensen, Pirmohamed, Shin, Suarez-Kurtz, Kimmel, Johnson, Klein and Wagner2010; Johnson et al., Reference Johnson, Caudle, Gong, Whirl-Carrillo, Stein, Scott, Lee, Gage, Kimmel, Perera, Anderson, Pirmohamed, Klein, Limdi, Cavallari and Wadelius2017; Zhou and Lauschke, Reference Zhou and Lauschke2022). Consequently, warfarin dose requirements are, respectively, lower and higher in Asian and African ancestry patients, respectively, as compared to Europeans (Limdi et al., Reference Limdi, Wadelius, Cavallari, Eriksson, Crawford, Lee, Chen, Motsinger-Reif, Sagreiya, Liu, Wu, Gage, Jorgensen, Pirmohamed, Shin, Suarez-Kurtz, Kimmel, Johnson, Klein and Wagner2010).

The CYP4F2 enzyme contributes to the variation in warfarin dose requirements not by metabolising warfarin, but rather by metabolising 75–90% of all vitamin K consumed by humans. Vitamin K1 reduction to vitamin K hydroquinone is critical to clotting factor activation. The *3 allele (rs2108622) is associated with reduced CYP4F2 activity resulting in higher concentrations of vitamin K1 and, consequently, higher warfarin dose requirements compared to the *1 allele, but this affects only European and Asian populations, with no impact on African ancestry individuals (Danese et al., Reference Danese, Raimondi, Montagnana, Tagetti, Langaee, Borgiani, Ciccacci, Carcas, Borobia, Tong, Davila-Fajardo, Rodrigues Botton, Bourgeois, Deloukas, Caldwell, Burmester, Berg, Cavallari, Drozda, Huang, Zhao, Cen, Gonzalez-Conejero, Roldan, Nakamura, Mushiroda, Gong, Kim, Hirai, Itoh, Isaza, Beltran, Jimenez-Varo, Canadas-Garre, Giontella, Kringen, Haug, Gwak, Lee, Minuz, Lee, Lubitz, Scott, Mazzaccara, Sacchetti, Genc, Ozer, Pathare, Krishnamoorthy, Paldi, Siguret, Loriot, Kutala, Suarez-Kurtz, Perini, Denny, Ramirez, Mittal, Rathore, Sagreiya, Altman, Shahin, Khalifa, Limdi, Rivers, Shendre, Dillon, Suriapranata, Zhou, Tan, Tatarunas, Lesauskaite, Zhang, Maitland-van der Zee, Verhoef, de Boer, Taljaard, Zambon, Pengo, Zhang, Pirmohamed, Johnson and Fava2019; Zhou and Lauschke, Reference Zhou and Lauschke2022).

While VKORC1 and CYP2C9 variants have emerged as the main genetic contributors to warfarin dose requirements in European and Asian ancestry populations (Cooper et al., Reference Cooper, Johnson, Langaee, Feng, Stanaway, Schwarz, Ritchie, Stein, Roden, Smith, Veenstra, Rettie and Rieder2008), the associations in African ancestry populations include single nucleotide polymorphisms (SNPs) in the chromosome 10 CYP2C cluster and in chromosome 6 upstream of EPHA7 (Perera et al., Reference Perera, Cavallari, Limdi, Gamazon, Konkashbaev, Daneshjou, Pluzhnikov, Crawford, Wang, Liu, Tatonetti, Bourgeois, Takahashi, Bradford, Burkley, Desnick, Halperin, Khalifa, Langaee, Lubitz, Nutescu, Oetjens, Shahin, Patel, Sagreiya, Tector, Weck, Rieder, Scott, Wu, Burmester, Wadelius, Deloukas, Wagner, Mushiroda, Kubo, Roden, Cox, Altman, Klein, Nakamura and Johnson2013; De et al., Reference De, Alarcon, Hernandez, Liko, Cavallari, Duarte and Perera2018; Zhou and Lauschke, Reference Zhou and Lauschke2022).

Validation of PGx-based warfarin dosing

The complexity of estimating initial warfarin dosing has been significantly diminished by the development of dosing algorithms, which take into account not only an individual’s clinical features (e.g., age, BMI and use of CYP2C9 inhibiting drugs), but also their genotype (VKORC1 −1639G>A, CYP2C9*2 and CYP2C9*3 alleles) (Gage et al., Reference Gage, Eby, Johnson, Deych, Rieder, Ridker, Milligan, Grice, Lenzini, Rettie, Aquilante, Grosso, Marsh, Langaee, Farnett, Voora, Veenstra, Glynn, Barrett and McLeod2008; International Warfarin Pharmacogenetics et al., Reference Klein, Altman, Eriksson, Gage, Kimmel, Lee, Limdi, Page, Roden, Wagner, Caldwell and Johnson2009). However, CYP2C9*5, *6, *8, *11 and rs12777823 are not represented in the algorithms significantly reducing their utility in patients of African ancestry. The Gage algorithm incorporates CYP2C9*5, *6 and CYP4F2*3 allele (Gage et al., Reference Gage, Eby, Johnson, Deych, Rieder, Ridker, Milligan, Grice, Lenzini, Rettie, Aquilante, Grosso, Marsh, Langaee, Farnett, Voora, Veenstra, Glynn, Barrett and McLeod2008; International Warfarin Pharmacogenetics et al., Reference Klein, Altman, Eriksson, Gage, Kimmel, Lee, Limdi, Page, Roden, Wagner, Caldwell and Johnson2009).

Three large multi-site RCTs (EU-PACT, COAG and GIFT) have evaluated the efficacy of genotype-guided warfarin dosing (Kimmel et al., Reference Kimmel, French, Kasner, Johnson, Anderson, Gage, Rosenberg, Eby, Madigan, McBane, Abdel-Rahman, Stevens, Yale, Mohler, Fang, Shah, Horenstein, Limdi, Muldowney, Gujral, Delafontaine, Desnick, Ortel, Billett, Pendleton, Geller, Halperin, Goldhaber, Caldwell, Califf, Ellenberg and Investigators2013; Pirmohamed et al., Reference Pirmohamed, Burnside, Eriksson, Jorgensen, Toh, Nicholson, Kesteven, Christersson, Wahlstrom, Stafberg, Zhang, Leathart, Kohnke, Maitland-van der Zee, Williamson, Daly, Avery, Kamali and Wadelius2013; Gage et al., Reference Gage, Bass, Lin, Woller, Stevens, Al-Hammadi, Li, Rodriguez, Miller, McMillin, Pendleton, Jaffer, King, Whipple, Porche-Sorbet, Napoli, Merritt, Thompson, Hyun, Anderson, Hollomon, Barrack, Nunley, Moskowitz, Davila-Roman and Eby2017) incorporating VKORC1 −1639G>A and CYP2C9*2 and *3 variants in a PGx algorithm, with CYP4F2 additionally included in the GIFT trial (Gage et al., Reference Gage, Bass, Lin, Woller, Stevens, Al-Hammadi, Li, Rodriguez, Miller, McMillin, Pendleton, Jaffer, King, Whipple, Porche-Sorbet, Napoli, Merritt, Thompson, Hyun, Anderson, Hollomon, Barrack, Nunley, Moskowitz, Davila-Roman and Eby2017). The primary endpoint was TTR for the EU-PACT and COAG trials (Kimmel et al., Reference Kimmel, French, Kasner, Johnson, Anderson, Gage, Rosenberg, Eby, Madigan, McBane, Abdel-Rahman, Stevens, Yale, Mohler, Fang, Shah, Horenstein, Limdi, Muldowney, Gujral, Delafontaine, Desnick, Ortel, Billett, Pendleton, Geller, Halperin, Goldhaber, Caldwell, Califf, Ellenberg and Investigators2013; Pirmohamed et al., Reference Pirmohamed, Burnside, Eriksson, Jorgensen, Toh, Nicholson, Kesteven, Christersson, Wahlstrom, Stafberg, Zhang, Leathart, Kohnke, Maitland-van der Zee, Williamson, Daly, Avery, Kamali and Wadelius2013) and clinical outcomes for the GIFT trial (Gage et al., Reference Gage, Bass, Lin, Woller, Stevens, Al-Hammadi, Li, Rodriguez, Miller, McMillin, Pendleton, Jaffer, King, Whipple, Porche-Sorbet, Napoli, Merritt, Thompson, Hyun, Anderson, Hollomon, Barrack, Nunley, Moskowitz, Davila-Roman and Eby2017). PGx-guided dosing showed significant improvement in the primary endpoints for EU-PACT and GIFT, but not COAG trials. EU-PACT (Pirmohamed et al., Reference Pirmohamed, Burnside, Eriksson, Jorgensen, Toh, Nicholson, Kesteven, Christersson, Wahlstrom, Stafberg, Zhang, Leathart, Kohnke, Maitland-van der Zee, Williamson, Daly, Avery, Kamali and Wadelius2013) compared genotype-guided warfarin dosing on days 1–5 followed by routine practice to routine practice. At 12 weeks, TTR was 7% higher in the genotype-guided arm (67.4% vs. 60.3%, P < 0.001). Conversely, TTR was similar in both the genotype-guided and clinically guided dosing arms of the COAG trial (4-week TTR 45.2% vs. 45.4%) (Kimmel et al., Reference Kimmel, French, Kasner, Johnson, Anderson, Gage, Rosenberg, Eby, Madigan, McBane, Abdel-Rahman, Stevens, Yale, Mohler, Fang, Shah, Horenstein, Limdi, Muldowney, Gujral, Delafontaine, Desnick, Ortel, Billett, Pendleton, Geller, Halperin, Goldhaber, Caldwell, Califf, Ellenberg and Investigators2013). In the GIFT trial (Gage et al., Reference Gage, Bass, Lin, Woller, Stevens, Al-Hammadi, Li, Rodriguez, Miller, McMillin, Pendleton, Jaffer, King, Whipple, Porche-Sorbet, Napoli, Merritt, Thompson, Hyun, Anderson, Hollomon, Barrack, Nunley, Moskowitz, Davila-Roman and Eby2017), the primary composite endpoint (INR ≥ 4, 30-day major bleeding, 30-day mortality death, 60-day incident venous thromboembolism) was lower in the genotype-guided group (10.8% vs. 14.7%, P = 0.02). Participants included in both the EU-PACT and GIFT trials were predominantly European. Although 27% of the COAG trial participants were African American, only the CYP2C9 alleles common in Caucasians (*2 and *3) were genotyped. Thus, all the three trials were blind to African ancestry-specific variants, and failure to account for these variants resulted in substantial warfarin overdosing in African American participants in the genotype-guided arm of COAG (Kimmel et al., Reference Kimmel, French, Kasner, Johnson, Anderson, Gage, Rosenberg, Eby, Madigan, McBane, Abdel-Rahman, Stevens, Yale, Mohler, Fang, Shah, Horenstein, Limdi, Muldowney, Gujral, Delafontaine, Desnick, Ortel, Billett, Pendleton, Geller, Halperin, Goldhaber, Caldwell, Califf, Ellenberg and Investigators2013). The reason is that CYP2C9*5, *6, *8 or *11 allele (present in ~15% of patients of African ancestry) or rs12777823 A allele (>40% of patients) may be misclassified as normal metabolisers (e.g., *1/*1) and dosed accordingly (Drozda et al., Reference Drozda, Wong, Patel, Bress, Nutescu, Kittles and Cavallari2015).

Patients with two or more CYP2C9 or VKORC1 variants are more prone to rapid INR surges and supratherapeutic anticoagulation at warfarin initiation. This may explain the differences between EU-PACT which used a loading dose and COAG which did not (Arwood et al., Reference Arwood, Deng, Drozda, Pugach, Nutescu, Schmidt, Duarte and Cavallari2017).

Clinical implementation of warfarin PGx

CYP2C9*2, *3, *5, *6, *8, *11, and VKORC1 −1639G>A alleles (Pratt et al., Reference Pratt, Cavallari, Del Tredici, Hachad, Ji, Kalman, Ly, Moyer, Scott, Whirl-Carrillo and Weck2020) are the minimum set of panel variants supported by cost-effectiveness data on the implementation of multigene genotype-guided warfarin dosing (Zhu et al., Reference Zhu, Swanson, Rojas, Wang, St Sauver, Visscher, Prokop, Bielinski, Wang, Weinshilboum and Borah2020). Both the FDA and Dutch Pharmacogenetics Working Group (DPWG) genotype-guided dosing recommendations are limited to just VKORC1 −1639G>A and CYP2C9*2 and *3 alleles. CPIC, in contrast, provides African and non-African specific guidance, with the former requiring CYP2C9*5, *6, *8 and *11 genotypes, and the latter requiring on CYP2C9*2 and *3 and VKORC1 genotypes (Johnson et al., Reference Johnson, Caudle, Gong, Whirl-Carrillo, Stein, Scott, Lee, Gage, Kimmel, Perera, Anderson, Pirmohamed, Klein, Limdi, Cavallari and Wadelius2017). Presence of CYP4F2*3 allele in non-African individuals results in a 5–10% dose increase. For those of African ancestry, rs12777823 variant, if available, results in an additional 15–30% dose reduction (Johnson et al., Reference Johnson, Caudle, Gong, Whirl-Carrillo, Stein, Scott, Lee, Gage, Kimmel, Perera, Anderson, Pirmohamed, Klein, Limdi, Cavallari and Wadelius2017).

Clopidogrel

Antiplatelet therapy is a cornerstone of atherosclerotic CVD management involving aspirin or a P2Y12 receptor antagonist (clopidogrel, prasugrel and ticagrelor), either as single agent therapy for secondary prevention or dual agents after percutaneous coronary intervention (PCI) (Roffi et al., Reference Roffi, Patrono, Collet, Mueller, Valgimigli, Andreotti, Bax, Borger, Brotons, Chew, Gencer, Hasenfuss, Kjeldsen, Lancellotti, Landmesser, Mehilli, Mukherjee, Storey and Windecker2016; Ibanez et al., Reference Ibanez, James, Agewall, Antunes, Bucciarelli-Ducci, Bueno, ALP, Crea, Goudevenos, Halvorsen, Hindricks, Kastrati, Lenzen, Prescott, Roffi, Valgimigli, Varenhorst, Vranckx and Widimsky2018). Prasugrel and ticagrelor are more potent P2Y12 receptor antagonists with an increased bleeding risk but are preferred over clopidogrel in high-risk cases (Wallentin et al., Reference Wallentin, Becker, Budaj, Cannon, Emanuelsson, Held, Horrow, Husted, James, Katus, Mahaffey, Scirica, Skene, Steg, Storey, Harrington, Investigators, Freij and Thorsen2009). Genetic variation is partly responsible for the observed variability in effectiveness of antiplatelet therapy (Angiolillo et al., Reference Angiolillo, Rollini, Storey, Bhatt, James, Schneider, Sibbing, So, Trenk, Alexopoulos, Gurbel, Hochholzer, De Luca, Bonello, Aradi, Cuisset, Tantry, Wang, Valgimigli, Waksman, Mehran, Montalescot, Franchi and Price2017). Assessment of platelet function status is time-consuming, lacks standard reference values and is hence not clinically feasible for tailoring antiplatelet therapy. The prospect of a genotype profile providing a measure of antiplatelet efficacy and thus predicting adverse cardiovascular outcomes makes a compelling case for the use of PGx to personalise treatment.

Clopidogrel, the most commonly prescribed antiplatelet drug, is a prodrug that undergoes a two-step transformation to its active metabolite which irreversibly inhibits platelet activation (Kazui et al., Reference Kazui, Nishiya, Ishizuka, Hagihara, Farid, Okazaki, Ikeda and Kurihara2010). CYP2C19 is involved in both activation steps, and thus, plays a crucial role in the bioactivation process of clopidogrel (Sangkuhl et al., Reference Sangkuhl, Klein and Altman2010). CYP2C19 is highly polymorphic with alleles representing a range of metaboliser phenotypes (summarised in Table 1; Kazui et al., Reference Kazui, Nishiya, Ishizuka, Hagihara, Farid, Okazaki, Ikeda and Kurihara2010; Sangkuhl et al., Reference Sangkuhl, Klein and Altman2010; Scott et al., Reference Scott, Sangkuhl, Stein, Hulot, Mega, Roden, Klein, Sabatine, Johnson, Shuldiner and Implementation2013; Pratt et al., Reference Pratt, Del Tredici, Hachad, Ji, Kalman, Scott and Weck2018; Zhou and Lauschke, Reference Zhou and Lauschke2022; Zhou et al., Reference Zhou, Nevosadova, Eliasson and Lauschke2023).

Table 1. CYP2C19 allele dependent enzyme activity

Abbreviations: EM, extensive metaboliser; IM, intermediate metaboliser; PM, poor metaboliser; UM, ultrarapid metaboliser.

The CYP2C19 poor metaboliser (PM) and intermediate metaboliser (IM) phenotypes have higher on-treatment platelet reactivity and an increased risk of ischaemic events compared to the normal metaboliser (NM) phenotype (*1/*1 genotype) (Varenhorst et al., Reference Varenhorst, James, Erlinge, Brandt, Braun, Man, Siegbahn, Walker, Wallentin, Winters and Close2009; Mega et al., Reference Mega, Close, Wiviott, Shen, Hockett, Brandt, Walker, Antman, Macias, Braunwald and Sabatine2009a). The equivalent of a 75 mg dose of clopidogrel in NMs is 225 mg in IMs, but 300 mg is insufficient in PMs (Mega et al., Reference Mega, Hochholzer, Frelinger, Kluk, Angiolillo, Kereiakes, Isserman, Rogers, Ruff, Contant, Pencina, Scirica, Longtine, Michelson and Sabatine2011; Price et al., Reference Price, Murray, Angiolillo, Lillie, Smith, Tisch, Schork, Teirstein, Topol and Investigators2012; Carreras et al., Reference Carreras, Hochholzer, Frelinger, Nordio, O’Donoghue, Wiviott, Angiolillo, Michelson, Sabatine and Mega2016). The antiplatelet drugs prasugrel and ticagrelor are not affected by the CYP2C19 genotype, offering the option for switching IMs and PMs to these drugs in preference to clopidogrel dose escalation in the absence of contraindications (Varenhorst et al., Reference Varenhorst, James, Erlinge, Brandt, Braun, Man, Siegbahn, Walker, Wallentin, Winters and Close2009; Mega et al., Reference Mega, Close, Wiviott, Shen, Hockett, Brandt, Walker, Antman, Macias, Braunwald and Sabatine2009b; Wallentin et al., Reference Wallentin, James, Storey, Armstrong, Barratt, Horrow, Husted, Katus, Steg, Shah, Becker and investigators2010).

Several real-world studies showed a significantly higher risk of major adverse cardiovascular events (MACE) in CYP2C19 PMs and IMs compared to NMs (Hulot et al., Reference Hulot, Collet, Silvain, Pena, Bellemain-Appaix, Barthelemy, Cayla, Beygui and Montalescot2010; Mega et al., Reference Mega, Simon, Collet, Anderson, Antman, Bliden, Cannon, Danchin, Giusti, Gurbel, Horne, Hulot, Kastrati, Montalescot, Neumann, Shen, Sibbing, Steg, Trenk, Wiviott and Sabatine2010; Holmes et al., Reference Holmes, Perel, Shah, Hingorani and Casas2011; Zabalza et al., Reference Zabalza, Subirana, Sala, Lluis-Ganella, Lucas, Tomas, Masia, Marrugat, Brugada and Elosua2012; Sorich et al., Reference Sorich, Rowland, McKinnon and Wiese2014; Cavallari et al., Reference Cavallari, Lee, Beitelshees, Cooper-DeHoff, Duarte, Voora, Kimmel, McDonough, Gong, Dave, Pratt, Alestock, Anderson, Alsip, Ardati, Brott, Brown, Chumnumwat, Clare-Salzler, Coons, Denny, Dillon, Elsey, Hamadeh, Harada, Hillegass, Hines, Horenstein, Howell, Jeng, Kelemen, Lee, Magvanjav, Montasser, Nelson, Nutescu, Nwaba, Pakyz, Palmer, Peterson, Pollin, Quinn, Robinson, Schub, Skaar, Smith, Sriramoju, Starostik, Stys, Stevenson, Varunok, Vesely, Wake, Weck, Weitzel, Wilke, Willig, Zhao, Kreutz, Stouffer, Empey, Limdi, Shuldiner, Winterstein, Johnson and Network2018; Kheiri et al., Reference Kheiri, Simpson, Osman, Kumar, Przybylowicz, Merrill, Golwala, Rahmouni, Cigarroa and Zahr2020). However, the higher risk of MACE in clopidogrel-treated PMs and IMs was less evident in lower-risk populations, such as atrial fibrillation or medically managed acute coronary syndrome (ACS) cases (Bauer et al., Reference Bauer, Bouman, van Werkum, Ford, ten Berg and Taubert2011; Holmes et al., Reference Holmes, Perel, Shah, Hingorani and Casas2011). Two prospective trials, POPular Genetics (Claassens et al., Reference Claassens, Vos, Hermanides, van’t Hof, van der Harst, Barbato, Morisco, Tjon Joe Gin, Asselbergs, Mosterd, Herrman, WJM, PWA, Kelder, Postma, de Boer, Boersma, VHM and Ten Berg2019) and TAILOR-PCI (Pereira et al., Reference Pereira, Farkouh, So, Lennon, Geller, Mathew, Bell, Bae, Jeong, Chavez, Gordon, Abbott, Cagin, Baudhuin, Fu, Goodman, Hasan, Iturriaga, Lerman, Sidhu, Tanguay, Wang, Weinshilboum, Welsh, Rosenberg, Bailey and Rihal2020) trials stratified IMs and PMs to prasugrel or ticagrelor while NMs received clopidogrel. The CYP2C19-guided approach reduced bleeding risk and was non-inferior to treatment with prasugrel or ticagrelor in preventing atherothrombotic events in the POPular Genetics study that enrolled post ST-segment elevation MI patients undergoing PCI (Claassens et al., Reference Claassens, Vos, Hermanides, van’t Hof, van der Harst, Barbato, Morisco, Tjon Joe Gin, Asselbergs, Mosterd, Herrman, WJM, PWA, Kelder, Postma, de Boer, Boersma, VHM and Ten Berg2019). In the TAILOR-PCI trial (Pereira et al., Reference Pereira, Farkouh, So, Lennon, Geller, Mathew, Bell, Bae, Jeong, Chavez, Gordon, Abbott, Cagin, Baudhuin, Fu, Goodman, Hasan, Iturriaga, Lerman, Sidhu, Tanguay, Wang, Weinshilboum, Welsh, Rosenberg, Bailey and Rihal2020), patients with either stable coronary disease or ACS undergoing PCI showed lower rates of the composite cardiovascular primary endpoint in the genotype-guided group compared to the non-genotype-guided cohort at 1-year follow-up, but this did not reach statistical significance (HR 0.66; 95% CI 0.43–1.02; P = 0.06) (Pereira et al., Reference Pereira, Farkouh, So, Lennon, Geller, Mathew, Bell, Bae, Jeong, Chavez, Gordon, Abbott, Cagin, Baudhuin, Fu, Goodman, Hasan, Iturriaga, Lerman, Sidhu, Tanguay, Wang, Weinshilboum, Welsh, Rosenberg, Bailey and Rihal2020). A post hoc analysis indicated benefit in the genotype-directed group during the first 3 months after PCI (HR 0.21; 95% CI 0.08–0.54; P = 0.001) (Pereira et al., Reference Pereira, Farkouh, So, Lennon, Geller, Mathew, Bell, Bae, Jeong, Chavez, Gordon, Abbott, Cagin, Baudhuin, Fu, Goodman, Hasan, Iturriaga, Lerman, Sidhu, Tanguay, Wang, Weinshilboum, Welsh, Rosenberg, Bailey and Rihal2020). Other indications for clopidogrel include stroke prevention and peripheral arterial disease. PMs and IMs show reduced rates of stent patency after endovascular treatment for peripheral arterial disease (Guo et al., Reference Guo, Tan, Guo, Shi, Zhang and Guo2014; Diaz-Villamarin et al., Reference Diaz-Villamarin, Davila-Fajardo, Martinez-Gonzalez, Carmona-Saez, Sanchez-Ramos, Alvarez Cubero, Salmeron-Febres, Cabeza Barrera and Fernandez-Quesada2016). For stroke, a large randomised controlled trial (RCT) showed that absence of the CYP2C19 no-function allele in patients with a minor ischaemic stroke or transient ischaemic attack (TIA) predicted better effectiveness of clopidogrel plus aspirin over aspirin alone (Wang et al., Reference Wang, Zhao, Lin, Li, Johnston, Lin, Pan, Liu, Wang, Wang, Meng, Xu, Wang and investigators2016). A meta-analysis including nearly 5,000 clopidogrel-treated patients with ischaemic stroke or TIA confirmed higher risk of new stroke in PMs and IMs (Pan et al., Reference Pan, Chen, Xu, Yi, Han, Yang, Li, Huang, Johnston, Zhao, Liu, Zhang, Wang, Wang and Wang2017).

Clinical implementation of clopidogrel PGx

Since 2010, the FDA, European Medicine Agency (EMA) and other regulatory bodies recommend alternative P2Y12 inhibitors to clopidogrel in PMs (but not IMs) in their labels (Holmes et al., Reference Holmes, Dehmer, Kaul, Leifer, O’Gara and Stein2010). The FDA table of gene–drug pairs includes therapeutic management recommendations for IMs and PMs (FDA, 2023a, 2023b), which is echoed by CPIC guidelines citing ‘strong’ evidence for IMs and PMs with ACS or undergoing PCI, and ‘moderate’ evidence PMs for all indications. In all of the above cases, alternative antiplatelet agents are recommended (Lee et al., Reference Lee, Luzum, Sangkuhl, Gammal, Sabatine, Stein, Kisor, Limdi, Lee, Scott, Hulot, Roden, Gaedigk, Caudle, Klein, Johnson and Shuldiner2022).

Joint PCI guidelines from 2016 by the American College of Cardiology (ACC) and the American Heart Association (AHA) recommend against routine genotyping for all patients undergoing PCI, but to consider testing high-risk patients and use either prasugrel or ticagrelor for patients with the no-function allele. The 2020 European Society of Cardiology (ESC) guidelines were influenced by the POPular Genetics trial to recommend genotype-guided de-escalation for post-PCI patients deemed to be at high bleeding risk (Claassens and Sibbing, Reference Claassens and Sibbing2020; Collet et al., Reference Collet, Thiele, Barbato, Barthelemy, Bauersachs, Bhatt, Dendale, Dorobantu, Edvardsen, Folliguet, Gale, Gilard, Jobs, Juni, Lambrinou, Lewis, Mehilli, Meliga, Merkely, Mueller, Roffi, Rutten, Sibbing and GCM2021).

CYP2C19-guided antiplatelet therapy after PCI is one of the most common PGx tests in clinical practice (Empey et al., Reference Empey, Stevenson, Tuteja, Weitzel, Angiolillo, Beitelshees, Coons, Duarte, Franchi, Jeng, Johnson, Kreutz, Limdi, Maloney, Owusu Obeng, Peterson, Petry, Pratt, Rollini, Scott, Skaar, Vesely, Stouffer, Wilke, Cavallari, Lee and Network2018) conducted either for patients at high risk of MACE in line with ACC/AHA guidelines or for all-comers (Empey et al., Reference Empey, Stevenson, Tuteja, Weitzel, Angiolillo, Beitelshees, Coons, Duarte, Franchi, Jeng, Johnson, Kreutz, Limdi, Maloney, Owusu Obeng, Peterson, Petry, Pratt, Rollini, Scott, Skaar, Vesely, Stouffer, Wilke, Cavallari, Lee and Network2018). If point-of-care genotyping is not available, a de-escalation approach is proposed where patients are commenced on prasugrel or ticagrelor initially pending genotype results and then switched to clopidogrel if the genotype results indicate the NM phenotype. This approach maximises benefit given the high risk of atherothrombotic events early after ACS and PCI, while reducing the high risk of bleeding with prasugrel and ticagrelor during long-term therapy (Becker et al., Reference Becker, Bassand, Budaj, Wojdyla, James, Cornel, French, Held, Horrow, Husted, Lopez-Sendon, Lassila, Mahaffey, Storey, Harrington and Wallentin2011; Rollini et al., Reference Rollini, Franchi and Angiolillo2016; Angiolillo et al., Reference Angiolillo, Rollini, Storey, Bhatt, James, Schneider, Sibbing, So, Trenk, Alexopoulos, Gurbel, Hochholzer, De Luca, Bonello, Aradi, Cuisset, Tantry, Wang, Valgimigli, Waksman, Mehran, Montalescot, Franchi and Price2017). The case for implementing pre-emptive CYP2C19 genotyping (Peterson et al., Reference Peterson, Field, Unertl, Schildcrout, Johnson, Shi, Danciu, Cleator, Pulley, McPherson, Denny, Laposata, Roden and Johnson2016) is evident due to the impact of CYP2C19 genotype on other drugs in addition to clopidogrel, such as proton pump inhibitors (Lima et al., Reference Lima, Thomas, Barbarino, Desta, Van Driest, El Rouby, Johnson, Cavallari, Shakhnovich, Thacker, Scott, Schwab, Uppugunduri, Formea, Franciosi, Sangkuhl, Gaedigk, Klein, Gammal and Furuta2021) and selective serotonin reuptake inhibitors (SSRIs) (Hicks et al., Reference Hicks, Bishop, Sangkuhl, Muller, Ji, Leckband, Leeder, Graham, Chiulli, LLerena, Skaar, Scott, Stingl, Klein, Caudle and Gaedigk2015).

Direct-acting oral anti-coagulants

Apixaban, dabigatran, edoxaban and rivaroxaban are direct-acting oral anticoagulants (DOACs) with several advantages compared to warfarin – wider therapeutic index, regular monitoring not required, lower risk of intracranial haemorrhage, stroke or systemic embolic events (Proietti et al., Reference Proietti, Romanazzi, Romiti, Farcomeni and Lip2018). Despite the favourable profile of DOACs, their higher cost, lower adherence rates, limited indications, and the high cost of reversal agents has limited uptake of DOAC compared to warfarin (Zhu et al., Reference Zhu, Alexander, Nazarian, Segal and Wu2018; Ho et al., Reference Ho, van Hove and Leng2020). Pharmacokinetic variation related to genetic variation is indicated but there is no data on clinical outcomes yet.

In a sub-study of the ENGAGE AF TIMI-48 trial (which compared warfarin and edoxaban in atrial fibrillation patients; Mega et al., Reference Mega, Walker, Ruff, Vandell, Nordio, Deenadayalu, Murphy, Lee, Mercuri, Giugliano, Antman, Braunwald and Sabatine2015) warfarin-treated participants with a sensitive or highly sensitive genotype (e.g., VKORC1 −1639AA or CYP2C9*1/*3) spent a greater proportion of time within the supratherapeutic INR range (i.e., INR >4) and had higher rates of bleeding in the initial 90 days of treatment, as compared to those with non-sensitive genotypes. In a genetic sub-study of the RE-LY trial (dabigatran versus warfarin in atrial fibrillation), carriers of the CES1 rs2244613 minor allele had a reduced risk of bleeding with dabigatran than with warfarin (Shi et al., Reference Shi, Wang, Nguyen, Bleske, Liang, Liu and Zhu2016).

Statins

Lipid lowering treatment by statins (HMG-CoA reductase inhibitors) are used in the prevention of CVD (Catapano et al., Reference Catapano, Graham, De Backer, Wiklund, Chapman, Drexel, Hoes, Jennings, Landmesser, Pedersen, Reiner, Riccardi, Taskinen, Tokgozoglu, WMM, Vlachopoulos, Wood, Zamorano and Cooney2016). Statin-associated muscle symptoms (SAMS) (range from mild myalgia without an elevation in creatine kinase to life-threatening rhabdomyolysis or autoimmune-necrotizing myositis) are the commonest reasons for treatment discontinuation (Alfirevic et al., Reference Alfirevic, Neely, Armitage, Chinoy, Cooper, Laaksonen, Carr, Bloch, Fahy, Hanson, Yue, Wadelius, Maitland-van Der Zee, Voora, Psaty, Palmer and Pirmohamed2014). A number of enzymes and transporters are responsible for intracellular skeletal myocyte entry that underlie disruption of muscle function leading to SAMS (Turner and Pirmohamed, Reference Turner and Pirmohamed2019). Hepatic uptake and elimination of statins are mainly carried out by the solute carrier anion transporter family 1B1 gene (SLCO1B1) encoding the organic anion transporting polypeptide 1B1 (OATP1B1) (Shitara, Reference Shitara2011). The rs4149056 SNP in the SLCO1B1 gene (SLCO1B1*5) is linked to OATP1B1 function (Tirona et al., Reference Tirona, Leake, Merino and Kim2001) with the C allele being associated with decreased OATP1B1 transporter function with greatest reduction in homozygous patients resulting in significantly increased plasma concentrations of all statins, except fluvastatin (Tirona et al., Reference Tirona, Leake, Merino and Kim2001). Additionally, the risk of myopathy increases by 2.6 and 4.3 per copy of SLCO1B1*5 in patients, respectively, on simvastatin 40 mg and 80 mg daily (Tirona et al., Reference Tirona, Leake, Merino and Kim2001). The mechanism of SLCO1B1*5 variant causing statin-related myopathy is through the accumulation of circulating simvastatin acid (the active form of simvastatin) reflecting liver transport (Choi et al., Reference Choi, Bae, Cho, Ghim, Choe, Jung, Jin, Kim and Lim2015). This effect is most prominent for simvastatin followed by pitavastatin, lovastatin and atorvastatin (Ramsey et al., Reference Ramsey, Johnson, Caudle, Haidar, Voora, Wilke, Maxwell, McLeod, Krauss, Roden, Feng, Cooper-DeHoff, Gong, Klein, Wadelius and Niemi2014). Each copy of the C allele of rs4149056 increases the risk of statin-induced myopathy threefold in genome-wide association studies (GWAS) (Carr et al., Reference Carr, Francis, Jorgensen, Zhang, Chinoy, Heckbert, Bis, Brody, Floyd, Psaty, Molokhia, Lapeyre-Mestre, Conforti, Alfirevic, van Staa and Pirmohamed2019). Atorvastatin is partially metabolised by the CYP3A and UDP-glucuronosyltransferase 1A1 (UGT1A) enzyme families. One study showed the SNP rs45446698 just upstream of CYP3A7 and another, rs887829, located in multiple overlapping UGT1A genes, to be associated with atorvastatin-to-metabolite ratios in patients with ACS (Turner et al., Reference Turner, Fontana, Zhang, Carr, Yin, FitzGerald, Morris and Pirmohamed2020). Inconsistent associations with SAMS have been reported for polymorphisms in CYP3A4, ABCB1, COQ2 (involved in coenzyme Q10 synthesis) and GATM (involved in creatine synthesis) (Fiegenbaum et al., Reference Fiegenbaum, da Silveira, Van der Sand, Van der Sand, Ferreira, Pires and Hutz2005; Hoenig et al., Reference Hoenig, Walker, Gurnsey, Beadle and Johnson2011; Mangravite et al., Reference Mangravite, Engelhardt, Medina, Smith, Brown, Chasman, Mecham, Howie, Shim, Naidoo, Feng, Rieder, Chen, Rotter, Ridker, Hopewell, Parish, Armitage, Collins, Wilke, Nickerson, Stephens and Krauss2013; Carr et al., Reference Carr, Francis, Jorgensen, Zhang, Chinoy, Heckbert, Bis, Brody, Floyd, Psaty, Molokhia, Lapeyre-Mestre, Conforti, Alfirevic, van Staa and Pirmohamed2019).

Validation of PGx-based statin dosing

The pragmatic SLCO1B1 genotype-informed statin therapy (GIST) trial randomised patients who had discontinued any statins due to myalgia to SLCO1B1 genotype guided therapy (rosuvastatin, pravastatin, or fluvastatin for SLCO1B1*5 carriers and any statin for non-carriers) or standard care (Peyser et al., Reference Peyser, Perry, Singh, Gill, Mehan, Haga, Musty, Milazzo, Savard, Li, Trujilio and Voora2018). At the end of 8-month follow-up, increased statin re-initiation, reduced LDL-C levels, and no change in self-reported medication adherence were seen in those randomised to genotype guided (Peyser et al., Reference Peyser, Perry, Singh, Gill, Mehan, Haga, Musty, Milazzo, Savard, Li, Trujilio and Voora2018). Whilst these results are interesting, the inclusion of patients who developed myopathy from any statins in the trial limits the translational potential of the results. This is because the impact of SLCO1B1 variation is highest for simvastatin and variable for other statins, hence the results of the trial do not present a clear case for genotype-guided simvastatin therapy.

Clinical implementation of statin PGx

The SLCO1B1*5 variant (rs4149056) shows wide population differences (1%, 8% and 16% in African, Asian and European populations, respectively). CPIC recommends not exceeding a dose of simvastatin 20 mg/day or, prescribing another statin (rosuvastatin or pravastatin) in patients who carry at least one rs4149056 C allele (Voora et al., Reference Voora, Shah, Spasojevic, Ali, Reed, Salisbury and Ginsburg2009; Danik et al., Reference Danik, Chasman, MacFadyen, Nyberg, Barratt and Ridker2013; Ramsey et al., Reference Ramsey, Johnson, Caudle, Haidar, Voora, Wilke, Maxwell, McLeod, Krauss, Roden, Feng, Cooper-DeHoff, Gong, Klein, Wadelius and Niemi2014; Lamoureux et al., Reference Lamoureux and Duflot2017). The French National Network of Pharmacogenetics recommends commencing statins in patients with risk factors for myopathy only after rs4149056 genotyping (Lamoureux et al., Reference Lamoureux and Duflot2017). The DPWG recommends that homozygotes avoid simvastatin entirely and individuals with other clinical risk factors for SAMS avoid atorvastatin (de Keyser et al., Reference de Keyser, Peters, Becker, Visser, Uitterlinden, Klungel, Verstuyft, Hofman, Maitland-van der Zee and Stricker2014; Bank et al., Reference Swen and Guchelaar2019; Linskey et al., Reference Linskey, English, Perry, Ochs-Balcom, Ma, Isackson, Vladutiu and Luzum2020; Turner et al., Reference Turner, Fontana, Zhang, Carr, Yin, FitzGerald, Morris and Pirmohamed2020).

Beta blockers

β-Adrenergic receptor antagonists, or beta blockers, are indicated for treatment of heart failure, hypertension, and secondary prevention of myocardial infarction. CYP2D6 is responsible for biotransformation of 70–80% of an oral dose of metoprolol and has negligible effects on other beta blockers (Ingelman-Sundberg et al., Reference Ingelman-Sundberg, Sim, Gomez and Rodriguez-Antona2007; Baudhuin et al., Reference Baudhuin, Miller, Train, Bryant, Hartman, Phelps, Larock and Jaffe2010; Blake et al., Reference Blake, Kharasch, Schwab and Nagele2013; Zisaki et al., Reference Zisaki, Miskovic and Hatzimanikatis2015; Vieira et al., Reference Vieira, Neves, Coelho and Lanchote2018). There is only weak evidence for PGx-guided prescribing of beta blockers (PharmGKB level 2–3, CPIC level B/C). Compared to EMs, IMs and PMs are associated with a decreased heart rate (Bijl et al., Reference Bijl, Visser, van Schaik, Kors, Witteman, Hofman, Vulto, van Gelder and Stricker2009; Batty et al., Reference Batty, Hall, White, Wikstrand, de Boer, van Veldhuisen, van der Harst, Waagstein, Hjalmarson, Kjekshus and Balmforth2014; Anstensrud et al., Reference Anstensrud, Molden, Haug, Qazi, Muriq, Fosshaug, Spigset and Oie2020) and lower diastolic BP (Bijl et al., Reference Bijl, Visser, van Schaik, Kors, Witteman, Hofman, Vulto, van Gelder and Stricker2009; Batty et al., Reference Batty, Hall, White, Wikstrand, de Boer, van Veldhuisen, van der Harst, Waagstein, Hjalmarson, Kjekshus and Balmforth2014; Hamadeh et al., Reference Hamadeh, Langaee, Dwivedi, Garcia, Burkley, Skaar, Chapman, Gums, Turner, Gong, Cooper-DeHoff and Johnson2014; Anstensrud et al., Reference Anstensrud, Molden, Haug, Qazi, Muriq, Fosshaug, Spigset and Oie2020). These studies have not studied the entire spectrum of major variations in CYP2D6 and have not been independently validated.

Three other genes (ADRB1, ADRB2 and GRK5) have been associated with the beta blocker pharmacodynamics rather than pharmacokinetics, but there is no evidence of clinical utility for using these variants to guide prescribing (White et al., Reference White, de Boer, Maqbool, Greenwood, van Veldhuisen, Cuthbert, Ball, Hall and Balmforth2003; Pacanowski et al., Reference Pacanowski, Gong, Cooper-Dehoff, Schork, Shriver, Langaee, Pepine, Johnson and Investigators2008; Magvanjav et al., Reference Magvanjav, McDonough, Gong, McClure, Talbert, Horenstein, Shuldiner, Benavente, Mitchell, Johnson and SiGN2017; Huang et al., Reference Huang, Li, Song, Fan, You, Tan, Xiao, Li, Ruan, Hu, Cui, Li, Ni, Chen, Woo, Xiao and Wang2018).

FDA and DPWG have slightly different recommendations on metoprolol dosing. The FDA recommends caution with co-administration of strong CYP2D6 inhibitors (SSRIs, antipsychotics) or substrates. The DPWG recommend cautious dose titration and reduced maximal doses in CYP2D6 IMs and PMs supramaximal metoprolol dose or an alternative beta blocker in UMs (Brouwer et al., Reference Brouwer, Nijenhuis, Soree, Guchelaar, Swen, van Schaik, Weide, Rongen, Buunk, de Boer-Veger, Houwink, van Westrhenen, Wilffert, VHM and Mulder2022).

Hydralazine

Hydralazine is a direct vasodilator seldom used in the treatment of hypertension (Whelton et al., Reference Whelton, Carey, Aronow, Casey, Collins, Dennison Himmelfarb, DePalma, Gidding, Jamerson, Jones, MacLaughlin, Muntner, Ovbiagele, Smith, Spencer, Stafford, Taler, Thomas, Williams, Williamson and Wright2018). Hydralazine is metabolised primarily by hepatic N-acetyltransferase type 2 (NAT2) acetylation. The common NAT2*4 genetic variant defines a ‘rapid acetylator’ phenotype with decreased hydralazine levels after drug administration (Gonzalez-Fierro et al., Reference Gonzalez-Fierro, Vasquez-Bahena, Taja-Chayeb, Vidal, Trejo-Becerril, Perez-Cardenas, de la Cruz-Hernandez, Chavez-Blanco, Gutierrez, Rodriguez, Fernandez and Duenas-Gonzalez2011; Han et al., Reference Han, Ryu, Cusumano, Easterling, Phillips, Risler, Shen and Hebert2019). Homozygous NAT2*5, *6, and *7 indicate a ‘slow acetylator’ phenotype, while heterozygous individuals (e.g., *4/*5) are ‘intermediate acetylators’. One study of resistant hypertension patients demonstrated that only those with the slow acetylator phenotype showed notable blood pressure reductions in response to hydralazine (Spinasse et al., Reference Spinasse, Santos, Suffys, Muxfeldt and Salles2014).

One of the rare side effects of hydralazine is the occurrence of lupus-like symptoms, with indirect evidence suggesting slow acetylators are more prone to developing this ADR (Weber and Hein, Reference Weber and Hein1985; Mazari et al., Reference Mazari, Ouarzane and Zouali2007; Schoonen et al., Reference Schoonen, Thomas, Somers, Smeeth, Kim, Evans and Hall2010). However, clinical utility and cost-effectiveness data are lacking.

Antiarrhythmic drugs

Inhibition of the rapid component of the delayed rectifier potassium current, I kr, encoded by KCNH2 is the commonest cause of drug induced long QT syndrome (LQTS) and torsades des pointes (TdP; ventricular tachycardia (Roden and Viswanathan, Reference Roden and Viswanathan2005; Wada et al., Reference Wada, Yang, Shaffer, Daniel, Glazer, Davogustto, Lowery, Farber-Eger, Wells and Roden2022).

Similar to beta blockers, the class 1 antiarrhythmic drugs flecainide and propafenone are metabolised by CYP2D6 (PharmGKB level 2A, CPIC level B/C; Doki et al., Reference Doki, Sekiguchi, Kuga, Aonuma and Homma2015; Rouini and Afshar, Reference Rouini and Afshar2017) with CYP2D6 genotype-related differences in QTc interval (Lim et al., Reference Lim, Jang, Kim, Kim, Jeon, Tae, Yi, Eum, Cho, Shin and Yu2010). The FDA recommends caution in the use of propafenone in patients with CYP2D6 deficiency when combined with CYP3A4 inhibition. The DPWG recommends a dose reduction of 50% and 30%, respectively, for flecainide and propafenone in CYP2D6 PMs.

Quinidine- or dofetilide-induced QT prolongation and drug-induced TdP was significantly associated with a polygenic risk score constructed from 61 SNPs excluding the CYP2D6 locus (Arking et al., Reference Arking, Pulit, Crotti, van der Harst, Munroe, Koopmann, Sotoodehnia, Rossin, Morley, Wang, Johnson, Lundby, Gudbjartsson, Noseworthy, Eijgelsheim, Bradford, Tarasov, Dorr, Muller-Nurasyid and Lahtinen2014; Strauss et al., Reference Strauss, Vicente, Johannesen, Blinova, Mason, Weeke, Behr, Roden, Woosley, Kosova, Rosenberg and Newton-Cheh2017). Though not validated, this highlights the potential for using polygenic risk scores in predicting drug-induced arrhythmias.

PGx implementation

Successful pharmacogenomic implementation in healthcare require strong scientific evidence, comprehensive and updated clinical guidelines, clinician champions and stakeholder engagement (Manolio et al., Reference Manolio, Chisholm, Ozenberger, Roden, Williams, Wilson, Bick, Bottinger, Brilliant, Eng, Frazer, Korf, Ledbetter, Lupski, Marsh, Mrazek, Murray, O’Donnell, Rader, Relling, Shuldiner, Valle, Weinshilboum, Green and Ginsburg2013).

Laboratory

Characterisation of pharmacogenomic variants in patients requires a certified molecular pathology laboratory to ensure analytical accuracy, precision, sensitivity and specificity of the results (Tayeh et al., Reference Tayeh, Gaedigk, Goetz, Klein, Lyon, McMillin, Rentas, Shinawi, Pratt and Scott2022). Most clinical PGx tests based on selected panel of clinically relevant variants (single gene or multigene) are more cost-effective than sequencing panels. It is likely that the decreasing cost of sequencing will make sequencing cost-competitive over multi-gene panels in the future (Figure 1).

Figure 1. Pharmacogenomic implementation. The top panel shows the range of stakeholders, technology, knowledge and evidence that need to be harnessed to realise the value of PGx. The middle panel depicts the uses of PGx in the clinical prescribing pathway. The bottom panel presents the applications of PGx. CPIC, the Clinical Pharmacogenetics Implementation Consortium; DPWG, Dutch Pharmacogenetics Working Groups; PharmGKB, the Pharmacogenomics Knowledge Base.

Guidelines and clinical decision support systems

Effective pharmacogenomic guided prescribing requires evidence from multiple sources to be distilled into guidelines and made available through clinical decision support systems (CDSS) that distil information on drug–gene interactions from published guidelines or prescribing labels. Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Groups (DPWG) have published guidelines covering 66 medications across several drug classes. However, the major PGx guideline and recommendation sources are not completely concordant in terms of their advice. A recent study found inconsistencies in clinical PGx recommendations (48.1%) and in 93.3% of recommendations from CPIC, FDA and clinical practice guidelines (Shugg et al., Reference Shugg, Pasternak, London and Luzum2020). These inconsistencies were spread across a range of domains – recommendation category (29.8%), the patient group (35.4%) and routine screening (15.2%), suggesting a potential barrier to rapid PGx implementation until this is resolved.

CDSS is an effective tool to guide clinicians with limited PGx knowledge (van der Wouden et al., Reference van der Wouden, Cambon-Thomsen, Cecchin, Cheung, Davila-Fajardo, Deneer, Dolzan, Ingelman-Sundberg, Jonsson, Karlsson, Kriek, Mitropoulou, Patrinos, Pirmohamed, Samwald, Schaeffeler, Schwab, Steinberger, Stingl, Sunder-Plassmann, Toffoli, Turner, van Rhenen, Swen and Guchelaar2017). In pre-emptive PGx, patient-specific CDSS alerts prompt and guide clinicians to use genetic information when prescribing drugs with known genetically-determined ADRs (Overby et al., Reference Overby, Erwin, Abul-Husn, Ellis, Scott, Obeng, Kannry, Hripcsak, Bottinger and Gottesman2014; Dunnenberger et al., Reference Dunnenberger, Crews, Hoffman, Caudle, Broeckel, Howard, Hunkler, Klein, Evans and Relling2015).

PGx may be implemented either reactively on a gene-by-gene basis at the time of prescribing a drug, or pre-emptively where a single sample is assessed for several pharmacogenes simultaneously with the results stored for future prescribing encounters. Reactive implementation is expensive and has a slow turnaround time and is unsuitable in situations where rapid drug initiation is required. In contrast, pre-emptive screening of multiple pharmacogenes is likely to be more cost-effective and provides the patient with a lifetime’s worth of test results readily available whenever a drug is prescribed, especially when integrated into electronic health records (EHRs) and drug prescription systems (Relling and Evans, Reference Relling and Evans2015). This further underlines the importance of efficient interoperability between different healthcare systems. A patient may be screened for CYP2C19 prior to being prescribed clopidogrel. These results could inform the prescription of an SSRI or proton pump inhibitor in the future – but only if the results have been stored in an EHR in an accessible format and trigger a CDSS alert at the point of prescription.

Health informatics

PGx implementation in healthcare can be developed in-house if there is availability of capabilities in laboratory and informatics infrastructure and expertise or outsourced to commercial partners. Due to the considerable diversity in commercial PGx products, it is essential to ensure the clinical, IT integration and interoperability requirements along with robust and continuous updating of evidence are rigorously assessed before deciding on the PGx service provider. The significant costs associated with the use of PGx in clinical practice are now in the domain of decision support, IT integration and interoperability, rather than in laboratory genetic testing (Dunnenberger et al., Reference Dunnenberger, Crews, Hoffman, Caudle, Broeckel, Howard, Hunkler, Klein, Evans and Relling2015; Relling and Evans, Reference Relling and Evans2015; van der Wouden et al., Reference van der Wouden, Cambon-Thomsen, Cecchin, Cheung, Davila-Fajardo, Deneer, Dolzan, Ingelman-Sundberg, Jonsson, Karlsson, Kriek, Mitropoulou, Patrinos, Pirmohamed, Samwald, Schaeffeler, Schwab, Steinberger, Stingl, Sunder-Plassmann, Toffoli, Turner, van Rhenen, Swen and Guchelaar2017). Informatics builds within the EHR are easier for a single gene–drug pair as opposed to the multiple pairs and networks that form as drug interactions and clinical factors are also considered. However, the cost-effectiveness data on the pre-emptive panel approach must be assessed, particularly when considering implementation early in life.

Patient and provider acceptability

Patient and healthcare professional acceptability is critical for effective and successful PGx implementation. This requires early and continuous engagement with both clinicians and patients, preferably with champions who are committed (Dressler et al., Reference Dressler, Bell, Ruch, Retamal, Krug and Paulus2018; McDermott et al., Reference McDermott, Wright, Sharma, Newman, Payne and Wilson2022). The main barriers to be tackled in the route to implementation are demonstrating that the system will not overburden the physicians, seamlessly integrate into hospital cornerstone systems, provide sufficient support for the users of the system to navigate the pharmacogenetic evidence base through education and decision support systems, demonstrate utility and cost-effectiveness (Stanek et al., Reference Stanek, Sanders, Taber, Khalid, Patel, Verbrugge, Agatep, Aubert, Epstein and Frueh2012; Just et al., Reference Just, Turner, Dolzan, Cecchin, Swen, Gurwitz and Stingl2019; Bagautdinova et al., Reference Bagautdinova, Lteif, Eddy, Terrell, Fisher and Duarte2022; Scheuner et al., Reference Scheuner, Sales, Hoggatt, Zhang, Whooley and Kelley2023).

Pharmacists are crucial in the PGx service for evaluating appropriate patient eligibility, providing informative post-test counselling, or leading a PGx consult service (Crews et al., Reference Crews, Cross, McCormick, Baker, Molinelli, Mullins, Relling and Hoffman2011; Brown et al., Reference Brown, MacDonald, Yapel, Luczak, Hanson and Stenehjem2021; Bagautdinova et al., Reference Bagautdinova, Lteif, Eddy, Terrell, Fisher and Duarte2022; Krause and Dowd, Reference Krause and Dowd2022).

Health economics

Implementation of PGx in clinical practice requires demonstration of its value and cost-effectiveness to key decision makers and a lack of RCTs that compare genotype-guided prescribing with conventional therapy has not helped. Conducting RCTs for each single drug–gene pair across different ethnicities is not a viable option. Big data analysis of EHRs has the advantage of being able to study diverse populations, limiting concerns about external validity of data and health equality (as is exemplified by the warfarin dosing algorithms that fail to serve patients of African descent). There is limited data on cost-effectiveness multiplexed pre-emptive strategies which are likely to be the preferred solution and the majority of existing cost-effectiveness PGx data are from single gene–drug pair studies (Roden et al., Reference Roden, Van Driest, Mosley, Wells, Robinson, Denny and Peterson2018). Most of the cost-effectiveness studies have been conducted separate from implementation initiatives and they indicate that PGx testing results in a reduction in per-patient treatment cost (Winner et al., Reference Winner, Carhart, Altar, Goldfarb, Allen, Lavezzari, Parsons, Marshak, Garavaglia and Dechairo2015; Deenen et al., Reference Deenen, Meulendijks, Cats, Sechterberger, Severens, Boot, Smits, Rosing, Mandigers, Soesan, Beijnen and Schellens2016), lower cost-per-QALY (Mitropoulou et al., Reference Mitropoulou, Fragoulakis, Bozina, Vozikis, Supe, Bozina, Poljakovic, van Schaik and Patrinos2015) and cost savings in long-term care (Saldivar et al., Reference Saldivar, Taylor, Sugarman, Cullors, Garces, Oades and Centeno2016). A recent systematic appraisal of economic evaluations of PGx testing to prevent ADRs found a number of deficiencies in the quality of data used in cost-effectiveness and cost-utility analyses (Turongkaravee et al., Reference Turongkaravee, Jittikoon, Rochanathimoke, Boyd, Wu and Chaikledkaew2021). Of the 14 economic evaluation studies of CYP2C9 and VKORC1 testing, 10 studies showed that CYP2C9 and VKORC1 testing would be a variably cost-effective and four studies suggested otherwise (Turongkaravee et al., Reference Turongkaravee, Jittikoon, Rochanathimoke, Boyd, Wu and Chaikledkaew2021). In contrast, all nine economic evaluation studies of CYP2C19 testing before prescription of clopidogrel ACS patients undergoing PCI showed that CYP2C19 testing would be a potentially cost-effective treatment strategy for avoiding MACE.

The clopidogrel–CYP2C19 implementation successes need to be contrasted with the difficulties faced in the implementation of warfarin–CYP2C9/CYP4F2/VKORC1 PGx. The key enablers for clopidogrel–CYP2C19 implementation include a discrete patient population (post-PCI), single-gene testing, a high frequency of actionable results, clinically well-established alternative therapies, and a focused group of providers (interventional cardiologists) (Crisamore et al., Reference Crisamore, Nolin, Coons and Empey2019).

Implementation in diverse health care systems

Whilst the above discussion related to healthcare systems in high-income countries, the specific challenges in implementing PGx low- and middle-income countries need to be recognised – lack of clinical efficacy and effectiveness data, under-resourced clinical settings, socio-cultural issues and the identification of population specific pharmacogenomic markers (Tata et al., Reference Tata, Ambele and Pepper2020; Magavern et al., Reference Magavern, Gurdasani, Ng and Lee2022; Sukri et al., Reference Sukri, Salleh, Masimirembwa and Teh2022). The lack of consistent and widely accepted definitions of race, ethnicity and ancestry in genomics and clinical research has resulted in erroneous, inconclusive or absent data on non-European ancestry populations (Popejoy et al., Reference Popejoy, Crooks, Fullerton, Hindorff, Hooker, Koenig, Pino, Ramos, Ritter, Wand, Wright, Yudell, Zou, Plon, Bustamante and Ormond2020). Initiatives such as Human Heredity and Health in Africa (H3Africa) Consortium and the African Pharmacogenomics Research Consortium attempt to increase the representativeness of pharmacogenomic panels (Matimba et al., Reference Matimba, Dhoro and Dandara2016). It is imperative that progress in pharmacogenomic research and implementation occurs at pace in diverse populations so that health disparities are not amplified when PGx becomes more mainstream in clinical practice.

Author contribution

S.P., C.T. and A.F.D. made substantial contributions to the conception drafting, and revision of the manuscript, it critically for important intellectual content. S.P., C.T. and A.F.D. provided final approval of the version to be published.

Abbreviations

ABCB1

ATP-binding cassette sub-family B member 1

ACC

American College of Cardiology

ACS

acute coronary syndrome

ADR

adverse drug reaction

ADRB1

β1-adrenergic receptor

ADRB2

β2-adrenergic receptor

AHA

American Heart Association

BMI

body mass index

CDSS

clinical decision support systems

CES1

liver carboxylesterase 1

CPIC

the Clinical Pharmacogenetics Implementation Consortium

CVD

cardiovascular disease

CYP

cytochrome P450

DOAC

direct-acting oral anticoagulants

DPYD

dihydropyrimidine dehydrogenase

DPWG

Dutch Pharmacogenetics Working Groups

EHR

electronic health records

EMA

European Medicines Agency

EPHA7

ephrin type A receptor 7

ESC

European Society of Cardiology

FDA

U.S. Food and Drug Administration

GATM

Glycine amidinotransferase, mitochondrial

GIST

genotype-informed statin therapy

GRK5

G protein-coupled receptor kinase 5

GWAS

genome-wide association studies

HLA-B

human leukocyte antigen B

IM

intermediate metaboliser

INR

international normalised ratio

KCNH2/hERG

human Ether-à-go-go-related gene

LQTS

long QT syndrome

MACE

major adverse cardiovascular event

NAT2

N-acetyltransferase 2

NM

normal metaboliser

OATP1B1

organic anion transporting polypeptide 1B1

PCI

percutaneous coronary intervention

PM

poor metaboliser

PGx

pharmacogenomics

QALY

quality-adjusted life year

RCT

randomised controlled trial

SAMS

statin-associated muscle symptoms

SLCO1A2

organic anion transporter family member 1A2

SLCO1B1

solute carrier anion transporter family 1B1

SNP

single nucleotide polymorphism

SSRI

selective serotonin reuptake inhibitors

STEMI

ST-segment elevation myocardial infarction

TdP

torsades des pointes

TIA

transient ischaemic attack

TTR

time in the therapeutic INR range

UGT1A

UDP-glucuronosyltransferase 1A1

UM

ultrarapid metaboliser

VKORC1

vitamin K epoxide reductase complex subunit 1

Open peer review

To view the open peer review materials for this article, please visit http://doi.org/10.1017/pcm.2023.17.

Financial support

S.P. and A.F.D. are supported by the British Heart Foundation Centre of Excellence Award (RE/18/6/34217) and the UKRI Strength in Places Fund (SIPF00007/1).

Competing interest

The authors have no competing interest to disclose.

References

Alfirevic, A, Neely, D, Armitage, J, Chinoy, H, Cooper, RG, Laaksonen, R, Carr, DF, Bloch, KM, Fahy, J, Hanson, A, Yue, QY, Wadelius, M, Maitland-van Der Zee, AH, Voora, D, Psaty, BM, Palmer, CN and Pirmohamed, M (2014) Phenotype standardization for statin-induced myotoxicity. Clinical Pharmacology and Therapeutics 96(4), 470476.CrossRefGoogle ScholarPubMed
Amstutz, U, Henricks, LM, Offer, SM, Barbarino, J, Schellens, JHM, Swen, JJ, Klein, TE, McLeod, HL, Caudle, KE, Diasio, RB and Schwab, M (2018) Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for dihydropyrimidine dehydrogenase genotype and fluoropyrimidine dosing: 2017 update. Clinical Pharmacology and Therapeutics 103(2), 210216.CrossRefGoogle ScholarPubMed
Angiolillo, DJ, Rollini, F, Storey, RF, Bhatt, DL, James, S, Schneider, DJ, Sibbing, D, So, DYF, Trenk, D, Alexopoulos, D, Gurbel, PA, Hochholzer, W, De Luca, L, Bonello, L, Aradi, D, Cuisset, T, Tantry, US, Wang, TY, Valgimigli, M, Waksman, R, Mehran, R, Montalescot, G, Franchi, F and Price, MJ (2017) International expert consensus on switching platelet P2Y12 receptor-inhibiting therapies. Circulation 136(20), 19551975.CrossRefGoogle ScholarPubMed
Anstensrud, AK, Molden, E, Haug, HJ, Qazi, R, Muriq, H, Fosshaug, LE, Spigset, O and Oie, E (2020) Impact of genotype-predicted CYP2D6 metabolism on clinical effects and tolerability of metoprolol in patients after myocardial infarction – A prospective observational study. European Journal of Clinical Pharmacology 76(5), 673683.CrossRefGoogle ScholarPubMed
Arking, DE, Pulit, SL, Crotti, L, van der Harst, P, Munroe, PB, Koopmann, TT, Sotoodehnia, N, Rossin, EJ, Morley, M, Wang, X, Johnson, AD, Lundby, A, Gudbjartsson, DF, Noseworthy, PA, Eijgelsheim, M, Bradford, Y, Tarasov, KV, Dorr, M, Muller-Nurasyid, M, Lahtinen, AM, et al. (2014) Genetic association study of QT interval highlights role for calcium signaling pathways in myocardial repolarization. Nature Genetics 46(8), 826836.CrossRefGoogle ScholarPubMed
Arwood, MJ, Deng, J, Drozda, K, Pugach, O, Nutescu, EA, Schmidt, S, Duarte, JD and Cavallari, LH (2017) Anticoagulation endpoints with clinical implementation of warfarin pharmacogenetic dosing in a real-world setting: A proposal for a new pharmacogenetic dosing approach. Clinical Pharmacology and Therapeutics 101(5), 675683.CrossRefGoogle Scholar
Bagautdinova, D, Lteif, C, Eddy, E, Terrell, J, Fisher, CL and Duarte, JD (2022) Patients’ perspectives of factors that influence pharmacogenetic testing uptake: Enhancing patient counseling and results dissemination. Journal of Personalized Medicine 12(12), 2046.CrossRefGoogle ScholarPubMed
Bank PCD, Swen, JJ and Guchelaar, HJ (2019) Estimated nationwide impact of implementing a preemptive pharmacogenetic panel approach to guide drug prescribing in primary care in the Netherlands. BMC Medicine 17(1), 110.Google Scholar
Batty, JA, Hall, AS, White, HL, Wikstrand, J, de Boer, RA, van Veldhuisen, DJ, van der Harst, P, Waagstein, F, Hjalmarson, A, Kjekshus, J, Balmforth, AJ and Group M-HS (2014) An investigation of CYP2D6 genotype and response to metoprolol CR/XL during dose titration in patients with heart failure: A MERIT-HF substudy. Clinical Pharmacology and Therapeutics 95(3), 321330.CrossRefGoogle ScholarPubMed
Baudhuin, LM, Miller, WL, Train, L, Bryant, S, Hartman, KA, Phelps, M, Larock, M and Jaffe, AS (2010) Relation of ADRB1, CYP2D6, and UGT1A1 polymorphisms with dose of, and response to, carvedilol or metoprolol therapy in patients with chronic heart failure. American Journal of Cardiology 106(3), 402408.CrossRefGoogle ScholarPubMed
Bauer, T, Bouman, HJ, van Werkum, JW, Ford, NF, ten Berg, JM and Taubert, D (2011) Impact of CYP2C19 variant genotypes on clinical efficacy of antiplatelet treatment with clopidogrel: Systematic review and meta-analysis. BMJ 343, d4588.CrossRefGoogle ScholarPubMed
Becker, RC, Bassand, JP, Budaj, A, Wojdyla, DM, James, SK, Cornel, JH, French, J, Held, C, Horrow, J, Husted, S, Lopez-Sendon, J, Lassila, R, Mahaffey, KW, Storey, RF, Harrington, RA and Wallentin, L (2011) Bleeding complications with the P2Y12 receptor antagonists clopidogrel and ticagrelor in the PLATelet inhibition and patient outcomes (PLATO) trial. European Heart Journal 32(23), 29332944.CrossRefGoogle ScholarPubMed
Bell, GC, Caudle, KE, Whirl-Carrillo, M, Gordon, RJ, Hikino, K, Prows, CA, Gaedigk, A, Agundez, J, Sadhasivam, S, Klein, TE and Schwab, M (2017) Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2D6 genotype and use of ondansetron and tropisetron. Clinical Pharmacology and Therapeutics 102(2), 213218.CrossRefGoogle ScholarPubMed
Bijl, MJ, Visser, LE, van Schaik, RH, Kors, JA, Witteman, JC, Hofman, A, Vulto, AG, van Gelder, T and Stricker, BH (2009) Genetic variation in the CYP2D6 gene is associated with a lower heart rate and blood pressure in beta-blocker users. Clinical Pharmacology and Therapeutics 85(1), 4550.CrossRefGoogle ScholarPubMed
Blake, CM, Kharasch, ED, Schwab, M and Nagele, P (2013) A meta-analysis of CYP2D6 metabolizer phenotype and metoprolol pharmacokinetics. Clinical Pharmacology and Therapeutics 94(3), 394399.CrossRefGoogle ScholarPubMed
Bourgeois, S, Jorgensen, A, Zhang, EJ, Hanson, A, Gillman, MS, Bumpstead, S, Toh, CH, Williamson, P, Daly, AK, Kamali, F, Deloukas, P and Pirmohamed, M (2016) A multi-factorial analysis of response to warfarin in a UK prospective cohort. Genome Medicine 8(1), 2.CrossRefGoogle Scholar
Brouwer, J, Nijenhuis, M, Soree, B, Guchelaar, HJ, Swen, JJ, van Schaik, RHN, Weide, JV, Rongen, G, Buunk, AM, de Boer-Veger, NJ, Houwink, EJF, van Westrhenen, R, Wilffert, B, VHM, Deneer and Mulder, H (2022) Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction between CYP2C19 and CYP2D6 and SSRIs. European Journal of Human Genetics 30, 11141120.CrossRefGoogle Scholar
Brown, JT, MacDonald, D, Yapel, A, Luczak, T, Hanson, A and Stenehjem, DD (2021) Integrating pharmacogenetic testing via medication therapy management in an outpatient family medicine clinic. Pharmacogenomics 22(4), 203212.CrossRefGoogle Scholar
Cancino, RS, Hylek, EM, Reisman, JI and Rose, AJ (2014) Comparing patient-level and site-level anticoagulation control as predictors of adverse events. Thrombosis Research 133(4), 652656.CrossRefGoogle ScholarPubMed
Carr, DF, Francis, B, Jorgensen, AL, Zhang, E, Chinoy, H, Heckbert, SR, Bis, JC, Brody, JA, Floyd, JS, Psaty, BM, Molokhia, M, Lapeyre-Mestre, M, Conforti, A, Alfirevic, A, van Staa, T and Pirmohamed, M (2019) Genomewide association study of statin-induced myopathy in patients recruited using the UK clinical practice research datalink. Clinical Pharmacology and Therapeutics 106(6), 13531361.CrossRefGoogle ScholarPubMed
Carreras, ET, Hochholzer, W, Frelinger, AL, Nordio, F, O’Donoghue, ML, Wiviott, SD, Angiolillo, DJ, Michelson, AD, Sabatine, MS and Mega, JL (2016) Diabetes mellitus, CYP2C19 genotype, and response to escalating doses of clopidogrel. Insights from the ELEVATE-TIMI 56 trial. Thrombosis and Haemostasis 116(1), 6977.Google ScholarPubMed
Catapano, AL, Graham, I, De Backer, G, Wiklund, O, Chapman, MJ, Drexel, H, Hoes, AW, Jennings, CS, Landmesser, U, Pedersen, TR, Reiner, Z, Riccardi, G, Taskinen, MR, Tokgozoglu, L, WMM, Verschuren, Vlachopoulos, C, Wood, DA, Zamorano, JL, Cooney, MT and Group ESCSD (2016) 2016 ESC/EAS guidelines for the management of dyslipidaemias. European Heart Journal 37(39), 29993058.CrossRefGoogle ScholarPubMed
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 and Network, I (2018) Multisite investigation of outcomes with implementation of CYP2C19 genotype-guided antiplatelet therapy after percutaneous coronary intervention. JACC. Cardiovascular Interventions 11(2), 181191.CrossRefGoogle ScholarPubMed
Chanfreau-Coffinier, C, Hull, LE, Lynch, JA, DuVall, SL, Damrauer, SM, Cunningham, FE, Voight, BF, Matheny, ME, Oslin, DW, Icardi, MS and Tuteja, S (2019) Projected prevalence of actionable pharmacogenetic variants and level A drugs prescribed among US veterans health administration pharmacy users. JAMA Network Open 2(6), e195345.CrossRefGoogle Scholar
Choi, HY, Bae, KS, Cho, SH, Ghim, JL, Choe, S, Jung, JA, Jin, SJ, Kim, HS and Lim, HS (2015) Impact of CYP2D6, CYP3A5, CYP2C19, CYP2A6, SLCO1B1, ABCB1, and ABCG2 gene polymorphisms on the pharmacokinetics of simvastatin and simvastatin acid. Pharmacogenetics and Genomics 25(12), 595608.CrossRefGoogle ScholarPubMed
Claassens, DM and Sibbing, D (2020) De-escalation of antiplatelet treatment in patients with myocardial infarction who underwent percutaneous coronary intervention: A review of the current literature. Journal of Clinical Medicine 9(9), 2983.CrossRefGoogle ScholarPubMed
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, WJM, Dewilde, PWA, Janssen, Kelder, JC, Postma, MJ, de Boer, A, Boersma, C, VHM, Deneer and Ten Berg, JM (2019) A genotype-guided strategy for oral P2Y12 inhibitors in primary PCI. New England Journal of Medicine 381(17), 16211631.CrossRefGoogle ScholarPubMed
Collet, JP, Thiele, H, Barbato, E, Barthelemy, O, Bauersachs, J, Bhatt, DL, Dendale, P, Dorobantu, M, Edvardsen, T, Folliguet, T, Gale, CP, Gilard, M, Jobs, A, Juni, P, Lambrinou, E, Lewis, BS, Mehilli, J, Meliga, E, Merkely, B, Mueller, C, Roffi, M, Rutten, FH, Sibbing, D, GCM, Siontis and Group ESCSD (2021) 2020 ESC guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. European Heart Journal 42(14), 12891367.CrossRefGoogle ScholarPubMed
Collins, FS and Varmus, H (2015) A new initiative on precision medicine. New England Journal of Medicine 372(9), 793795.CrossRefGoogle ScholarPubMed
Cooper, GM, Johnson, JA, Langaee, TY, Feng, H, Stanaway, IB, Schwarz, UI, Ritchie, MD, Stein, CM, Roden, DM, Smith, JD, Veenstra, DL, Rettie, AE and Rieder, MJ (2008) A genome-wide scan for common genetic variants with a large influence on warfarin maintenance dose. Blood 112(4), 10221027.CrossRefGoogle ScholarPubMed
CPIC (2022) Genes-drugs. Available at https://cpicpgx.org/genes-drugs/ (accessed 20 March 2023).Google Scholar
Crews, KR, Cross, SJ, McCormick, JN, Baker, DK, Molinelli, AR, Mullins, R, Relling, MV and Hoffman, JM (2011) Development and implementation of a pharmacist-managed clinical pharmacogenetics service. American Journal of Health-System Pharmacy 68(2), 143150.CrossRefGoogle ScholarPubMed
Crews, KR, Gaedigk, A, Dunnenberger, HM, Leeder, JS, Klein, TE, Caudle, KE, Haidar, CE, Shen, DD, Callaghan, JT, Sadhasivam, S, Prows, CA, Kharasch, ED, Skaar, TC and Implementation, Clinical Pharmacogenetics (2014) Clinical Pharmacogenetics Implementation Consortium guidelines for cytochrome P450 2D6 genotype and codeine therapy: 2014 update. Clinical Pharmacology and Therapeutics 95(4), 376382.CrossRefGoogle ScholarPubMed
Crisamore, KR, Nolin, TD, Coons, JC and Empey, PE (2019) Engaging and empowering stakeholders to advance pharmacogenomics. Clinical Pharmacology and Therapeutics 106(2), 305308.CrossRefGoogle ScholarPubMed
Danese, E, Raimondi, S, Montagnana, M, Tagetti, A, Langaee, T, Borgiani, P, Ciccacci, C, Carcas, AJ, Borobia, AM, Tong, HY, Davila-Fajardo, C, Rodrigues Botton, M, Bourgeois, S, Deloukas, P, Caldwell, MD, Burmester, JK, Berg, RL, Cavallari, LH, Drozda, K, Huang, M, Zhao, LZ, Cen, HJ, Gonzalez-Conejero, R, Roldan, V, Nakamura, Y, Mushiroda, T, Gong, IY, Kim, RB, Hirai, K, Itoh, K, Isaza, C, Beltran, L, Jimenez-Varo, E, Canadas-Garre, M, Giontella, A, Kringen, MK, Haug, KBF, Gwak, HS, Lee, KE, Minuz, P, Lee, MTM, Lubitz, SA, Scott, S, Mazzaccara, C, Sacchetti, L, Genc, E, Ozer, M, Pathare, A, Krishnamoorthy, R, Paldi, A, Siguret, V, Loriot, MA, Kutala, VK, Suarez-Kurtz, G, Perini, J, Denny, JC, Ramirez, AH, Mittal, B, Rathore, SS, Sagreiya, H, Altman, R, Shahin, MHA, Khalifa, SI, Limdi, NA, Rivers, C, Shendre, A, Dillon, C, Suriapranata, IM, Zhou, HH, Tan, SL, Tatarunas, V, Lesauskaite, V, Zhang, Y, Maitland-van der Zee, AH, Verhoef, TI, de Boer, A, Taljaard, M, Zambon, CF, Pengo, V, Zhang, JE, Pirmohamed, M, Johnson, JA and Fava, C (2019) Effect of CYP4F2, VKORC1, and CYP2C9 in influencing coumarin dose: A single-patient data meta-analysis in more than 15,000 individuals. Clinical Pharmacology and Therapeutics 105(6), 14771491.CrossRefGoogle Scholar
Danik, JS, Chasman, DI, MacFadyen, JG, Nyberg, F, Barratt, BJ and Ridker, PM (2013) Lack of association between SLCO1B1 polymorphisms and clinical myalgia following rosuvastatin therapy. American Heart Journal 165(6), 10081014.CrossRefGoogle ScholarPubMed
de Keyser, CE, Peters, BJ, Becker, ML, Visser, LE, Uitterlinden, AG, Klungel, OH, Verstuyft, C, Hofman, A, Maitland-van der Zee, AH and Stricker, BH (2014) The SLCO1B1 c.521T>C polymorphism is associated with dose decrease or switching during statin therapy in the Rotterdam study. Pharmacogenetics and Genomics 24(1), 4351.CrossRefGoogle ScholarPubMed
De, T, Alarcon, C, Hernandez, W, Liko, I, Cavallari, LH, Duarte, JD and Perera, MA (2018) Association of genetic variants with warfarin-associated bleeding among patients of African descent. JAMA 320(16), 16701677.CrossRefGoogle ScholarPubMed
Deenen, MJ, Meulendijks, D, Cats, A, Sechterberger, MK, Severens, JL, Boot, H, Smits, PH, Rosing, H, Mandigers, CM, Soesan, M, Beijnen, JH and Schellens, JH (2016) Upfront genotyping of DPYD*2A to individualize fluoropyrimidine therapy: A safety and cost analysis. Journal of Clinical Oncology 34(3), 227234.CrossRefGoogle ScholarPubMed
Diaz-Villamarin, X, Davila-Fajardo, CL, Martinez-Gonzalez, LJ, Carmona-Saez, P, Sanchez-Ramos, J, Alvarez Cubero, MJ, Salmeron-Febres, LM, Cabeza Barrera, J and Fernandez-Quesada, F (2016) Genetic polymorphisms influence on the response to clopidogrel in peripheral artery disease patients following percutaneous transluminal angioplasty. Pharmacogenomics 17(12), 13271338.CrossRefGoogle ScholarPubMed
Doki, K, Sekiguchi, Y, Kuga, K, Aonuma, K and Homma, M (2015) Serum flecainide S/R ratio reflects the CYP2D6 genotype and changes in CYP2D6 activity. Drug Metabolism and Pharmacokinetics 30(4), 257262.CrossRefGoogle ScholarPubMed
Dressler, LG, Bell, GC, Ruch, KD, Retamal, JD, Krug, PB and Paulus, RA (2018) Implementing a personalized medicine program in a community health system. Pharmacogenomics 19(17), 13451356.CrossRefGoogle Scholar
Drozda, K, Wong, S, Patel, SR, Bress, AP, Nutescu, EA, Kittles, RA and Cavallari, LH (2015) Poor warfarin dose prediction with pharmacogenetic algorithms that exclude genotypes important for African Americans. Pharmacogenetics and Genomics 25(2), 7381.CrossRefGoogle ScholarPubMed
Dunnenberger, HM, Crews, KR, Hoffman, JM, Caudle, KE, Broeckel, U, Howard, SC, Hunkler, RJ, Klein, TE, Evans, WE and Relling, MV (2015) Preemptive clinical pharmacogenetics implementation: Current programs in five US medical centers. Annual Review of Pharmacology and Toxicology 55, 89106.CrossRefGoogle ScholarPubMed
Ehmann, F, Caneva, L, Prasad, K, Paulmichl, M, Maliepaard, M, Llerena, A, Ingelman-Sundberg, M and Papaluca-Amati, M (2015) Pharmacogenomic information in drug labels: European medicines agency perspective. Pharmacogenomics Journal 15(3), 201210.CrossRefGoogle ScholarPubMed
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 and Network, I (2018) Multisite investigation of strategies for the implementation of CYP2C19 genotype-guided antiplatelet therapy. Clinical Pharmacology and Therapeutics 104(4), 664674.CrossRefGoogle ScholarPubMed
FDA (2023a) Table of Pharmacogenetic Associations. U.S. Food and Drug Administration. Available at https://www.fda.gov/medical-devices/precision-medicine/table-pharmacogenetic-associations (accessed 20 March 2023).Google Scholar
FDA (2023b) Table of Pharmacogenomic Biomarkers in Drug Labeling. U.S. Food and Drug Administration. Available at https://www.fda.gov/drugs/science-and-research-drugs/table-pharmacogenomic-biomarkers-drug-labeling (accessed 20 March 2023).Google Scholar
Fiegenbaum, M, da Silveira, FR, Van der Sand, CR, Van der Sand, LC, Ferreira, ME, Pires, RC and Hutz, MH (2005) The role of common variants of ABCB1, CYP3A4, and CYP3A5 genes in lipid-lowering efficacy and safety of simvastatin treatment. Clinical Pharmacology and Therapeutics 78(5), 551558.CrossRefGoogle ScholarPubMed
Gage, BF, Bass, AR, Lin, H, Woller, SC, Stevens, SM, Al-Hammadi, N, Li, J, Rodriguez, 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, Davila-Roman, V and 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(12), 11151124.CrossRefGoogle ScholarPubMed
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 and McLeod, HL (2008) Use of pharmacogenetic and clinical factors to predict the therapeutic dose of warfarin. Clinical Pharmacology and Therapeutics 84(3), 326331.CrossRefGoogle ScholarPubMed
Goldberger, JJ and Buxton, AE (2013) Personalized medicine vs guideline-based medicine. JAMA 309(24), 25592560.CrossRefGoogle ScholarPubMed
Gonzalez-Fierro, A, Vasquez-Bahena, D, Taja-Chayeb, L, Vidal, S, Trejo-Becerril, C, Perez-Cardenas, E, de la Cruz-Hernandez, E, Chavez-Blanco, A, Gutierrez, O, Rodriguez, D, Fernandez, Z and Duenas-Gonzalez, A (2011) Pharmacokinetics of hydralazine, an antihypertensive and DNA-demethylating agent, using controlled-release formulations designed for use in dosing schedules based on the acetylator phenotype. International Journal of Clinical Pharmacology and Therapeutics 49(8), 519524.CrossRefGoogle ScholarPubMed
Greden, JF, Parikh, SV, Rothschild, AJ, Thase, ME, Dunlop, BW, DeBattista, C, Conway, CR, Forester, BP, Mondimore, FM, Shelton, RC, Macaluso, M, Li, J, Brown, K, Gilbert, A, Burns, L, Jablonski, MR and Dechairo, B (2019) Impact of pharmacogenomics on clinical outcomes in major depressive disorder in the GUIDED trial: A large, patient- and rater-blinded, randomized, controlled study. Journal of Psychiatric Research 111, 5967.CrossRefGoogle Scholar
Guo, B, Tan, Q, Guo, D, Shi, Z, Zhang, C and Guo, W (2014) Patients carrying CYP2C19 loss of function alleles have a reduced response to clopidogrel therapy and a greater risk of in-stent restenosis after endovascular treatment of lower extremity peripheral arterial disease. Journal of Vascular Surgery 60(4), 9931001.CrossRefGoogle Scholar
Hamadeh, IS, Langaee, TY, Dwivedi, R, Garcia, S, Burkley, BM, Skaar, TC, Chapman, AB, Gums, JG, Turner, ST, Gong, Y, Cooper-DeHoff, RM and Johnson, JA (2014) Impact of CYP2D6 polymorphisms on clinical efficacy and tolerability of metoprolol tartrate. Clinical Pharmacology and Therapeutics 96(2), 175181.CrossRefGoogle ScholarPubMed
Han, LW, Ryu, RJ, Cusumano, M, Easterling, TR, Phillips, BR, Risler, LJ, Shen, DD and Hebert, MF (2019) Effect of N-acetyltransferase 2 genotype on the pharmacokinetics of hydralazine during pregnancy. Journal of Clinical Pharmacology 59(12), 16781689.CrossRefGoogle ScholarPubMed
Heise, CW, Gallo, T, Curry, SC and Woosley, RL (2020) Identification of populations likely to benefit from pharmacogenomic testing. Pharmacogenetics and Genomics 30(5), 9195.CrossRefGoogle ScholarPubMed
Hicks, JK, Bishop, JR, Sangkuhl, K, Muller, DJ, Ji, Y, Leckband, SG, Leeder, JS, Graham, RL, Chiulli, DL, LLerena, A, Skaar, TC, Scott, SA, Stingl, JC, Klein, TE, Caudle, KE, Gaedigk, A and Clinical Pharmacogenetics Implementation (2015) Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2D6 and CYP2C19 genotypes and dosing of selective serotonin reuptake inhibitors. Clinical Pharmacology and Therapeutics 98(2), 127134.CrossRefGoogle ScholarPubMed
Hicks, JK, El Rouby, N, Ong, HH, Schildcrout, JS, Ramsey, LB, Shi, Y, Anne Tang, L, Aquilante, CL, Beitelshees, AL, Blake, KV, Cimino, JJ, Davis, BH, Empey, PE, Kao, DP, Lemkin, DL, Limdi, NA, PL, G, Rosenman, MB, Skaar, TC, Teal, E, Tuteja, S, Wiley, LK, Williams, H, Winterstein, AG, Van Driest, SL, Cavallari, LH, Peterson, JF and Group IPW (2021) Opportunity for genotype-guided prescribing among adult patients in 11 US health systems. Clinical Pharmacology and Therapeutics 110(1), 179188.CrossRefGoogle ScholarPubMed
Ho, KH, van Hove, M and Leng, G (2020) Trends in anticoagulant prescribing: A review of local policies in English primary care. BMC Health Services Research 20(1), 279.CrossRefGoogle ScholarPubMed
Hoenig, MR, Walker, PJ, Gurnsey, C, Beadle, K and Johnson, L (2011) The C3435T polymorphism in ABCB1 influences atorvastatin efficacy and muscle symptoms in a high-risk vascular cohort. Journal of Clinical Lipidology 5(2), 9196.CrossRefGoogle Scholar
Holmes, DR, Dehmer, GJ, Kaul, S, Leifer, D, O’Gara, PT and Stein, CM (2010) ACCF/AHA clopidogrel clinical alert: Approaches to the FDA “boxed warning”: A report of the American College of Cardiology Foundation task force on clinical expert consensus documents and the American Heart Association endorsed by the Society for Cardiovascular Angiography and Interventions and the Society of Thoracic Surgeons. Journal of the American College of Cardiology 56(4), 321341.CrossRefGoogle Scholar
Holmes, MV, Perel, P, Shah, T, Hingorani, AD and Casas, JP (2011) CYP2C19 genotype, clopidogrel metabolism, platelet function, and cardiovascular events: A systematic review and meta-analysis. JAMA 306(24), 27042714.CrossRefGoogle ScholarPubMed
Huang, J, Li, C, Song, Y, Fan, X, You, L, Tan, L, Xiao, L, Li, Q, Ruan, G, Hu, S, Cui, W, Li, Z, Ni, L, Chen, C, Woo, AY, Xiao, RP and Wang, DW (2018) ADRB2 polymorphism Arg16Gly modifies the natural outcome of heart failure and dictates therapeutic response to beta-blockers in patients with heart failure. Cell Discovery 4, 57.CrossRefGoogle ScholarPubMed
Hulot, JS, Collet, JP, Silvain, J, Pena, A, Bellemain-Appaix, A, Barthelemy, O, Cayla, G, Beygui, F and Montalescot, G (2010) 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. Journal of the American College of Cardiology 56(2), 134143.CrossRefGoogle ScholarPubMed
Hylek, EM, Evans-Molina, C, Shea, C, Henault, LE and Regan, S (2007) Major hemorrhage and tolerability of warfarin in the first year of therapy among elderly patients with atrial fibrillation. Circulation 115(21), 26892696.CrossRefGoogle ScholarPubMed
Ibanez, B, James, S, Agewall, S, Antunes, MJ, Bucciarelli-Ducci, C, Bueno, H, ALP, Caforio, Crea, F, Goudevenos, JA, Halvorsen, S, Hindricks, G, Kastrati, A, Lenzen, MJ, Prescott, E, Roffi, M, Valgimigli, M, Varenhorst, C, Vranckx, P, Widimsky, P and Group ESCSD (2018) 2017 ESC guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The task force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). European Heart Journal 39(2), 119177.CrossRefGoogle ScholarPubMed
Ingelman-Sundberg, M, Sim, SC, Gomez, A and Rodriguez-Antona, C (2007) Influence of cytochrome P450 polymorphisms on drug therapies: Pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacology & Therapeutics 116(3), 496526.CrossRefGoogle ScholarPubMed
International Warfarin Pharmacogenetics, Klein, TE, Altman, RB, Eriksson, N, Gage, BF, Kimmel, SE, Lee, MT, Limdi, NA, Page, D, Roden, DM, Wagner, MJ, Caldwell, MD and Johnson, JA (2009) Estimation of the warfarin dose with clinical and pharmacogenetic data. New England Journal of Medicine 360(8), 753764.Google ScholarPubMed
Johnson, JA, Caudle, KE, Gong, L, Whirl-Carrillo, M, Stein, CM, Scott, SA, Lee, MT, Gage, BF, Kimmel, SE, Perera, MA, Anderson, JL, Pirmohamed, M, Klein, TE, Limdi, NA, Cavallari, LH and Wadelius, M (2017) Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for pharmacogenetics-guided warfarin dosing: 2017 update. Clinical Pharmacology and Therapeutics 102(3), 397404.CrossRefGoogle ScholarPubMed
Jones, M, McEwan, P, Morgan, CL, Peters, JR, Goodfellow, J and Currie, CJ (2005) Evaluation of the pattern of treatment, level of anticoagulation control, and outcome of treatment with warfarin in patients with non-valvar atrial fibrillation: A record linkage study in a large British population. Heart 91(4), 472477.CrossRefGoogle Scholar
Just, KS, Turner, RM, Dolzan, V, Cecchin, E, Swen, JJ, Gurwitz, D and Stingl, JC (2019) Educating the next generation of pharmacogenomics experts: Global educational needs and concepts. Clinical Pharmacology and Therapeutics 106(2), 313316.CrossRefGoogle ScholarPubMed
Kaminsky, LS and Zhang, ZY (1997) Human P450 metabolism of warfarin. Pharmacology & Therapeutics 73(1), 6774.CrossRefGoogle ScholarPubMed
Kazui, M, Nishiya, Y, Ishizuka, T, Hagihara, K, Farid, NA, Okazaki, O, Ikeda, T and Kurihara, A (2010) Identification of the human cytochrome P450 enzymes involved in the two oxidative steps in the bioactivation of clopidogrel to its pharmacologically active metabolite. Drug Metabolism and Disposition 38(1), 9299.CrossRefGoogle ScholarPubMed
Kheiri, B, Simpson, TF, Osman, M, Kumar, K, Przybylowicz, R, Merrill, M, Golwala, H, Rahmouni, H, Cigarroa, JE and Zahr, F (2020) Genotype-guided strategy for P2Y12 inhibitors in coronary artery disease: A meta-analysis of randomized clinical trials. JACC. Cardiovascular Interventions 13(5), 659661.CrossRefGoogle ScholarPubMed
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 III, Fang, MC, Shah, V, Horenstein, RB, Limdi, NA, Muldowney, JA III, Gujral, J, Delafontaine, P, Desnick, RJ, Ortel, TL, Billett, HH, Pendleton, RC, Geller, NL, Halperin, JL, Goldhaber, SZ, Caldwell, MD, Califf, RM, Ellenberg, JH and Investigators, C (2013) A pharmacogenetic versus a clinical algorithm for warfarin dosing. New England Journal of Medicine 369(24), 22832293.CrossRefGoogle ScholarPubMed
Krause, DS and Dowd, D (2022) Use of a consultation service following pharmacogenetic testing in psychiatry. Pharmacogenomics 23(5), 327333.CrossRefGoogle ScholarPubMed
Lamoureux, F, Duflot, T and French Network of Pharmacogenetics (2017) Pharmacogenetics in cardiovascular diseases: State of the art and implementation-recommendations of the French National Network of pharmacogenetics (RNPGx). Thérapie 72(2), 257267.Google ScholarPubMed
Landefeld, CS and Beyth, RJ (1993) Anticoagulant-related bleeding: Clinical epidemiology, prediction, and prevention. American Journal of Medicine 95(3), 315328.CrossRefGoogle ScholarPubMed
Lau, WCY, Li, X, Wong, ICK, Man, KKC, Lip, GYH, Leung, WK, Siu, CW and Chan, EW (2017) Bleeding-related hospital admissions and 30-day readmissions in patients with non-valvular atrial fibrillation treated with dabigatran versus warfarin. Journal of Thrombosis and Haemostasis 15(10), 19231933.CrossRefGoogle ScholarPubMed
Lee, CR, Luzum, JA, Sangkuhl, K, Gammal, RS, Sabatine, MS, Stein, CM, Kisor, DF, Limdi, NA, Lee, YM, Scott, SA, Hulot, JS, Roden, DM, Gaedigk, A, Caudle, KE, Klein, TE, Johnson, JA and Shuldiner, AR (2022) Clinical Pharmacogenetics Implementation Consortium guideline for CYP2C19 genotype and clopidogrel therapy: 2022 update. Clinical Pharmacology and Therapeutics 112(5), 959967.CrossRefGoogle ScholarPubMed
Lim, KS, Jang, IJ, Kim, BH, Kim, J, Jeon, JY, Tae, YM, Yi, S, Eum, S, Cho, JY, Shin, SG and Yu, KS (2010) Changes in the QTc interval after administration of flecainide acetate, with and without coadministered paroxetine, in relation to cytochrome P450 2D6 genotype: Data from an open-label, two-period, single-sequence crossover study in healthy Korean male subjects. Clinical Therapeutics 32(4), 659666.CrossRefGoogle ScholarPubMed
Lima, JJ, Thomas, CD, Barbarino, J, Desta, Z, Van Driest, SL, El Rouby, N, Johnson, JA, Cavallari, LH, Shakhnovich, V, Thacker, DL, Scott, SA, Schwab, M, Uppugunduri, CRS, Formea, CM, Franciosi, JP, Sangkuhl, K, Gaedigk, A, Klein, TE, Gammal, RS and Furuta, T (2021) Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2C19 and proton pump inhibitor dosing. Clinical Pharmacology and Therapeutics 109(6), 14171423.CrossRefGoogle ScholarPubMed
Limdi, NA, Wadelius, M, Cavallari, L, Eriksson, N, Crawford, DC, Lee, MT, Chen, CH, Motsinger-Reif, A, Sagreiya, H, Liu, N, Wu, AH, Gage, BF, Jorgensen, A, Pirmohamed, M, Shin, JG, Suarez-Kurtz, G, Kimmel, SE, Johnson, JA, Klein, TE, Wagner, MJ and International Warfarin Pharmacogenetics (2010) Warfarin pharmacogenetics: A single VKORC1 polymorphism is predictive of dose across 3 racial groups. Blood 115(18), 38273834.CrossRefGoogle ScholarPubMed
Linskey, DW, English, JD, Perry, DA, Ochs-Balcom, HM, Ma, C, Isackson, PJ, Vladutiu, GD and Luzum, JA (2020) Association of SLCO1B1 c.521T>C (rs4149056) with discontinuation of atorvastatin due to statin-associated muscle symptoms. Pharmacogenetics and Genomics 30(9), 208211.CrossRefGoogle ScholarPubMed
Magavern, EF, Gurdasani, D, Ng, FL and Lee, SS (2022) Health equality, race and pharmacogenomics. British Journal of Clinical Pharmacology 88(1), 2733.CrossRefGoogle ScholarPubMed
Magvanjav, O, McDonough, CW, Gong, Y, McClure, LA, Talbert, RL, Horenstein, RB, Shuldiner, AR, Benavente, OR, Mitchell, BD, Johnson, JA and SiGN, N (2017) Pharmacogenetic associations of beta1-adrenergic receptor polymorphisms with cardiovascular outcomes in the SPS3 trial (secondary prevention of small subcortical strokes). Stroke 48(5), 13371343.CrossRefGoogle ScholarPubMed
Mallal, S, Phillips, E, Carosi, G, Molina, JM, Workman, C, Tomazic, J, Jagel-Guedes, E, Rugina, S, Kozyrev, O, Cid, JF, Hay, P, Nolan, D, Hughes, S, Hughes, A, Ryan, S, Fitch, N, Thorborn, D, Benbow, A and Team P-S (2008) HLA-B*5701 screening for hypersensitivity to abacavir. New England Journal of Medicine 358(6), 568579.CrossRefGoogle ScholarPubMed
Mangravite, LM, Engelhardt, BE, Medina, MW, Smith, JD, Brown, CD, Chasman, DI, Mecham, BH, Howie, B, Shim, H, Naidoo, D, Feng, Q, Rieder, MJ, Chen, YD, Rotter, JI, Ridker, PM, Hopewell, JC, Parish, S, Armitage, J, Collins, R, Wilke, RA, Nickerson, DA, Stephens, M and Krauss, RM (2013) A statin-dependent QTL for GATM expression is associated with statin-induced myopathy. Nature 502(7471), 377380.CrossRefGoogle ScholarPubMed
Manolio, TA, Chisholm, RL, Ozenberger, B, Roden, DM, Williams, MS, Wilson, R, Bick, D, Bottinger, EP, Brilliant, MH, Eng, C, Frazer, KA, Korf, B, Ledbetter, DH, Lupski, JR, Marsh, C, Mrazek, D, Murray, MF, O’Donnell, PH, Rader, DJ, Relling, MV, Shuldiner, AR, Valle, D, Weinshilboum, R, Green, ED and Ginsburg, GS (2013) Implementing genomic medicine in the clinic: The future is here. Genetics in Medicine 15(4), 258267.CrossRefGoogle Scholar
Matimba, A, Dhoro, M and Dandara, C (2016) Is there a role of pharmacogenomics in Africa. Global Health, Epidemiology and Genomics 1, e9.CrossRefGoogle Scholar
Mazari, L, Ouarzane, M and Zouali, M (2007) Subversion of B lymphocyte tolerance by hydralazine, a potential mechanism for drug-induced lupus. Proceedings of the National Academy of Sciences of the United States of America 104(15), 63176322.CrossRefGoogle ScholarPubMed
McDermott, JH, Wright, S, Sharma, V, Newman, WG, Payne, K and Wilson, P (2022) Characterizing pharmacogenetic programs using the consolidated framework for implementation research: A structured scoping review. Frontiers in Medicine (Lausanne) 9, 945352.CrossRefGoogle ScholarPubMed
Mega, JL, Close, SL, Wiviott, SD, Shen, L, Hockett, RD, Brandt, JT, Walker, JR, Antman, EM, Macias, W, Braunwald, E and Sabatine, MS (2009a) Cytochrome p-450 polymorphisms and response to clopidogrel. New England Journal of Medicine 360(4), 354362.CrossRefGoogle ScholarPubMed
Mega, JL, Close, SL, Wiviott, SD, Shen, L, Hockett, RD, Brandt, JT, Walker, JR, Antman, EM, Macias, WL, Braunwald, E and Sabatine, MS (2009b) Cytochrome P450 genetic polymorphisms and the response to prasugrel: Relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Circulation 119(19), 25532560.CrossRefGoogle ScholarPubMed
Mega, JL, Hochholzer, W, Frelinger, AL III, Kluk, MJ, Angiolillo, DJ, Kereiakes, DJ, Isserman, S, Rogers, WJ, Ruff, CT, Contant, C, Pencina, MJ, Scirica, BM, Longtine, JA, Michelson, AD and Sabatine, MS (2011) Dosing clopidogrel based on CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovascular disease. JAMA 306(20), 22212228.CrossRefGoogle ScholarPubMed
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 and 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(16), 18211830.CrossRefGoogle ScholarPubMed
Mega, JL, Walker, JR, Ruff, CT, Vandell, AG, Nordio, F, Deenadayalu, N, Murphy, SA, Lee, J, Mercuri, MF, Giugliano, RP, Antman, EM, Braunwald, E and Sabatine, MS (2015) Genetics and the clinical response to warfarin and edoxaban: Findings from the randomised, double-blind ENGAGE AF-TIMI 48 trial. Lancet 385(9984), 22802287.CrossRefGoogle ScholarPubMed
Mehta, D, Uber, R, Ingle, T, Li, C, Liu, Z, Thakkar, S, Ning, B, Wu, L, Yang, J, Harris, S, Zhou, G, Xu, J, Tong, W, Lesko, L and Fang, H (2020) Study of pharmacogenomic information in FDA-approved drug labeling to facilitate application of precision medicine. Drug Discovery Today 25(5), 813820.CrossRefGoogle ScholarPubMed
Mitropoulou, C, Fragoulakis, V, Bozina, N, Vozikis, A, Supe, S, Bozina, T, Poljakovic, Z, van Schaik, RH and Patrinos, GP (2015) Economic evaluation of pharmacogenomic-guided warfarin treatment for elderly Croatian atrial fibrillation patients with ischemic stroke. Pharmacogenomics 16(2), 137148.CrossRefGoogle ScholarPubMed
Overby, CL, Erwin, AL, Abul-Husn, NS, Ellis, SB, Scott, SA, Obeng, AO, Kannry, JL, Hripcsak, G, Bottinger, EP and Gottesman, O (2014) Physician attitudes toward adopting genome-guided prescribing through clinical decision support. Journal of Personalized Medicine 4(1), 3549.CrossRefGoogle ScholarPubMed
Pacanowski, MA, Gong, Y, Cooper-Dehoff, RM, Schork, NJ, Shriver, MD, Langaee, TY, Pepine, CJ, Johnson, JA and Investigators, I (2008) Beta-adrenergic receptor gene polymorphisms and beta-blocker treatment outcomes in hypertension. Clinical Pharmacology and Therapeutics 84(6), 715721.CrossRefGoogle ScholarPubMed
Pan, Y, Chen, W, Xu, Y, Yi, X, Han, Y, Yang, Q, Li, X, Huang, L, Johnston, SC, Zhao, X, Liu, L, Zhang, Q, Wang, G, Wang, Y and Wang, Y (2017) Genetic polymorphisms and clopidogrel efficacy for acute ischemic stroke or transient ischemic attack: A systematic review and meta-analysis. Circulation 135(1), 2133.CrossRefGoogle ScholarPubMed
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 and 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(8), 761771.CrossRefGoogle Scholar
Perera, MA, Cavallari, LH, Limdi, NA, Gamazon, ER, Konkashbaev, A, Daneshjou, R, Pluzhnikov, A, Crawford, DC, Wang, J, Liu, N, Tatonetti, N, Bourgeois, S, Takahashi, H, Bradford, Y, Burkley, BM, Desnick, RJ, Halperin, JL, Khalifa, SI, Langaee, TY, Lubitz, SA, Nutescu, EA, Oetjens, M, Shahin, MH, Patel, SR, Sagreiya, H, Tector, M, Weck, KE, Rieder, MJ, Scott, SA, Wu, AH, Burmester, JK, Wadelius, M, Deloukas, P, Wagner, MJ, Mushiroda, T, Kubo, M, Roden, DM, Cox, NJ, Altman, RB, Klein, TE, Nakamura, Y and Johnson, JA (2013) Genetic variants associated with warfarin dose in African-American individuals: A genome-wide association study. Lancet 382(9894), 790796.CrossRefGoogle ScholarPubMed
Peterson, JF, Field, JR, Unertl, KM, Schildcrout, JS, Johnson, DC, Shi, Y, Danciu, I, Cleator, JH, Pulley, JM, McPherson, JA, Denny, JC, Laposata, M, Roden, DM and Johnson, KB (2016) Physician response to implementation of genotype-tailored antiplatelet therapy. Clinical Pharmacology and Therapeutics 100(1), 6774.CrossRefGoogle ScholarPubMed
Peyser, B, Perry, EP, Singh, K, Gill, RD, Mehan, MR, Haga, SB, Musty, MD, Milazzo, NA, Savard, D, Li, YJ, Trujilio, G and Voora, D (2018) Effects of delivering SLCO1B1 pharmacogenetic information in randomized trial and observational settings. Circulation: Genomic and Precision Medicine 11(9), e002228.Google ScholarPubMed
PharmGKB (2023a) Drug Label Information and Legend. Available at https://www.pharmgkb.org/page/clinAnnLevels (accessed 20 March 2023).Google Scholar
PharmGKB (2023b) Very Important Pharmacogenes. Available at https://www.pharmgkb.org/vips (accessed 20 March 2023).Google Scholar
Pirmohamed, M, Burnside, G, Eriksson, N, Jorgensen, AL, Toh, CH, Nicholson, T, Kesteven, P, Christersson, C, Wahlstrom, B, Stafberg, C, Zhang, JE, Leathart, JB, Kohnke, H, Maitland-van der Zee, AH, Williamson, PR, Daly, AK, Avery, P, Kamali, F, Wadelius, M and Group E-P (2013) A randomized trial of genotype-guided dosing of warfarin. New England Journal of Medicine 369(24), 22942303.CrossRefGoogle ScholarPubMed
Pokorney, SD, Simon, DN, Thomas, L, Fonarow, GC, Kowey, PR, Chang, P, Singer, DE, Ansell, J, Blanco, RG, Gersh, B, Mahaffey, KW., Hylek, EM, Go, AS, Piccini, JP, Peterson, ED and Outcomes Registry for Better Informed Treatment of Atrial Fibrillation (2015) Patients’ time in therapeutic range on warfarin among US patients with atrial fibrillation: Results from ORBIT-AF registry. American Heart Journal 170(1), 141148 e1.CrossRefGoogle ScholarPubMed
Popejoy, AB, Crooks, KR, Fullerton, SM, Hindorff, LA, Hooker, GW, Koenig, BA, Pino, N, Ramos, EM, Ritter, DI, Wand, H, Wright, MW, Yudell, M, Zou, JY, Plon, SE, Bustamante, CD, Ormond, KE, Clinical Genome Resource A and Diversity Working G (2020) Clinical genetics lacks standard definitions and protocols for the collection and use of diversity measures. American Journal of Human Genetics 107(1), 7282.CrossRefGoogle ScholarPubMed
Pratt, VM, Cavallari, LH, Del Tredici, AL, Hachad, H, Ji, Y, Kalman, LV, Ly, RC, Moyer, AM, Scott, SA, Whirl-Carrillo, M and Weck, KE (2020) Recommendations for clinical warfarin genotyping allele selection: A report of the Association for Molecular Pathology and the College of American Pathologists. Journal of Molecular Diagnostics 22(7), 847859.CrossRefGoogle ScholarPubMed
Pratt, VM, Del Tredici, AL, Hachad, H, Ji, Y, Kalman, LV, Scott, SA and Weck, KE (2018) Recommendations for clinical CYP2C19 genotyping allele selection: A report of the Association for Molecular Pathology. Journal of Molecular Diagnostics 20(3), 269276.CrossRefGoogle ScholarPubMed
Price, MJ, Murray, SS, Angiolillo, DJ, Lillie, E, Smith, EN, Tisch, RL, Schork, NJ, Teirstein, PS, Topol, EJ and Investigators, G (2012) Influence of genetic polymorphisms on the effect of high- and standard-dose clopidogrel after percutaneous coronary intervention: The GIFT (genotype information and functional testing) study. Journal of the American College of Cardiology 59(22), 19281937.CrossRefGoogle ScholarPubMed
Pritchard, D, Patel, JN, Stephens, L and Mc Leod, HL (2022) Comparison of FDA Table of Pharmacogenetic Associations and Clinical Pharmacogenetics Implementation Consortium guidelines. American Society of Health-System Pharmacists 79(12), 9931005.CrossRefGoogle ScholarPubMed
Proietti, M, Romanazzi, I, Romiti, GF, Farcomeni, A and Lip, GYH (2018) Real-world use of Apixaban for stroke prevention in atrial fibrillation: A systematic review and meta-analysis. Stroke 49(1), 98106.CrossRefGoogle ScholarPubMed
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 and Niemi, M (2014) The clinical pharmacogenetics implementation consortium guideline for SLCO1B1 and simvastatin-induced myopathy: 2014 update. Clinical Pharmacology and Therapeutics 96(4), 423428.CrossRefGoogle ScholarPubMed
Relling, MV and Evans, WE (2015) Pharmacogenomics in the clinic. Nature 526(7573), 343350.CrossRefGoogle ScholarPubMed
Relling, MV, Klein, TE, Gammal, RS, Whirl-Carrillo, M, Hoffman, JM and Caudle, KE (2020) The Clinical Pharmacogenetics Implementation Consortium: 10 years later. Clinical Pharmacology and Therapeutics 107(1), 171175.CrossRefGoogle ScholarPubMed
Relling, MV, Schwab, M, Whirl-Carrillo, M, Suarez-Kurtz, G, Pui, CH, Stein, CM, Moyer, AM, Evans, WE, Klein, TE, Antillon-Klussmann, FG, Caudle, KE, Kato, M, Yeoh, AEJ, Schmiegelow, K and Yang, JJ (2019) Clinical Pharmacogenetics Implementation Consortium guideline for thiopurine dosing based on TPMT and NUDT15 genotypes: 2018 update. Clinical Pharmacology and Therapeutics 105(5), 10951105.CrossRefGoogle ScholarPubMed
Roden, DM, Van Driest, SL, Mosley, JD, Wells, QS, Robinson, JR, Denny, JC and Peterson, JF (2018) Benefit of preemptive pharmacogenetic information on clinical outcome. Clinical Pharmacology and Therapeutics 103(5), 787794.CrossRefGoogle ScholarPubMed
Roden, DM and Viswanathan, PC (2005) Genetics of acquired long QT syndrome. Journal of Clinical Investigation 115(8), 20252032.CrossRefGoogle ScholarPubMed
Roffi, M, Patrono, C, Collet, JP, Mueller, C, Valgimigli, M, Andreotti, F, Bax, JJ, Borger, MA, Brotons, C, Chew, DP, Gencer, B, Hasenfuss, G, Kjeldsen, K, Lancellotti, P, Landmesser, U, Mehilli, J, Mukherjee, D, Storey, RF, Windecker, S and Group ESCSD (2016) 2015 ESC guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: Task force for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation of the European Society of Cardiology (ESC). European Heart Journal 37(3), 267315.CrossRefGoogle ScholarPubMed
Rollini, F, Franchi, F and Angiolillo, DJ (2016) Switching P2Y12-receptor inhibitors in patients with coronary artery disease. Nature Reviews. Cardiology 13(1), 1127.CrossRefGoogle ScholarPubMed
Rost, S, Fregin, A, Ivaskevicius, V, Conzelmann, E, Hortnagel, K, Pelz, HJ, Lappegard, K, Seifried, E, Scharrer, I, Tuddenham, EG, Muller, CR, Strom, TM and Oldenburg, J (2004) Mutations in VKORC1 cause warfarin resistance and multiple coagulation factor deficiency type 2. Nature 427(6974), 537541.CrossRefGoogle ScholarPubMed
Rouini, MR and Afshar, M (2017) Effect of CYP2D6 polymorphisms on the pharmacokinetics of propafenone and its two main metabolites. Thérapie 72(3), 373382.Google ScholarPubMed
Saldivar, JS, Taylor, D, Sugarman, EA, Cullors, A, Garces, JA, Oades, K and Centeno, J (2016) Initial assessment of the benefits of implementing pharmacogenetics into the medical management of patients in a long-term care facility. Pharmacogenomics and Personalized Medicine 9, 16.CrossRefGoogle Scholar
Sangkuhl, K, Klein, TE and Altman, RB (2010) Clopidogrel pathway. Pharmacogenetics and Genomics 20(7), 463465.CrossRefGoogle ScholarPubMed
Scheuner, MT, Sales, P, Hoggatt, K, Zhang, N, Whooley, MA and Kelley, MJ (2023) Genetics professionals are key to the integration of genetic testing within the practice of frontline clinicians. Genetics in Medicine 25(1), 103114.CrossRefGoogle Scholar
Schoonen, WM, Thomas, SL, Somers, EC, Smeeth, L, Kim, J, Evans, S and Hall, AJ (2010) Do selected drugs increase the risk of lupus? A matched case-control study. British Journal of Clinical Pharmacology 70(4), 588596.CrossRefGoogle ScholarPubMed
Schork, NJ (2015) Personalized medicine: Time for one-person trials. Nature 520(7549), 609611.CrossRefGoogle ScholarPubMed
Scott, SA, Sangkuhl, K, Stein, CM, Hulot, JS, Mega, JL, Roden, DM, Klein, TE, Sabatine, MS, Johnson, JA, Shuldiner, AR and Implementation, Clinical Pharmacogenetics (2013) Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update. Clinical Pharmacology and Therapeutics 94(3), 317323.CrossRefGoogle ScholarPubMed
Shi, J, Wang, X, Nguyen, JH, Bleske, BE, Liang, Y, Liu, L and Zhu, HJ (2016) Dabigatran etexilate activation is affected by the CES1 genetic polymorphism G143E (rs71647871) and gender. Biochemical Pharmacology 119, 7684.CrossRefGoogle ScholarPubMed
Shitara, Y (2011) Clinical importance of OATP1B1 and OATP1B3 in drug-drug interactions. Drug Metabolism and Pharmacokinetics 26(3), 220227.CrossRefGoogle ScholarPubMed
Shugg, T, Pasternak, AL, London, B and Luzum, JA (2020) Prevalence and types of inconsistencies in clinical pharmacogenetic recommendations among major U.S. sources. NPJGenomic Medicine 5, 48.Google ScholarPubMed
Smith, DM, Weitzel, KW, Elsey, AR, Langaee, T, Gong, Y, Wake, DT, Duong, BQ, Hagen, M, Harle, CA, Mercado, E, Nagoshi, Y, Newsom, K, Wright, A, Rosenberg, EI, Starostik, P, Clare-Salzler, MJ, Schmidt, SO, Fillingim, RB, Johnson, JA and Cavallari, LH (2019) CYP2D6-guided opioid therapy improves pain control in CYP2D6 intermediate and poor metabolizers: A pragmatic clinical trial. Genetics in Medicine 21(8), 18421850.CrossRefGoogle ScholarPubMed
Sorich, MJ, Rowland, A, McKinnon, RA and Wiese, MD (2014) CYP2C19 genotype has a greater effect on adverse cardiovascular outcomes following percutaneous coronary intervention and in Asian populations treated with clopidogrel: A meta-analysis. Circulation. Cardiovascular Genetics 7(6), 895902.CrossRefGoogle Scholar
Spinasse, LB, Santos, AR, Suffys, PN, Muxfeldt, ES and Salles, GF (2014) Different phenotypes of the NAT2 gene influences hydralazine antihypertensive response in patients with resistant hypertension. Pharmacogenomics 15(2), 169178.CrossRefGoogle ScholarPubMed
Stanek, EJ, Sanders, CL, Taber, KA, Khalid, M, Patel, A, Verbrugge, RR, Agatep, BC, Aubert, RE, Epstein, RS and Frueh, FW (2012) Adoption of pharmacogenomic testing by US physicians: Results of a nationwide survey. Clinical Pharmacology and Therapeutics 91(3), 450458.CrossRefGoogle ScholarPubMed
Strauss, DG, Vicente, J, Johannesen, L, Blinova, K, Mason, JW, Weeke, P, Behr, ER, Roden, DM, Woosley, R, Kosova, G, Rosenberg, MA and Newton-Cheh, C (2017) Common genetic variant risk score is associated with drug-induced QT prolongation and torsade de pointes risk: A pilot study. Circulation 135(14), 13001310.CrossRefGoogle ScholarPubMed
Sukri, A, Salleh, MZ, Masimirembwa, C and Teh, LK (2022) A systematic review on the cost effectiveness of pharmacogenomics in developing countries: Implementation challenges. Pharmacogenomics Journal 22(3), 147159.CrossRefGoogle ScholarPubMed
Swen, JJ, van der Wouden, CH, Manson, LE, Abdullah-Koolmees, H, Blagec, K, Blagus, T, Bohringer, S, Cambon-Thomsen, A, Cecchin, E, Cheung, KC, Deneer, VH, Dupui, M, Ingelman-Sundberg, M, Jonsson, S, Joefield-Roka, C, Just, KS, Karlsson, MO, Konta, L, Koopmann, R, Kriek, M, Lehr, T, Mitropoulou, C, Rial-Sebbag, E, Rollinson, V, Roncato, R, Samwald, M, Schaeffeler, E, Skokou, M, Schwab, M, Steinberger, D, Stingl, JC, Tremmel, R, Turner, RM, van Rhenen, MH, Davila Fajardo, CL, Dolzan, V, Patrinos, GP, Pirmohamed, M, Sunder-Plassmann, G, Toffoli, G and Guchelaar, HJ (2023) A 12-gene pharmacogenetic panel to prevent adverse drug reactions: An open-label, multicentre, controlled, cluster-randomised crossover implementation study. Lancet 401(10374), 347356.CrossRefGoogle ScholarPubMed
Tata, EB, Ambele, MA and Pepper, MS (2020) Barriers to implementing clinical pharmacogenetics testing in Sub-Saharan Africa. A critical review. Pharmaceutics 12(9), 809.CrossRefGoogle Scholar
Tayeh, MK, Gaedigk, A, Goetz, MP, Klein, TE, Lyon, E, McMillin, GA, Rentas, S, Shinawi, M, Pratt, VM, Scott, SA and ACMG Laboratory Quality Assurance Committee (2022) Clinical pharmacogenomic testing and reporting: A technical standard of the American College of Medical Genetics and Genomics (ACMG). Genetics in Medicine 24(4), 759768.CrossRefGoogle ScholarPubMed
Tirona, RG, Leake, BF, Merino, G and Kim, RB (2001) Polymorphisms in OATP-C: Identification of multiple allelic variants associated with altered transport activity among European- and African-Americans. Journal of Biological Chemistry 276(38), 3566935675.CrossRefGoogle ScholarPubMed
Turner, RM, Fontana, V, Zhang, JE, Carr, D, Yin, P, FitzGerald, R, Morris, AP and Pirmohamed, M (2020) A genome-wide association study of circulating levels of atorvastatin and its major metabolites. Clinical Pharmacology and Therapeutics 108(2), 287297.CrossRefGoogle ScholarPubMed
Turner, RM and Pirmohamed, M (2019) Statin-related myotoxicity: A comprehensive review of pharmacokinetic, pharmacogenomic and muscle components. Journal of Clinical Medicine 9(1), 22.CrossRefGoogle ScholarPubMed
Turongkaravee, S, Jittikoon, J, Rochanathimoke, O, Boyd, K, Wu, O and Chaikledkaew, U (2021) Pharmacogenetic testing for adverse drug reaction prevention: Systematic review of economic evaluations and the appraisal of quality matters for clinical practice and implementation. BMC Health Services Research 21(1), 1042.CrossRefGoogle ScholarPubMed
van der Wouden, CH, Cambon-Thomsen, A, Cecchin, E, Cheung, KC, Davila-Fajardo, CL, Deneer, VH, Dolzan, V, Ingelman-Sundberg, M, Jonsson, S, Karlsson, MO, Kriek, M, Mitropoulou, C, Patrinos, GP, Pirmohamed, M, Samwald, M, Schaeffeler, E, Schwab, M, Steinberger, D, Stingl, J, Sunder-Plassmann, G, Toffoli, G, Turner, RM, van Rhenen, MH, Swen, JJ, Guchelaar, HJ and Ubiquitous Pharmacogenomics (2017) Implementing pharmacogenomics in Europe: Design and implementation strategy of the ubiquitous pharmacogenomics consortium. Clinical Pharmacology and Therapeutics 101(3), 341358.CrossRefGoogle ScholarPubMed
Varenhorst, C, James, S, Erlinge, D, Brandt, JT, Braun, OO, Man, M, Siegbahn, A, Walker, J, Wallentin, L, Winters, KJ and Close, SL (2009) Genetic variation of CYP2C19 affects both pharmacokinetic and pharmacodynamic responses to clopidogrel but not prasugrel in aspirin-treated patients with coronary artery disease. European Heart Journal 30(14), 17441752.CrossRefGoogle Scholar
Verhoef, TI, Redekop, WK, Daly, AK, van Schie, RM, de Boer, A and Maitland-van der Zee, AH (2014) Pharmacogenetic-guided dosing of coumarin anticoagulants: Algorithms for warfarin, acenocoumarol and phenprocoumon. British Journal of Clinical Pharmacology 77(4), 626641.CrossRefGoogle ScholarPubMed
Vieira, CP, Neves, DV, Coelho, EB and Lanchote, VL (2018) Effect of CYP2D6 poor metabolizer phenotype on stereoselective nebivolol pharmacokinetics. Drug Metabolism Letters 12(1), 6870.CrossRefGoogle ScholarPubMed
Voora, D, Shah, SH, Spasojevic, I, Ali, S, Reed, CR, Salisbury, BA and Ginsburg, GS (2009) The SLCO1B1*5 genetic variant is associated with statin-induced side effects. Journal of the American College of Cardiology 54(17), 16091616.CrossRefGoogle ScholarPubMed
Wada, Y, Yang, T, Shaffer, CM, Daniel, LL, Glazer, AM, Davogustto, GE, Lowery, BD, Farber-Eger, EH, Wells, QS and Roden, DM (2022) Common ancestry-specific ion channel variants predispose to drug-induced arrhythmias. Circulation 145(4), 299308.CrossRefGoogle ScholarPubMed
Wadelius, M, Chen, LY, Lindh, JD, Eriksson, N, Ghori, MJ, Bumpstead, S, Holm, L, McGinnis, R, Rane, A and Deloukas, P (2009) The largest prospective warfarin-treated cohort supports genetic forecasting. Blood 113(4), 784792.CrossRefGoogle ScholarPubMed
Wallentin, L, Becker, RC, Budaj, A, Cannon, CP, Emanuelsson, H, Held, C, Horrow, J, Husted, S, James, S, Katus, H, Mahaffey, KW, Scirica, BM, Skene, A, Steg, PG, Storey, RF, Harrington, RA, Investigators, P, Freij, A and Thorsen, M (2009) Ticagrelor versus clopidogrel in patients with acute coronary syndromes. New England Journal of Medicine 361(11), 10451057.CrossRefGoogle ScholarPubMed
Wallentin, L, James, S, Storey, RF, Armstrong, M, Barratt, BJ, Horrow, J, Husted, S, Katus, H, Steg, PG, Shah, SH, Becker, RC and investigators, P (2010) 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 376(9749), 13201328.CrossRefGoogle ScholarPubMed
Wang, Y, Zhao, X, Lin, J, Li, H, Johnston, SC, Lin, Y, Pan, Y, Liu, L, Wang, D, Wang, C, Meng, X, Xu, J, Wang, Y and investigators, C (2016) Association between CYP2C19 loss-of-function allele status and efficacy of clopidogrel for risk reduction among patients with minor stroke or transient ischemic attack. JAMA 316(1), 7078.CrossRefGoogle ScholarPubMed
Weber, WW and Hein, DW (1985) N-acetylation pharmacogenetics. Pharmacological Reviews 37(1), 2579.Google ScholarPubMed
Whelton, PK, Carey, RM, Aronow, WS, Casey, DE, Collins, KJ, Dennison Himmelfarb, C, DePalma, SM, Gidding, S, Jamerson, KA, Jones, DW, MacLaughlin, EJ, Muntner, P, Ovbiagele, B, Smith, SC, Spencer, CC, Stafford, RS, Taler, SJ, Thomas, RJ, Williams, KA Sr, Williamson, JD and Wright, JT Jr (2018) 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: Executive summary: A report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. Circulation 138(17), e426e483.Google Scholar
White, HL, de Boer, RA, Maqbool, A, Greenwood, D, van Veldhuisen, DJ, Cuthbert, R, Ball, SG, Hall, AS, Balmforth, AJ and Group M-HS (2003) An evaluation of the beta-1 adrenergic receptor Arg389Gly polymorphism in individuals with heart failure: A MERIT-HF sub-study. European Journal of Heart Failure 5(4), 463468.Google ScholarPubMed
Winner, JG, Carhart, JM, Altar, CA, Goldfarb, S, Allen, JD, Lavezzari, G, Parsons, KK, Marshak, AG, Garavaglia, S and Dechairo, BM (2015) Combinatorial pharmacogenomic guidance for psychiatric medications reduces overall pharmacy costs in a 1 year prospective evaluation. Current Medical Research and Opinion 31(9), 16331643.CrossRefGoogle Scholar
Zabalza, M, Subirana, I, Sala, J, Lluis-Ganella, C, Lucas, G, Tomas, M, Masia, R, Marrugat, J, Brugada, R and Elosua, R (2012) Meta-analyses of the association between cytochrome CYP2C19 loss- and gain-of-function polymorphisms and cardiovascular outcomes in patients with coronary artery disease treated with clopidogrel. Heart 98(2), 100108.CrossRefGoogle ScholarPubMed
Zhou, Y and Lauschke, VM (2022) Population pharmacogenomics: An update on ethnogeographic differences and opportunities for precision public health. Human Genetics 141(6), 11131136.CrossRefGoogle ScholarPubMed
Zhou, Y, Nevosadova, L, Eliasson, E and Lauschke, VM (2023) Global distribution of functionally important CYP2C9 alleles and their inferred metabolic consequences. Human Genomics 17(1), 15.CrossRefGoogle ScholarPubMed
Zhu, J, Alexander, GC, Nazarian, S, Segal, JB and Wu, AW (2018) Trends and variation in oral anticoagulant choice in patients with atrial fibrillation, 2010–2017. Pharmacotherapy 38(9), 907920.CrossRefGoogle ScholarPubMed
Zhu, Y, Swanson, KM, Rojas, RL, Wang, Z, St Sauver, JL, Visscher, SL, Prokop, LJ, Bielinski, SJ, Wang, L, Weinshilboum, R and Borah, BJ (2020) Systematic review of the evidence on the cost-effectiveness of pharmacogenomics-guided treatment for cardiovascular diseases. Genetics in Medicine 22(3), 475486.CrossRefGoogle ScholarPubMed
Zisaki, A, Miskovic, L and Hatzimanikatis, V (2015) Antihypertensive drugs metabolism: An update to pharmacokinetic profiles and computational approaches. Current Pharmaceutical Design 21(6), 806822.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. CYP2C19 allele dependent enzyme activity

Figure 1

Figure 1. Pharmacogenomic implementation. The top panel shows the range of stakeholders, technology, knowledge and evidence that need to be harnessed to realise the value of PGx. The middle panel depicts the uses of PGx in the clinical prescribing pathway. The bottom panel presents the applications of PGx. CPIC, the Clinical Pharmacogenetics Implementation Consortium; DPWG, Dutch Pharmacogenetics Working Groups; PharmGKB, the Pharmacogenomics Knowledge Base.

Author comment: Cardiovascular precision medicine – A pharmacogenomic perspective — R0/PR1

Comments

No accompanying comment.

Review: Cardiovascular precision medicine – A pharmacogenomic perspective — R0/PR2

Conflict of interest statement

Reviewer declares none.

Comments

Title: Cardiovascular precision medicine – a pharmacogenomic perspective

Authors: Sandosh Padmanabhan et al

Although there are many published reviews on the pharmacogenetics and pharmacogenomics of drugs used in cardiovascular diseases, I found this review to be largely comprehensive, up to date and concise. However, I believe that the readers would benefit from including a section on future perspectives in CVD pharmacogenomics including approaches to expedite the implementation of PGx in diverse healthcare systems (i.e. in developed and developing countries).

The global geographical distribution of alleles and phenotypes of most drug metabolizing enzymes have been covered extensively in the literature (PMID: 34652573, PMID: 36855170 as examples) and I am therefore wondering if figures 1-3 are necessary? Are the data presented here presenting anything new or distinct from what have been published?

Minor point

On page 5, the authors state “ …. pharmacogenomics (PGx), the study of the effect of inherited genetic variation on drug….” I believe that acquired variations are also relevant to pharmacogenomics.

Recommendation: Cardiovascular precision medicine – A pharmacogenomic perspective — R0/PR3

Comments

No accompanying comment.

Decision: Cardiovascular precision medicine – A pharmacogenomic perspective — R0/PR4

Comments

No accompanying comment.

Author comment: Cardiovascular precision medicine – A pharmacogenomic perspective — R1/PR5

Comments

Dear Editor-in-Chief,

We are pleased to submit our revised article taking into consideration all the reviewer comments. We have added a new figure and highlighted the changes made in the text. We are ver grateful for considering our manuscript.

Yours sincerely

Anna Dominiczak and co-authors

Review: Cardiovascular precision medicine – A pharmacogenomic perspective — R1/PR6

Conflict of interest statement

Reviewer declares none.

Comments

The authors have addressed my previous comments adequately.

Recommendation: Cardiovascular precision medicine – A pharmacogenomic perspective — R1/PR7

Comments

No accompanying comment.

Decision: Cardiovascular precision medicine – A pharmacogenomic perspective — R1/PR8

Comments

No accompanying comment.