Open access peer-reviewed chapter - ONLINE FIRST

Pharmacogenomics of Cardiovascular Diseases: The Path to Precision Therapy

Written By

Georges Nemer and Nagham Nafiz Hendi

Submitted: 07 September 2023 Reviewed: 18 September 2023 Published: 14 October 2023

DOI: 10.5772/intechopen.113236

Pharmacogenomics and Pharmacogenetics in Drug Therapy IntechOpen
Pharmacogenomics and Pharmacogenetics in Drug Therapy Edited by Madhu Khullar

From the Edited Volume

Pharmacogenomics and Pharmacogenetics in Drug Therapy [Working Title]

Prof. Madhu Khullar, Dr. Anupam Mittal and Associate Prof. Amol Patil

Chapter metrics overview

76 Chapter Downloads

View Full Metrics

Abstract

Cardiovascular diseases (CVD) represent a substantial global health burden, leading to significant morbidity and mortality rates. However, the efficacy and safety of CVD therapies are markedly influenced by individual variability in drug responses and adverse reactions, often attributable to genetic factors. This chapter discusses how pharmacogenomics impacts the safety and efficacy of cardiovascular therapies through advanced genetic testing methods, like genome-wide association studies, polygenic risk scores, and multi-omics analyses. Additionally, the chapter addresses challenges and future perspectives, with a focus on the role of artificial intelligence and machine learning in integrating pharmacogenomics and genotype-based personalized interventions into the routine CVD care to improve long-term health outcomes.

Keywords

  • pharmacogenomics
  • cardiovascular therapies
  • gene polymorphisms
  • precision medicines
  • genome-wide association studies

1. Introduction

Cardiovascular diseases (CVD) remain the leading cause of global disability and mortality, responsible for about 38% of all deaths and straining healthcare systems. These disorders influence the heart and blood circulations, including coronary heart disease, peripheral arterial disease, cerebrovascular disease, and other conditions [1]. However, the response to CVD drugs varies significantly among individuals, leading to suboptimal outcomes and potential adverse reactions. Genetics plays a vital role in drug response variability, with specific CVD therapies showing up to 90% dependence on genetic factors [2]. Given the multifactorial nature of CVD, which involves genetic, environmental, and lifestyle influences, precise and personalized therapeutic strategies become essential for effective management [3].

In recent years, pharmacogenomics has emerged as a promising approach in cardiovascular therapy, offering new opportunities for safe and effective targeted therapies. The field of pharmacogenomics focuses on understanding how genetic variations influence an individual’s response to medications. Genetic variations primarily alter cardiovascular drug’s concentration (pharmacokinetics), mechanism of action (pharmacodynamics), and the underlying mechanism of CVD [2]. By identifying the genetic factors affecting drug metabolism, receptor interactions, and signaling pathways, pharmacogenomics paves the way for tailored cardiovascular treatment regimens aligned with an individual’s genetic profile [4]. This holds particular significance due to the widespread prevalence of CVD, affecting approximately 523 million individuals [1].

The significant advancements in genome-wide association studies (GWAS) and ‘omic’ technologies, such as proteomics, metabolomics, and transcriptomics, have identified numerous genetic loci linked to cardiovascular conditions, deepening our understanding of the genetic basis of CVD [5, 6]. This information has facilitated pharmacogenomics research, bringing us closer to the goal of precision medicine in managing the global health challenges caused by these diseases [7]. This chapter focuses on utilizing genetic information to optimize standard cardiovascular therapy and improve patient outcomes. It provides comprehensive discussions on key cardiovascular pharmacogenomics, including antithrombotic (antiplatelets and anticoagulants), lipid-lowering agents (statins), and antihypertensive treatments (beta-blockers, angiotensin-converting enzyme inhibitors (ACEI), angiotensin II receptor blockers (ARBs), vasodilators, and diuretics), along with future directions in this field.

Advertisement

2. Cardiovascular pharmacogenetics

The influence of genetic makeup on individual response to cardiovascular medications has gained substantial recognition. By 2023, genetic information had been integrated into the Food and Drug Administration (FDA, https://www.fda.gov/drugs/science-and-research-drugs/table-pharmacogenomic-biomarkers-drug-labeling) approved labeling for around 15 cardiovascular treatments. The Clinical Pharmacogenetics Implementation Consortium (CPIC, https://cpicpgx.org/genes-drugs/) [8] and Dutch Pharmacogenetics Working Group (DPWG, https://www.knmp.nl/dossiers/farmacogenetica) [9] provide authoritative guidance through the Pharmacogenomics Knowledgebase (PharmGKB, https://www.pharmgkb.org) on integrating genetics into drug prescribing [10]. These organizations offer evidence-based guidelines for specific gene-drug pairs, optimizing pharmacotherapy decisions in the clinical practice. CPIC and PharmGKB categorize evidence from A (strong) to D (limited/conflicting) [8, 10]. CPIC has issued level A guidelines for several cardiovascular drugs, including clopidogrel, warfarin, and simvastatin [8]. Table 1 provides an overview of pharmacogenetic biomarkers and clinical recommendations derived from prevalent guidelines for cardiovascular medications.

MedicationsBiomarkerSNP IDGenotypeEffect on response/safetyClinical recommendationsGuideline
Antiplatelets
Acetylsalicylic acid (Aspirin) & abciximabPTGS1rs3842787TReduce responseMonitor for thrombosis in patients with resistance.PharmGKB
PTGS2rs20417C
GPIIIa PlArs2317676AGReduce response & increase CVEConsider higher doses of Aspirin & abciximab.
ITGA2rs1126643CT/TTReduce responseLimited evidence & require further validation.
rs1062535AA/AG
CYP2C9*1Increase responseConsider lower doses of Aspirin & abciximab.CPIC
rs4244285*2
rs4986893*3
P2Y1Rrs1065776TT/CTLimited evidence & require further validation.PharmGKB
ClopidogrelCYP2C19rs4244285*2Reduce response & increase CVEConsider alternative antiplatelet agents, such as prasugrel or ticagrelor.CPIC, DPWG, & FDA “Black Box”
rs4986893*3
rs3758581*17Increase response & AE (bleeding risk)Limited evidence & require further validation.
rs28399504*4Reduce response & increase CVE
rs56337013*5
rs72552267*6
rs72558186*7
rs41291556*8
rs17884712*9
rs140278421*10
rs6413438*22
rs118203757*24
rs12769205*35
ABCB1rs1045642AA/AGInconsistent & require further validation especially on adverse CVE.PharmGKB
PON1rs662TTCPIC
CYP3A5rs776746*3Inconsistent effectRequire further validation.CPIC
CYP2C9rs1799853*2Reduce responseLimited evidence & require further validation.CPIC
rs1057910*3Reduce response & increase CVECPIC
Prasugrel & ticagrelorCYP2C19rs3758581*17Increase response & AE (bleeding risk)DPWG & FDA
CYP3A4rs35599367*22Inconsistent effect
CYP4F2rs3093135TTInconsistent reduce response & increase CVE (MI & CAS)
SLCO1B1rs113681054CInconsistent increase response
Anticoagulants
WarfarinCYP2C9rs1799853*2Increase response & AEConsider lower warfarine dose & more frequently INR monitoring.CPIC, DPWG, & FDA
rs1057910*3
rs12777823*8Consider 15–30% lower warfarine dose & more frequently INR monitoring in African.
rs56165452*4
rs28371686*5
rs9332131*6
rs28371685*11
CYP2C19rs11188082T
CYP4F2rs2108622*3Reduce responseConsider 5–10% higher warfarine dose in European & Asian.CPIC
VKORC1rs9923231TTIncrease response & AE (bleeding risk)Consider lower warfarine dose & more frequently INR monitoring.CPIC, DPWG, & FDA
rs749671G
rs7294CT/TT
rs9934438AG/GG
AcenocoumarolCYP2C9rs1799853*2Increase response & AEConsider lower acenocoumarol dose & more frequently INR monitoring, especially with NSAID.
CYP4F2rs2108622*3Reduce responseConsider higher acenocoumarol dose.DPWG
VKORC1rs9923231AG/GGIncrease response & AE (bleeding risk)Consider 50% lower acenocoumarol dose & more frequent INR monitoring, especially with NSAID.
HeparinITGB3, FCGR2A, TDAG8, PTPRJ, IL10, KIR2DS5, HLA-DRA, HLA-DR*3Increase risk of heparin-induced thrombocytopeniaLimited evidence & require further validation.PharmGKB
Direct oral anticoagulants (apixaban, dabigatran, edoxaban, & rivaroxaban)ABCB1rs4148738CT/TTReduce response & increase CVE (thromboembolism)
rs1045642AA
CES1rs2244613GT/TTIncrease response AE (bleeding risk)
AGTrs699TTReduce response
PRKCQrs500766CT/TTReduced AE
ETV6rs2724635AA/AG
CYP3A4, CES1, SLCO1B1, ABCG2, & SULTIncrease response & AEAvoid combining this medication with potent CYP3A4/5 inhibitors, require further validation.FDA
Lipid-lowering agents
PravastatinKIF6rs20455AA/GAIncrease response & reduce CVELimited evidence & require further validation.PharmGKB
SLCO1B1rs4149015*17Increase response & AE (statin-induced myopathy)Consider patient genotyping prior to dosing with lowering the initial dose.CPIC, DPWG, & FDA
rs2306283
rs4149056
Atorvastatin, simvastatin, rosuvastatin, pravastatin, & lovastatin, pitavastatinrs4149056*5
rs2306283*14
rs4363657
rs4149056
rs2306283*3
rs11045819
Fluvastatinrs4149056
CYP2C9rs1057910*3Increase responseCPIC guidelines now cover fluvastatin dosing.CPIC
Atorvastatin, simvastatin, rosuvastatin, pravastatin, & lovastatin, pitavastatinCYP3A5rs776746*3Inconsistent effectLimited evidence & require further validation.PharmGKB
CYP3A4rs35599367*22
rs55951658*4
rs4986910*3Reduce response
rs2740574*1B
rs2242480*1G
CYP3A7rs45446698G
ABCB1rs1128503AAIncrease response
rs2032582
rs1045642
ABCG2rs2231142CCConsider lower statin dose, alternatives, or combination therapy (e.g., statin + ezetimibe).CPIC
UGT1Ars887829*2Reduce responseLimited evidence & require further validation.PharmGKB
HMGCRrs17244841ATReduce response
CLMNrs8014194AVariable (increase) response
APOErs429358E4Increase response & reduce CVE
rs7412E2
CETPrs708272AAReduce response & increase CVE
Beta-blockers
Atenolol, bisoprolol, carvedilol, metoprolol, nebivolol, & propranololADRB1rs1801252AG/GGReduce response & increase CVELimited evidence & require further validation.PharmGKB
rs1801253
ADRB2rs1042713GG
rs1042714CG/GG
rs1800888AA
ADD1rs4961GT/TTIncrease response & reduce CVE
NPPArs5065GG
rs4149601AAPharmGKB
rs292449CG/GG
NEDD4Lrs75982813AG/GG
GRK5rs2230345TT/ATIncrease response & reduce CVE
CYP2D6rs1135840GGIncrease response & AE (bradycardia)FDA
CYP2D6rs1065852AG/GGReduce response
Direct-acting vasodilators
Hydralazine, minoxidil, & sodium nitroprussideNAT2*4Reduce responseLimited evidence & require further validation.PharmGKB
rs1801280*5DIncrease response & AE (lupus-like symptoms)
rs1799930*6B
rs1799931*7A
rs1801279*14B
Renin-angiotensin system inhibitors
ACEI (saptopril, enalapril, & lisinopril)ACErs4646994DDReduce response & increase CVE (mortality)Limited evidence & require further validation.PharmGKB
AGTrs699AAReduce response & increase CVE (blood pressure)
PRKCQrs500766CCIncrease AE (angioedema)
ETV6rs2724635GG
ARBs (candesartan, losartan, & olmesartan)AGTR1rs5186AAIncrease response
Diuretics
Aldosterone antagonist (Spironolactone)ADD1rs4961GG/GTIncrease response & reduce CVE (MI)Limited evidence & require further validation.PharmGKB
Loop diuretics (Furosemide)
Thiazide diuretics (Hydrochlorothiazide, Chlorthalidone & indapamide)
NPPArs5065GG
rs4149601CC/CGIncrease response
rs292449
NEDD4Lrs75982813AG/GG
PRKCArs16960228A
YEATS4rs7297610CC
Antiarrhythmics
Amiodarone, flecainide, sotalolKCNE1rs1805128TIncrease AD (DITdP risk)Limited evidence & require further validation.PharmGKB
SCN5Ars7626962GT/TTReduce response & increase CVE (arrhythmias)
NOS1APrs10919035TIncrease AD (ventricular arrhythmia & QT prolongation risk)
ProcainamideNAT2Increase response & AEFDA
Amiodarone, Sotalol flecainide, quinidine, & propafenonePITX2rs10033464GT/TTReduced response & CVE (atrial fibrillation recurrence)PharmGKB
ADRB1rs1801253CCIncrease response
Quinidine & propafenoneCYP2D6 & CYP3A4Increase risk (pro-arrhythmia)Consider patient genotyping prior to dosing with lowering the initial dose.DPWG & FDA

Table 1.

Pharmacogenomics and clinical recommendations of cardiovascular medications based on common guidelines.

The sources of drug-gene interactions are from common pharmacogenomic guidelines, including PharmGKB (https://www.pharmgkb.org), CPIC (Clinical Pharmacogenetics Implementation Consortium, https://cpicpgx.org/genes-drugs/), DPWG (Dutch Pharmacogenetics Working Group, https://www.knmp.nl/dossiers/farmacogenetica), & FDA (U.S. Food & Drug Administration, https://www.fda.gov/drugs/science-&-research-drugs/table-pharmacogenomic-biomarkers-drug-labeling). Abbreviations: CVE, cardiovascular events; AE, adverse effect; DITdP, drug-induced Torsade de Pointes; MI, myocardial infarction; SNP, single nucleotide polymorphism.

2.1 Antiplatelets

Anti-platelet therapy stands as a cornerstone in managing CVDs, effectively preventing thrombotic incidents of acute coronary syndromes (ACS) and post-percutaneous coronary intervention (PCI) [11]. Gene polymorphisms potently influence various individual responses to this treatment, encompassing acetylsalicylic acid (aspirin), thienopyridine derivatives (clopidogrel, ticlopidine, ticagrelor, and prasugrel), and GP IIb/IIIa receptor inhibitors (tirofiban, lamifiban, epifibatide, and abciximab) [12].

Acetylsalicylic acid modifies cyclooxygenase (COX1) activities to lower thromboxane A2 levels and prevent platelet aggregation. Despite acetylsalicylic acid use, resistance leads to thrombosis in 5–57% of patients [13]. Notably, two specific variants in the genes encoding COX1 and COX2, rs3842787 in prostaglandin-Endoperoxide Synthase (PTGS1) and rs20417 in PTGS2 affect thromboxane B2 levels [14, 15, 16]. The ITGB3 gene encoding glycoprotein (GP)IIIa harbors different variants (Pro33Leu, rs2317676GG/AG) that necessitate higher aspirin and abciximab doses and augment major ischemic incidents [14]. Genetic variants in ITGA2 encoding the glycoprotein Ia (GPIa) (rs1126643 and rs1062535) heighten platelet reactivity [16], while the purinergic (P2RY1) receptor variant (rs1065776TT/CT, C893T) reduces platelet aggregation [14]. These genetic insights underscore aspirin’s complexity in patients’ outcomes.

The prodrug clopidogrel, a widely used medication, is metabolized and activated by hepatic cytochrome P450 (CYP) enzymes, majorly CYP2C19, CYP3A5, and CYP3A4 (Figure 1) [12]. A widely observed loss-of-function variant of CYP2C19*2, rs4244285, has been linked to diminished treatment responses and attenuated anti-platelet effects through genome-wide association studies. Individuals with homozygous loss-of-function alleles, such as CY2C19*2/*2, display a complete lack of enzyme functionality and poor metabolism compared to those with reference CY2C19*1/*1 or heterozygous CY2C19*1/*2 alleles, which show extensive or intermediate metabolism [17]. This polymorphism is most prevalent among Asian, African, and European populations, constituting approximately 33%, 18%, and 17% of their respective allele frequencies (Table 2) [18].

Figure 1.

Pharmacogenetic associations supported by clinical evidence with cardiovascular medications. The pharmacokinetic and pharmacodynamic of cardiovascular medications, including antiplatelets, anticoagulants, DOACs, statins, beta-blockers, direct-acting vasodilators (vasodilators), and antiarrhythmics, are influenced by genetic variations within essential genes. These genes include members of cytochrome P450 family (CYP2C9, CYP2C19, CYP3A5, CYP4F2, and CYP2D6), as well as PON1, VKORC1, SLCO1B1, ABCG2, and NAT2. The illustration is generated with BioRender.com.

GeneSNP IDConsequencesAllele frequency in different populationsHGVScFunctional role
GlobalAfricanEuropeanAmericanSouth AsianMiddle Eastern
ABCB1rs4148738Intron0.6110.7700.5400.6030.4030.570ENST00000622132.5:c.2482-2236G>TTransports molecules and drugs across cellular membranes.
rs1045642Missense0.5720.7970.4690.5680.4110.519ENST00000622132.5:c.3435T>G
ABCG2rs2231142Missense0.0000.0000.0000.0000.0000.000ENST00000237612.8:c.421C>G
ADD1rs4961Missense0.1690.0790.1940.1820.1890.127ENST00000683351.1:c.1378G>APart of the cytoskeleton and regulates ion transport, mainly in renal cells.
ADRB1rs1801253Missense0.6960.5920.7320.7760.7380.624ENST00000369295.4:c.1165G>AMediates the physiological effects of the epinephrine and norepinephrine, mainly in heart.
rs1801252Missense0.1670.2270.1300.2160.1250.067ENST00000369295.4:c.145A>G
ADRB2rs1800888Missense0.0090.0020.0140.0130.0040.025ENST00000305988.6:c.491C TRegulates specific and rapid signaling by the G protein-coupled receptor and gene transcription.
rs1042714Stop gained0.6820.8210.6320.7570.7940.760ENST00000305988.6:c.79G>T
rs1042713Missense0.4260.4940.3720.4350.4420.402ENST00000305988.6:c.46G>A
AGTR1rs51863 prime UTR0.2060.0630.2960.2580.0810.206ENST00000349243.8:c.*86A>CRegulates the cardiovascular effects of angiotensin II.
APOErs7412Missense0.0780.1050.0790.0420.0370.045ENST00000252486.9:c.526C>TCritical for the catabolism of atherogenic lipoprotein constituents.
rs429358Missense0.1570.2150.1380.1120.1050.067ENST00000252486.9:c.388T>C
CES1rs2244613Intron0.7690.7550.8160.7600.5920.740ENST00000360526.8:c.1171-33C>TRegulates drugs hydrolysis or transesterification.
CETPrs708272Intron0.3850.2640.4350.4190.4610.402ENST00000200676.8:c.118 + 279G>ATransfer cholesteryl ester from high density lipoprotein (HDL) to other lipoproteins.
CTBP2P2rs12777823Downstream gene0.1900.2500.1510.1410.3340.095A transcriptional repressor, regulates epithelial to mesenchymal transition.
CYP2C19rs72558186Splice donorENST00000371321.9:c.819 + 2T>ACatalyze drug metabolism and synthesis reactions.
rs72552267Missense0.0000.0000.0000.0010.0000.000ENST00000371321.9:c.395G>A
rs6413438Missense0.0010.0030.0000.0010.0000.000ENST00000371321.9:c.680C>T
rs56337013Missense0.0000.0000.0000.0000.0000.000ENST00000371321.9:c.1297C>T
rs4986893Stop gained0.0030.0000.0000.0010.0050.000ENST00000371321.9:c.636G>A
rs4244285Synonymous0.1680.1770.1480.1300.3310.087ENST00000371321.9:c.681G>A
rs41291556Missense0.0020.0000.0030.0010.0010.000ENST00000371321.9:c.358T>C
rs3758581Missense0.9520.9870.9370.9530.8860.927ENST00000371321.9:c.991A>C
rs28399504Start lost0.0020.0010.0030.0060.0010.000ENST00000371321.9:c.1A>G
rs17884712Missense0.0040.0120.0000.0010.0000.000ENST00000371321.9:c.431G>A
rs140278421Missense0.0000.0000.0000.0000.0000.000ENST00000371321.9:c.557G>A
rs12769205Intron0.1730.1970.1480.1330.3300.092ENST00000371321.9:c.332-23A>G
rs118203757Missense0.0000.0010.0000.0000.0000.000ENST00000371321.9:c.1004G>A
rs11188082Intron0.9100.8400.9300.9400.8900.916ENST00000371321.9:c.820-12218A>C
CYP2C9rs1799853Missense0.0880.0240.1270.0980.0390.136ENST00000260682.8:c.430C>T
rs1057910Missense0.0490.0130.0660.0490.1140.073ENST00000260682.8:c.1075A>C
CYP2D6rs1135840Missense0.5770.6430.5590.4900.5520.621ENST00000645361.2:c.1457G>C
rs1065852Missense0.1910.1250.2230.1530.1680.114ENST00000645361.2:c.100C>T
CYP3A4rs4986910Missense0.0050.0010.0070.0020.0000.000ENST00000651514.1:c.1334T>C
rs35599367Intron0.0320.0090.0490.0250.0090.016ENST00000651514.1:c.522-191C>T
rs2242480Intron0.3100.7320.0940.3580.3460.228ENST00000651514.1:c.1026 + 12G>A
CYP3A5rs776746Intron0.7290.3050.9310.7800.6990.880ENST00000222982.8:c.219-237A>G
CYP3A7rs45446698Upstream gene0.0260.0100.0360.0280.0140.022
CYP4F2rs3093135Intron0.1510.1360.1660.1540.1560.174ENST00000221700.11:c.344-979T>A
rs2108622Missense0.2290.0990.2860.2580.4020.395ENST00000221700.11:c.1297G>C
ETV6rs2724635Intron0.5560.3960.6210.6610.4930.669ENST00000396373.9:c.34-5411G>AMaintenance hematopoiesis and vascular network development.
GRK5rs2230345Missense0.0860.2610.0130.0450.0680.047ENST00000392870.3:c.122A>TPhosphorylates the active G protein-coupled receptors, with unclear mechanisms in cardiovascular diseases.
ITGB3rs23176763 prime UTR0.0960.1330.0730.0870.1180.165ENST00000559488.7:c.*713A>GRegulates platelet function
KCNE1rs1805128MissenseENST00000399286.3:c.253G>AModulates potassium ions flow through ear and cardiac channels.
KIF6rs20455Missense0.4890.7980.3610.3860.4450.443ENST00000287152.12:c.2155T>CTransport cellular intracellular transport of protein complexes.
PITX2rs10033464Downstream gene0.8590.8190.9040.8170.7820.908Guiding the development of asymmetric cardiac morphology.
NAT2rs1801280Missense0.3820.3050.4470.3520.3380.452ENST00000286479.4:c.341T>CCatalyzes the acetylation, and thus, activation of drugs.
rs1801279Missense0.0240.0830.0000.0110.0000.003ENST00000286479.4:c.191G>A
rs1799931Missense0.0410.0330.0260.0960.0740.048ENST00000286479.4:c.857G>A
rs1799930Missense0.2730.2590.2920.2070.3660.287ENST00000286479.4:c.590G>A
NEDD4Lrs75982813Upstream gene0.0630.0910.0610.0410.0370.032Regulates the surface expression of renal epithelial sodium channels by ubiquitin-mediated endocytosis and lysosomal targeting.
rs4149601Intron0.3210.3440.3350.2630.1890.326ENST00000400345.8:c.49-16229G>A
rs292449Intron0.4210.5140.3430.4620.4440.440ENST00000400345.8:c.123-17578G>C
NOS1APrs10919035Intron0.1980.2140.1470.2140.2650.204ENST00000361897.10:c.178-13122C>GModulates intracellular calcium, and up-regulated in dystrophic cardiomyopathy.
P2RY1rs1065776Synonymous0.3090.6750.1490.2400.2160.209Regulate platelet aggregation.
PON1rs662Missense0.4200.6700.2800.4230.3900.338ENST00000222381.8:c.575A>TDegrades oxidized lipids in LDL, potentially delaying atherosclerosis.
PRKCArs16960228Intron0.1280.3090.0460.1090.0390.060ENST00000413366.8:c.1854 + 3730G>ARegulator heart contractility
PRKCQrs500766Intron0.2980.3770.2730.2480.2760.348ENST00000263125.10:c.319-1132G>APhosphorylate proteins that regulate heart functions and rates.
PTGS1rs3842787Missense0.0820.1320.0660.0330.0170.041ENST00000362012.7:c.50C>TModulates angiogenesis in endothelial cells.
PTGS2rs20417Upstream gene0.2010.3340.1580.1800.1640.220Regulates prostagland synthesis, and functions as a dioxygenase and peroxidase.
SCN5Ars7626962Missense0.0000.0000.0000.0000.0000.000ENST00000423572.7:c.3305C>TInitiates sodium upstroke of the action potential in the heart.
SLCO1B1rs4363657Intron0.1820.1480.1710.1770.0880.193ENST00000256958.3:c.1498-1331T>ARegulates the sodium-independent uptake of different molecules and drugs.
rs4149056Missense0.1210.0320.1590.1300.0490.206ENST00000256958.3:c.521T>C
rs2306283Missense0.5320.7680.4020.5600.5160.477ENST00000256958.3:c.388A>C
rs113681054Intergenic0.1830.1530.1710.1810.0930.199
UGT1A4rs887829Intron0.3610.4490.3210.3240.4100.339ENST00000373409.8:c.868-7110C>TCatalyze drug metabolism and synthesis reactions.
VKORC1rs9934438Intron0.3190.1000.3780.3910.1790.513ENST00000394975.3:c.174-136C>TReduces of inactive vitamin K 2,3-epoxide to active vitamin K in the endoplasmic reticulum membrane.
rs9923231Upstream gene0.3190.1000.3700.3900.1780.519
rs749671Upstream gene0.3160.0990.3740.3880.1780.472
rs72943 prime UTR0.4030.4580.3830.4000.7120.250ENST00000394975.3:c.*134G>A
YEATS4rs7297610Intergenic0.1240.3030.0630.0720.0220.079Involves in transcriptional activation.

Table 2.

Functions of cardiovascular genetic factors and their allele frequencies among diverse populations.

The allele frequencies of genes associated with the pharmacogenomic of cardiovascular medications are identified using the Genome Aggregation Database (gnomAD, https://gnomad.broadinstitute.org, accessed on September 5th, 2023), and annotated using the Ensembl Effect Predictor release 108 (VEP, https://grch37.ensembl.org/index.html, accessed on September 4th, 2023). Abbreviations: HGVSc, Human Genome Variation Society sequence nomenclature descriptions from Ensembl; SNP, single nucleotide polymorphism, AF; allele frequency.

Other rare loss-of-function alleles, namely CYP2C19 (*3, rs4986893), (*4, rs28399504), (*5, rs56337013), (*6, rs72552267), (*7, rs72558186), (*8, rs41291556), (*9, rs17884712), (*10, rs6413438), (*22, rs140278421), (*24, rs118203757), and (*35, rs12769205), along with CYP2C9 (*3, rs1057910) and (*2, rs1799853) have also been identified with uncertain impact, necessitating further validation [19]. Conversely, a gain-of-function variant in the CYP2C9 (*17, rs3758581) has significantly correlated with elevated enzyme functionality, rapid metabolism, and bleeding risk [20]. This variant exhibit minor allele frequencies of 0.3, 0.93, and 0.98 in Caucasian, European, and African populations, respectively (Table 2) [18]. However, the uncertain clinical implications for carriers of *17 necessitate additional research due to inconsistent findings.

Genetic polymorphisms in the CYP2C19 gene markedly impact enzymatic activity and link to adverse cardiovascular events, including mortality, myocardial infarction (MI), and stroke [17]. Consequently, the Food and Drug Administration (FDA) issued a “Black Box” warning for clopidogrel, suggesting CYP2C19 genotyping and the consideration of alternative antiplatelet agents before prescribing antiplatelet therapies for PCI or ACS patients with a high risk of poor responses, as also recommended by CPIC and DPWG [9].

CYP3A4, CYP2C19, and CYP2B6 primarily activate Prasugrel, while CYP3A4 activates ticagrelor’s active metabolite [12]. Clinical trials indicate better cardiovascular event risk reduction with ticagrelor and prasugrel than clopidogrel in ACS [21]. Polymorphisms in the ATP-binding cassette B1 (ABCB1, rs1045642) and paraoxonase 1 (PON1, rs662) genes lower clopidogrel bioavailability, with inconsistent impact on adverse cardiovascular outcomes. Clinical guidelines exclude ABCB1 or CYP2C19 variants for prasugrel or ticagrelor [22]. However, those agents have higher bleeding rates, costs, and discontinuation [12]. Genetic variants, including CYP3A4 (*22, rs35599367), CYP3A5 (*3, rs776746), CYP4F2 (rs3093135), CYP2C19 (*17, rs3758581), and solute carrier organic anion transporter 1B1 (SLCO1B1, rs113681054) [23, 24], may impact prasugrel and ticagrelor antiplatelet outcomes, necessitating further research.

2.2 Anticoagulants

Oral anticoagulants, particularly Vitamin K antagonists and direct-acting oral anticoagulants (DOACs), are critical in preventing and treating thromboembolic disorders [12]. These medications have achieved significant success in cardiovascular therapeutics through candidate genes and GWAS investigations [25, 26].

2.2.1 Vitamin K antagonists

The narrow therapeutic index for Vitamin K antagonists, such as warfarin and acenocoumarol, necessitates close monitoring of the international normalized ratio (INR). This monitoring ensures the achievement of optimal anticoagulation within the INR range of 2–3 and prevents hemorrhage [27]. The metabolism (inactivation) of warfarin occurs predominantly in the liver through CYP2C9, with minimal involvement of CYP4F2. These therapeutic agents function as inhibitors of the vitamin K epoxide reductase complex-1 (VKORC1), subsequently impeding the biological activation of vitamin K1. VKORC1 activates coagulation factors by enhancing Vitamin K reduction, thus disrupting the biological activation of vitamin K1 (Table 2) [12].

Common genotypes, particularly CYP2C9 (*3, rs1057910), (*2, rs1799853), and rs9332238, have been detected as key contributors to variability in dose requirements of warfarin, accounting for around 50% [28]. Individuals carrying loss-of-function alleles of CYP2C9 are at elevated risk of hemorrhagic consequences and demand reduced warfarin dosages [26]. GWAS have additionally linked the *3 allele (rs2108622, V433M) variant in CYP4F2 as a distinct factor influencing variability in warfarin dosing. CYP4F2 plays a role in metabolizing vitamin K1, acting alongside VKORC1 to prevent excessive vitamin K accumulation [29]. In European and Asian populations, the *3 allele requires 5–10% higher warfarin doses than the *1 allele. However, this association is absent within African populations where CYP2C9*8 (rs12777823) and the CYP2C19 intron variant (rs11188082) are predominant, primarily due to disparities in allele frequencies [25].

Genetic mutations in the promoter region of VKORC1, commonly rs9923231 (1639G>A), rs9934438 (1173T>C), rs749671, and rs7294 (3730G>A), impact the hepatic abundance of VKORC1 mRNA transcripts and the sensitivity of warfarin [26]. Individuals with reduced VKORC1 expression levels require less warfarin doses, whereas those with gain-of-function mutations require higher doses to maintain a steady anticoagulant effect. Numerous uncommon variations in VKORC1 with non-synonymous alterations (defined as alterations to the protein’s amino acid sequence) have been associated with warfarin resistance and heightened dosage requirements [30].

Pharmacogenomics of warfarin underscores the significant role of genetic heritage on drug responses. Asian individuals often demand reduced amounts due to the prevalence of VKORC1 loss-of-function variants, while increased doses are needed for African individuals because gain-of-function VKORC1 variants are more prevalent. Among individuals of African ancestry, there are additional CYP2C9 variants (*5, rs28371686; *6, rs28371686; *8, rs9332131; and *11, rs28371685 alleles) that reduce warfarin dose requirements by 15–30% and are more common than *2 and *3 alleles compared to Caucasians (Table 2) [31]. The Clarification of Optimal Anticoagulation through Genetics (COAG) trial underscored that ignoring these genetic variations resulted in warfarin overdosing in African Americans [32].

Acknowledging the significance of these genetic patterns, the FDA has updated drug packaging to include dosage instructions that consider pharmacogenetic testing of CYP2C9 and VKORC1 genotypes. The CPIC and DPWG guidelines recommend this therapeutic approach before initiating warfarin treatment in pediatrics and adults. Studies showed that genotype-guided early warfarin treatment shortened stabilization time, improved cost-effect anticoagulation, and led to better clinical outcomes [33].

Acenocoumarol, a coumarin derivative, is primarily utilized to prevent instances of thromboembolism and deep vein thrombosis, with its effectiveness influenced by genetic polymorphisms in the CYP2C9*2 rs1799853, CYP4F2*3 rs2108622, and VKORC1 (rs9923231, 1639G>A) genes. Several algorithms predict dosage requirements of acenocoumarol based on genetic factors, ethnicity, and demographic data, such as Verde et al.’s “acenocoumarol-dose genotype score” [34, 35, 36]. DPWG guidelines emphasize close INR monitoring during non-steroidal anti-inflammatory drug (NSAID) changes for those with specific genetic profiles on acenocoumarol. The VKORC1 genotype significantly impacts dosing variability, although precise recommendations remain limited due to rigorous monitoring requirements [37].

2.2.2 Heparin

Heparin-induced thrombocytopenia (HIT) is a dangerous immune reaction to heparin anticoagulants. Predicting HIT is challenging due to limited confirmed cases and genetic complexity. Studies have inconsistently identified risk genes associated with HIT-related clotting, such as integrin beta 3 (ITGB3, encodes GPIIIa), Fc gamma receptor IIa (FCGR2A), T-cell death-associated gene-8 (TDAG8), protein tyrosine phosphatase receptor type J (PTPRJ, also known as CD148), interleukin 10 (IL10), and human leukocyte antigen DR (HLA-DRA) [38, 39, 40, 41, 42].

A GWAS identified a substantial association of missense genetic variant near the TDAG8 with elevated platelet factor 4/heparin antibodies, even in non-heparin-treated patients [43]. Candidate genomic study determined a strong link between the HLA-DRB3*01:01 allele and a higher risk of HIT. They also noted an interaction between the killer cell immunoglobulin like receptor 2 domains, short cytoplasmic tail 5 (KIR2DS5) and the HLA-C1 KIR binding group. This suggests that HLA variations and CD4+ T cells might play a role in HIT development, pending confirmation through functional assays [38]. Furthermore, comprehensive multiethnic studies with confirmed HIT cases are needed to identify genomic biomarkers for effective preventive strategies.

2.2.3 Direct oral anticoagulants

DOACs, like apixaban, dabigatran, edoxaban, and rivaroxaban, have gained popularity due to their advantageous pharmacokinetics and pharmacodynamics, eliminating the need for regular INR monitoring. However, instances of varied plasma DOAC levels and increased hemorrhagic risks have enhanced pharmacogenomic research [27]. Plasma level variation mainly arises from drug interactions and genetics, particularly depending on the absorption and activation functions of intestinal P-glycoprotein, coded by the adenosine triphosphate (ATP)-binding cassette B1 transporter/multidrug resistance-1 (ABCB1/MDR1) gene, and hepatic carboxylesterase 1 (CES1) [12].

The ABCB1 polymorphisms (such as rs1045642 and rs4148738) and CES1 polymorphism (rs2244613) inconsistently affect dabigatran levels [44]. Plasma levels of rivaroxaban and apixaban were associated significantly with genetic variations in CYP3A4, ABCB1, ATP Binding Cassette G2 (ABCG2), and sulfotransferase (SULT), while edoxaban linked to CYP3A4, CES1, SLCO1B1, and ABCB1 [45]. The FDA recently advises against combining this medication with potent CYP3A4/5 inhibitors, like carbamazepine, phenobarbital, phenytoin, or rifampin, as well as inducers, such as boceprevir, itraconazole, and clarithromycin [46]. Exploring these genetic factors, particularly in elderly patients with multiple conditions and concurrent medications, is crucial for understanding drug-gene interactions.

2.3 Lipid-lowering agents

The high occurrence of CVD, obesity, and metabolic syndrome necessitates the utilization of statins, a class of drugs known as 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) inhibitors, including atorvastatin, rosuvastatin, simvastatin, pravastatin, fluvastatin, lovastatin and pitavastatin [47]. Statins effectively decrease total and low-density lipoprotein (LDL) cholesterol levels by up to 55%, leading to a 20–30% reduction in cardiovascular events. The clinical response to statin treatment varies significantly among individuals. Approximately one-third of individuals achieve insufficient desired LDL reductions, with rare yet severe adverse consequences to the medication, including myopathy and rhabdomyolysis. Genetic variations linked to cholesterol synthesis, absorption, and transport can impact the effectiveness of statin therapy alongside environmental influences and patient adherence. Genomic studies identified genes associated with lipid metabolisms, such as apolipoprotein E (APOE), apolipoprotein B (APOB), cholesteryl ester transfer protein (CETP), LDL receptor (LDLR), cholesterol transport, such as ABCG5/8, ABCG2, ABCB1/MDR1, kinesin-like protein 6 (KIF6), solute carrier organic anion transporter 1B1 (SLCO1B1), calmin (CLMN), and CYP450 metabolizing enzymes family [47].

The utilization of simvastatin, a prominent HMGCR inhibitor, has faced a decline attributed to its link to myopathy risk in about 28% of patients [48]. This risk exhibits a dose-dependent pattern, prompting regulatory measures by the FDA to restrict the maximum allowable dose [49]. GWAS suggest a link between SLCO1B1 genetic variations, including rs4149056 (c388A-c521C), rs2306283 (388A>G), and noncoding rs4363657 (c.1498-1331T>C), and statin-induced myopathy, including simvastatin and atorvastatin. These polymorphisms reduce the function of the organic anion transporting polypeptides B1 (OATP1B1) found on hepatocytes’ sinusoidal membrane, leading to increased circulating levels of simvastatin active form. GWAS revealed a threefold elevated risk associated with each C genotype, particularly in homozygous individuals. Carriers of SLCO1B1*5 allele have a 4- to 5-fold higher risk of severe creatine kinase-positive statin-induced myopathy and a 2- to 3-fold higher risk of creatine kinase-negative myopathy [50].

Variations in the CYP3A4 gene have been connected to different responses to statin treatment, including simvastatin, atorvastatin, and lovastatin. Individuals carrying the homozygous/heterozygous CYP3A4*22 (rs35599367) or homozygous CYP3A5*3 (rs776746) polymorphisms, along with CYP3A4*4 (rs55951658) haplotype on statin therapy exhibit an escalated response that decreased total and LDL cholesterol levels [51, 52]. Conversely, those with the missense CYP3A4*3 (rs4986910, M445T), CYP3A4 promoter polymorphism (rs2740574, A290G), or CYP3A4*1G haplotype (rs2242480) might not achieve the anticipated lipid-lowering effects from statins [53, 54, 55]. One GWAS on the plasma level of atorvastatin identified associations with polymorphisms near CYP3A7 (rs45446698) and UDP Glucuronosyltransferase-1A (UGT1A, rs887829) [56]. Studies suggest that genetic variability in CYP2C9*3 (rs1057910) is connected to elevated levels of fluvastatin [57].

The ABCB1 genetic polymorphisms influence the efficacy of simvastatin, with less common variants (rs1128503, 1236T; rs2032582, 2677 non-G; and rs1045642, 3435T) occurring in patients who experience muscle-related side effects [58]. The pharmacokinetics of atorvastatin and rosuvastatin are affected through variants in the ABCG2 gene, which encodes breast cancer resistance protein (BCRP) transporter. Individuals with the CC genotype at rs2231142 experience more significant reductions in LDL cholesterol levels compared to those with AA genotypes [59].

In some studies, kinesin-like protein 6 (KIF6), involved in intracellular transport, has been linked to coronary artery disease. These studies suggest potential statin benefits for carriers of the rs20455 (Trp719Arg) missense polymorphism in the KIF6 gene [60]. However, a meta-analysis of 19 studies discovered an inconsistent link between this polymorphism and nonfatal coronary artery disease [61]. The CLMN polymorphism, rs8014194, explains only 1% of statin response variability, with carriers experiencing notably greater reductions in total cholesterol. Initial GWAS suggest that the APOE E2 (rs7412) (526C>T) and E4 (rs429358, 388T>C) alleles might significantly reduce the LDL levels and lower instances of nonfatal myocardial infarction and mortality when treated with lipid-lowering therapy [62]. However, support for these associations varies across all studies [63].

Pharmacogenetic research has focused on the CETP gene, particularly examining the TaqIB variant, rs708272. Individuals with a B1B1 genotype on statin treatment experience slower coronary artery disease progression than B2B2 carriers. While some studies, including Boekholdt et al.’s meta-analysis, find no connection between the TaqIB polymorphism and pravastatin treatment [64], the Regression Growth Evaluation Statin Study (REGRESS) revealed potential pharmacogenetic interactions. This study identified higher 10-year mortality in male statin-treated patients with the B2 allele, compared to those with the B1B1 genotype (Table 1) [65]. In the future, there is an expectation of a more comprehensive understanding of statins’ function and the individual variations in response.

Consequently, clinical guidelines suggest adjusting simvastatin and atorvastatin dosages according to the SLCO1B1 genotype, although the significance has diminished due to the availability of safer statins, such as rosuvastatin and or pravastatin [66]. The FDA has eliminated the requirement for further confirmatory testing of SLCO1B1-simvastatin genotyping [67]. CPIC guidelines now cover rosuvastatin dosing based on ABCG2 and SLCO1B1 genotypes and fluvastatin dosing based on CYP2C9 genotype [66]. This expansion to high-intensity statins, including atorvastatin and rosuvastatin, is expected to enhance the use of pharmacogenetic-guided approaches for selecting and dosing statins in clinical practice.

2.4 Beta-blockers

Beta-blockers function through competitive antagonism of endogenous catecholamines at the beta-1 and beta-2 adrenergic receptors, encoded by ADBR1 and ADBR2, in the heart and blood vessels. These medications are commonly prescribed to reduce heart rate, lower blood pressure, and enhance survival and left ventricular ejection fraction following an MI, including atenolol, bisoprolol, carvedilol, metoprolol, nebivolol, propranolol. However, the response to beta-blockers can vary significantly among individuals due to genetic factors influencing the pharmacokinetics (CYP2D6) and the pharmacodynamics (ADBR1, ADBR2, and GRK5).

Genetic variants of ADBR1, commonly rs1801252 (Ser49Gly) and rs1801253 (Arg389Gly), have been linked to impaired down-regulation and altered signal transduction in vitro [68, 69]. Associations of Gly49 are linked to more significant reductions in the end diastolic diameter than the Ser49 genotype. Moreover, Ser49 carriers have higher heart rates and increased mortality than Gly49 carriers on beta-blocker treatment [70]. Clinical research indicates that individuals with homozygous Arg389 genotype tend to respond more positively to beta-blockers, enhancements in the left ventricular ejection fraction (LVEF), and overall risk reduction of hospitalizations and mortality compared to Gly389 [71, 72]. However, this correlation and reports of improved beta-blocker response on blood pressure and heart rate demonstrated inconsistency [73, 74].

Prevalent polymorphisms in the ADBR2, mainly rs1042713 (Arg16Gly), rs1042714 (Gln27Gly), and rs1800888 (Thr164Ile), have shown increased adenylyl cyclase activity, leading to enhanced agonist-induced downregulation of ADBR2 [75]. However, associations of these variants with cardiovascular outcomes are inconsistent. Most studies observed no link between these genetic variants and cardiovascular improvement [76], but smaller studies suggested the Glu27 allele might be associated with higher LVEF than the Gln27 allele when using beta-blockers [77]. A prevalent four amino-acid deletion (Del322-325) reduces alpha(2C)-AR (ADRA2C) activity in cells and associates with adverse heart failure outcomes [78], though it did not affect the beta-blocker Evaluation of Survival Trial (BEST) with bucindolol [72].

The CYP2D6 enzyme predominantly metabolizes carvedilol, metoprolol, nebivolol, propranolol, and timolol. CYP2D6 is unnecessary for metabolizing other beta-blockers, like atenolol, bisoprolol, and nadolol. Common genetic variants of CYP2D6 lead to a range of phenotypes, varying from increased enzyme function due to gene duplication to complete loss of function. Such as homozygous rs1135840 and rs1065852, caused by gene deletion or splicing defects [79]. Approximately, 5–10% of the population carries two or more CYP2D6 alleles, such as the *4 allele, with reduced activity. This raises the circulating levels of beta-blockers, leading to substantial blood pressure and heart rate reduction and a greater risk of adverse drug reactions [80]. Although CYP2D6 variations seem to affect heart rate response to beta-blockers, their significant influence on blood pressure response and cardiovascular risk reduction remains uncertain [81].

The G protein-coupled receptor kinase 5, encoded by GRK5, is an intracellular component that attenuates signaling from beta-adrenergic receptors. The rs2230345 (Gln41Leu) variant heightens GRK5 activity, mimicking the impact of a beta-blocker [82]. Those with the 41Leu variant show improved heart failure and hypertension outcomes, potentially reducing beta-blockers effectiveness [83]. This could favor alternative medications depending on the medical condition. Nevertheless, further research is needed to establish the clinical implications of these variants for guiding beta-blocker therapy.

Considering the established pharmacogenomic interactions of beta-blockers and ADBR1 polymorphisms, utilizing multiple risk alleles could enhance the strategic management of this therapy [84]. Genetic variations in CYP2D6 might impact beta-blocker pharmacokinetics, but application in prescribing is limited. The FDA-approved metoprolol label downplays the impact of CYP2D6-dependent metabolism on safety but advises caution with potent CYP2D6 inhibitors, such as quinidine, fluoxetine, paroxetine, and propafenone [85]. The DPWG guidance suggests tailored dosing, including gradual titration for intermediate and poor metabolizers and considering alternatives, like bisoprolol, for ultra-rapid metabolizers as needed [9].

2.5 Direct-acting vasodilators

Direct-acting vasodilators decrease blood pressure by relaxing vascular smooth muscle, including hydralazine, minoxidil, and sodium nitroprusside. Hydralazine is recommended for secondary hypertension treatment and primarily undergoes hepatic metabolism by N-acetyltransferase type 2 (NAT2). Genetic variants in NAT2 determine acetylation rates, with NAT2*4 indicating rapid acetylation, and the common *5 rs1801280 (341T>C), *6 rs1799930 (590G>A), *7 rs1799931 (857G>A), and the rare *14 rs1801279 (191G>A) suggesting slower rates [86]. Rapid acetylators have lower hydralazine exposure, affecting blood pressure response. Slow acetylators might experience more potent antihypertensive effects due to increased drug exposure [87], potentially elevating the risk of rare adverse reactions, like lupus-like symptoms [88]. Nonetheless, the evidence supporting NAT2 genotyping for predicting hydralazine’s safety and efficacy remains inconclusive.

2.6 Renin-angiotensin system inhibitors

Renin-angiotensin system (RAS), including ACEIs (such as captopril, enalapril, and lisinopril) and ARBs (such as candesartan, losartan, and olmesartan) serves as a cornerstone in the management of CVD, encompassing hypertension, ACS, heart failure, and nephropathy. Polymorphisms in renin-angiotensin-aldosterone-related genes, particularly ACE, angiotensinogen (AGT), and angiotensin-II receptors I and II (AGTR1 and AGTR2), can potentially impact the ACEIs and ARBs mechanisms. Individuals with DD homozygosity of the ACE insertion/deletion rs4646994 (287-Alu) variant exhibit elevated ACE activity in both plasma and tissues, which is associated with higher 10-year mortality rates [89]. Around 10% of patients on ACEIs developed a dry cough, likely caused by an accumulation of bradykinin during therapy. Studies have linked this cough to the ACE insertion/deletion variant and a specific polymorphism in the bradykinin B2 (B2R) gene’s promoter region [90].

The AGT rs699 (Met235Thr) allele carriers tend to have elevated angiotensin levels, which may be connected to increased blood pressure [91]. The AGTR1 rs5186 (A1166C) polymorphism has been studied for its beneficial influence on ARB response [92]. A GWAS identified two moderately associated variants in the protein kinase C theta (PRKCQ, rs500766) and ETS transcription 6 (ETV6, rs2724635), genes involved in immune regulation related to ACEI-induced angioedema [93]. Current evidence lacks definitive pharmacogenomic associations between ACE, AGT, or AGT1R polymorphisms and the effectiveness and safety of ACEIs or ARBs. The inconsistent results and study limitations undermine the reliability of these findings, warranting further research to improve precision medicine guidance.

2.7 Diuretics

Thiazide diuretics (such as hydrochlorothiazide, chlorthalidone, and indapamide) play a crucial role in managing hypertension, while loop diuretics (such as bumetanide, ethacrynic acid, and furosemide) and aldosterone antagonist (like spironolactone) are the preferred choice for addressing fluid retention in heart failure.

Research suggests that individuals carrying the rs4961 (Gly460>Trp) variant in the α-adducin gene (ADD1) exhibit more favorable blood pressure responses to thiazide, loop, and spironolactone diuretics, revealing an association with salt sensitivity [94]. Carriers of the Trp460 variant experience a more pronounced protective effect, approximately 38%, against heart attacks (MI) and strokes compared to individuals with the Gly460 genotype [95]. The NPPA rs5065 (T2238C) variants, encode atrial natriuretic peptide, showed that C allele carriers had lower CVD risk and more significant blood pressure reductions with chlorthalidone than amlodipine. However, those with the TT genotype had a higher risk of heart attack, stroke, and all-cause mortality with chlorthalidone [96].

Furthermore, genetic variations in the NEDD4L (rs4149601, rs292449, and rs75982813, encodes neural precursor cell expressed developmentally downregulated 4-like enzyme), PRKCA (rs16960228, encodes Protein kinase C alpha), and YEATS4 (rs7297610, encodes YEATS Domain Containing-4) consistently show a significant link to cardiovascular outcomes with thiazide diuretics (Table 1) [97, 98, 99]. Despite their potential, these gene variations have not yet impacted diuretic prescriptions due to conflicting research. Further studies may lead to personalized diuretic therapies for hypertension and related conditions.

2.8 Antiarrhythmics

Antiarrhythmic drugs block the potassium current channel (IKr) and prolong QT intervals, increasing the drug-induced Torsade de Pointes (DITdP) malignant arrhythmia risk, including amiodarone, flecainide, procainamide, and sotalol. Some common gene variants have been linked to DITdP, constituting around 10% of cases known as congenital long QT syndrome (cLQTS) incomplete penetrance. The rs1805128 (D85N) polymorphism in potassium voltage-gated channel E1 (KCNE1), encoding Ikr subunit, and rs7626962 (S1103Y) sodium voltage-gated Channel Alpha 5 (SCN5A), encoding cardiac sodium channel, exhibit a strong connection to DITdP development [100, 101]. A significant SNP, rs10919035, in nitric oxide synthase 1 adaptor protein (NOS1AP), is notably prevalent in amiodarone related TdP cases and associated with ventricular arrhythmia and QT prolongation [102].

Prevalent polymorphisms at the 4q25 locus near the PITX2 gene, rs10033464, have been linked to an increased risk of atrial fibrillation (AF) and reduced responsiveness to certain antiarrhythmic drugs (Table 1) [103]. Additionally, a genetic variant rs1801253 in ADRB1 (Arg389Gly) is associated with better outcomes with rhythm control strategies for managing AF [104]. CYP2D6 and CYP3A4 metabolize some antiarrhythmic drugs as beta-blockers, such as flecainide, quinidine, and propafenone. Limited evidence suggests that genetic variations in CYP2D6 and CYP3A4 may impact their pharmacokinetics and affect the QTc interval, as indicated by their classification in PharmGKB as level 2A and in CPIC level as B-C [105, 106].

Clinical studies use a QT alert system to monitor high-risk DITdP patients. A polygenic risk score of common variants shows potential for enhancing personalized prevention and guiding AF treatment based on an individual’s genotype [107]. The FDA label for procainamide highlights response differences based on acetylation speed by NAT2 but does not mention any specific genotype for acetylation status determination. The FDA recommends caution with quinidine and propafenone in individuals with CYP2D6 and CYP3A4 inhibitions due to high adverse effects risk, like pro-arrhythmia. Although there are no specific CPIC guidelines for these drugs, the DPWG recommends reducing the standard doses by 50% and 30% in CYP2D6 poor metabolizers (Table 1) [37].

Advertisement

3. Future perspectives in personalized cardiovascular therapy

Advancements in pharmacogenomics offer significant potential for personalized cardiovascular therapy, leveraging an individual’s genetic profile. High-throughput genotyping and next-generation sequencing technologies enhance our understanding of how genetics influence responses to cardiovascular drugs [108]. Integrating pharmacogenomic data into electronic health records enhances clinical decision-making, promising safer and more cost-effective medication regimens. Notably, genetic markers identified through GWAS, and multi-omics analyses enable personalized selection of antiplatelets, statins, and warfarin dosages, reducing the occurrence of adverse cardiovascular events [9, 17, 22, 33, 66].

Following the validation of identified polymorphisms, the transformative genetic approach revolutionizes cardiovascular therapy, including implementing polygenic risk scores. However, cardiovascular pharmacogenomics faces challenges due to patient variability and limited study sizes. These challenges encompass genetic data diversity limitations, difficulties in clinical integration, technical and sociopolitical constraints, and the complexity of genetic interactions. Addressing these issues necessitates collaborative efforts, supported by artificial intelligence and machine learning, to uncover polygenic predictors and fully realize the potential of precision medicine in complex CVDs with multiple drug regimens, like heart failure [109]. While logistical challenges persist in obtaining early genotype results, the prospect of pre-emptive or point-of-care testing offers potential solutions.

Advertisement

4. Conclusion

Substantial evidence underscores genetic associations with efficacy and tolerance of cardiovascular therapies, including antiplatelets, anticoagulants, lipid-lowering agents, and, to moderate extent, beta-blockers. Pharmacogenetic testing has become a standard practice, enhancing cardiovascular treatment outcomes. However, further research is necessary to guide genotype-based drug selection and validate genetic variation’s effect on drug responses and safety among different populations. Ultimately, precision medicine aims to prove its superiority of genotype-based personalized interventions over current standard care and improve overall health outcomes.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Roth GA et al. Global burden of cardiovascular diseases and risk factors, 1990-2019: Update from the GBD 2019 study. Journal of the American College of Cardiology. 2020;76(25):2982-3021
  2. 2. Matthaei J et al. Heritability of metoprolol and torsemide pharmacokinetics. Clinical Pharmacology and Therapeutics. 2015;98(6):611-621
  3. 3. Watkins WS et al. De novo and recessive forms of congenital heart disease have distinct genetic and phenotypic landscapes. Nature Communications. 2019;10(1):4722
  4. 4. Roden DM et al. Opportunities and challenges in cardiovascular pharmacogenomics: From discovery to implementation. Circulation Research. 2018;122(9):1176-1190
  5. 5. Khera AV et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nature Genetics. 2018;50(9):1219-1224
  6. 6. Reel PS et al. Machine learning for classification of hypertension subtypes using multi-omics: A multi-Centre, retrospective, data-driven study. eBioMedicine. 2022;84:104276
  7. 7. Mensah GA et al. Emerging concepts in precision medicine and cardiovascular diseases in racial and ethnic minority populations. Circulation Research. 2019;125(1):7-13
  8. 8. Relling MV et al. The clinical pharmacogenetics implementation consortium: 10 years later. Clinical Pharmacology and Therapeutics. 2020;107(1):171-175
  9. 9. Lee CR et al. Clinical pharmacogenetics implementation consortium guideline for CYP2C19 genotype and clopidogrel therapy: 2022 update. Clinical Pharmacology and Therapeutics. 2022;112(5):959-967
  10. 10. Gong L, Whirl-Carrillo M, Klein TE. PharmGKB, an integrated resource of pharmacogenomic knowledge. Current Protocols. 2021;1(8):e226
  11. 11. Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC, Becker K, et al. Correction to: Guidelines for the early management of patients with acute ischemic stroke: 2019 update to the 2018 guidelines for the early management of acute ischemic stroke: A guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2019;50(12):e440-e441
  12. 12. Magavern EF, Kaski JC, Turner RM, Drexel H, Janmohamed A, Scourfield A, et al. Corrigendum to: The role of pharmacogenomics in contemporary cardiovascular therapy: A position statement from the European Society of Cardiology Working Group on Cardiovascular Pharmacotherapy. European Heart Journal - Cardiovascular Pharmacotherapy. 2022;9(1):116
  13. 13. Liu Q et al. Model based on single-nucleotide polymorphism to discriminate aspirin resistance patients. Stroke and Vascular Neurology. 2023;16:svn-2022-002228
  14. 14. Li Q et al. Frequency of genetic polymorphisms of COX1, GPIIIa and P2Y1 in a Chinese population and association with attenuated response to aspirin. Pharmacogenomics. 2007;8(6):577-586
  15. 15. Yi X et al. Platelet response to aspirin in Chinese stroke patients is independent of genetic polymorphisms of COX-1 C50T and COX-2 G765C. Journal of Atherosclerosis and Thrombosis. 2013;20(1):65-72
  16. 16. Wang H et al. Association of GPIa and COX-2 gene polymorphism with aspirin resistance. Journal of Clinical Laboratory Analysis. 2018;32(4):e22331
  17. 17. Verma SS et al. Genomewide association study of platelet reactivity and cardiovascular response in patients treated with clopidogrel: A study by the international clopidogrel pharmacogenomics consortium. Clinical Pharmacology and Therapeutics. 2020;108(5):1067-1077
  18. 18. Ionova Y et al. CYP2C19 allele frequencies in over 2.2 million direct-to-consumer genetics research participants and the potential implication for prescriptions in a large health system. Clinical and Translational Science. 2020;13(6):1298-1306
  19. 19. Mega JL et al. Cytochrome p-450 polymorphisms and response to clopidogrel. The New England Journal of Medicine. 2009;360(4):354-362
  20. 20. Yadav AK et al. Substantiation of a clopidogrel metabolism-associated gene (CYP2C19) variation among healthy individuals. Indian Heart Journal. 2023;75(5):343-346
  21. 21. Claassens DMF et al. A genotype-guided strategy for oral P2Y(12) inhibitors in primary PCI. The New England Journal of Medicine. 2019;381(17):1621-1631
  22. 22. Her AY et al. Platelet function and genotype after DES implantation in east Asian patients: Rationale and characteristics of the PTRG-DES consortium. Yonsei Medical Journal. 2022;63(5):413-421
  23. 23. Liedes H et al. CYP3A4*22 may increase bleeding risk in ticagrelor users. Basic & Clinical Pharmacology & Toxicology. 2023;133(2):202-207
  24. 24. Varenhorst C et al. Effect of genetic variations on ticagrelor plasma levels and clinical outcomes. European Heart Journal. 2015;36(29):1901-1912
  25. 25. Asiimwe IG et al. A genome-wide association study of plasma concentrations of warfarin enantiomers and metabolites in sub-Saharan black-African patients. Frontiers in Pharmacology. 2022;13:967082
  26. 26. Parra EJ et al. Genome-wide association study of warfarin maintenance dose in a Brazilian sample. Pharmacogenomics. 2015;16(11):1253-1263
  27. 27. Pokorney SD et al. Patients' time in therapeutic range on warfarin among US patients with atrial fibrillation: Results from ORBIT-AF registry. American Heart Journal. 2015;170(1):141-148, 148 e1
  28. 28. Wadelius M et al. Association of warfarin dose with genes involved in its action and metabolism. Human Genetics. 2007;121(1):23-34
  29. 29. Singh O et al. Influence of CYP4F2 rs2108622 (V433M) on warfarin dose requirement in Asian patients. Drug Metabolism and Pharmacokinetics. 2011;26(2):130-136
  30. 30. Scott SA et al. Warfarin pharmacogenetics: CYP2C9 and VKORC1 genotypes predict different sensitivity and resistance frequencies in the Ashkenazi and Sephardi Jewish populations. American Journal of Human Genetics. 2008;82(2):495-500
  31. 31. Allabi AC, Gala JL, Horsmans Y. CYP2C9, CYP2C19, ABCB1 (MDR1) genetic polymorphisms and phenytoin metabolism in a Black Beninese population. Pharmacogenetics and Genomics. 2005;15(11):779-786
  32. 32. Kimmel SE et al. A pharmacogenetic versus a clinical algorithm for warfarin dosing. The New England Journal of Medicine. 2013;369(24):2283-2293
  33. 33. Johnson JA et al. Clinical pharmacogenetics implementation consortium (CPIC) guideline for pharmacogenetics-guided warfarin dosing: 2017 update. Clinical Pharmacology and Therapeutics. 2017;102(3):397-404
  34. 34. Ragia G et al. A novel acenocoumarol pharmacogenomic dosing algorithm for the Greek population of EU-PACT trial. Pharmacogenomics. 2017;18(1):23-34
  35. 35. Ragia G, Manolopoulos VG. Pharmacogenomics of anticoagulation therapy: The last 10 years. Pharmacogenomics. 2019;20(16):1113-1117
  36. 36. Verde Z et al. A novel, single algorithm approach to predict acenocoumarol dose based on CYP2C9 and VKORC1 allele variants. PLoS One. 2010;5(6):e11210
  37. 37. Yoon DY et al. Pharmacogenomic information from CPIC and DPWG guidelines and its application on drug labels. Translational and Clinical Pharmacology. 2020;28(4):189-198
  38. 38. Karnes JH et al. Influence of human leukocyte antigen (HLA) alleles and killer cell immunoglobulin-like receptors (KIR) types on heparin-induced thrombocytopenia (HIT). Pharmacotherapy. 2017;37(9):1164-1171
  39. 39. Rollin J et al. Increased risk of thrombosis in FcgammaRIIA 131RR patients with HIT due to defective control of platelet activation by plasma IgG2. Blood. 2015;125(15):2397-2404
  40. 40. Harris K, Nguyen P, Van Cott EM. Platelet PlA2 polymorphism and the risk for thrombosis in heparin-induced thrombocytopenia. American Journal of Clinical Pathology. 2008;129(2):282-286
  41. 41. Rollin J et al. Polymorphisms of protein tyrosine phosphatase CD148 influence FcgammaRIIA-dependent platelet activation and the risk of heparin-induced thrombocytopenia. Blood. 2012;120(6):1309-1316
  42. 42. Pouplard C et al. Interleukin-10 promoter microsatellite polymorphisms influence the immune response to heparin and the risk of heparin-induced thrombocytopenia. Thrombosis Research. 2012;129(4):465-469
  43. 43. Karnes JH et al. A genome-wide association study of heparin-induced thrombocytopenia using an electronic medical record. Thrombosis and Haemostasis. 2015;113(4):772-781
  44. 44. Sychev DA et al. The impact of ABCB1 (rs1045642 and rs4148738) and CES1 (rs2244613) gene polymorphisms on dabigatran equilibrium peak concentration in patients after total knee arthroplasty. Pharmacogenomics and Personalized Medicine. 2018;11:127-137
  45. 45. Kryukov AV et al. Influence of ABCB1 and CYP3A5 gene polymorphisms on pharmacokinetics of apixaban in patients with atrial fibrillation and acute stroke. Pharmacogenomics and Personalized Medicine. 2018;11:43-49
  46. 46. XARELTO (rivaroxaban). 2023. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/202439s001lbl.pdf
  47. 47. Hindi NN, Alenbawi J, Nemer G. Pharmacogenomics variability of lipid-lowering therapies in familial hypercholesterolemia. Journal of Personalized Medicine. 2021;11(9):877
  48. 48. Abed W et al. Statin induced myopathy among patients attending the National Center for Diabetes, Endocrinology, & Genetics. Annals of Medicine and Surgery. 2022;74:103304
  49. 49. FDA Drug Safety Communication: Revised Dose Limitation for Zocor (Simvastatin) When Taken with Amiodarone. 2011. Available from: https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-revised-dose-limitation-zocor-simvastatin-when-taken-amiodarone
  50. 50. Mykkanen AJH et al. Genomewide association study of simvastatin pharmacokinetics. Clinical Pharmacology and Therapeutics. 2022;112(3):676-686
  51. 51. Elalem EG et al. Association of cytochromes P450 3A4*22 and 3A5*3 genotypes and polymorphism with response to simvastatin in hypercholesterolemia patients. PLoS One. 2022;17(7):e0260824
  52. 52. Wang A et al. Ile118Val genetic polymorphism of CYP3A4 and its effects on lipid-lowering efficacy of simvastatin in Chinese hyperlipidemic patients. European Journal of Clinical Pharmacology. 2005;60(12):843-848
  53. 53. Thompson JF et al. An association study of 43 SNPs in 16 candidate genes with atorvastatin response. The Pharmacogenomics Journal. 2005;5(6):352-358
  54. 54. Kajinami K et al. CYP3A4 genotypes and plasma lipoprotein levels before and after treatment with atorvastatin in primary hypercholesterolemia. The American Journal of Cardiology. 2004;93(1):104-107
  55. 55. Peng C et al. Polymorphisms in CYP450 genes and the therapeutic effect of atorvastatin on ischemic stroke: A retrospective cohort study in Chinese population. Clinical Therapeutics. 2018;40(3):469-477 e2
  56. 56. Turner RM et al. A genome-wide association study of circulating levels of atorvastatin and its major metabolites. Clinical Pharmacology and Therapeutics. 2020;108(2):287-297
  57. 57. Buzkova H et al. Lipid-lowering effect of fluvastatin in relation to cytochrome P450 2C9 variant alleles frequently distributed in the Czech population. Medical Science Monitor. 2012;18(8):CR512-517
  58. 58. Fiegenbaum M et al. The role of common variants of ABCB1, CYP3A4, and CYP3A5 genes in lipid-lowering efficacy and safety of simvastatin treatment. Clinical Pharmacology and Therapeutics. 2005;78(5):551-558
  59. 59. Keskitalo JE et al. ABCG2 polymorphism markedly affects the pharmacokinetics of atorvastatin and rosuvastatin. Clinical Pharmacology and Therapeutics. 2009;86(2):197-203
  60. 60. Iakoubova OA et al. KIF6 Trp719Arg polymorphism and the effect of statin therapy in elderly patients: Results from the PROSPER study. European Journal of Cardiovascular Prevention and Rehabilitation. 2010;17(4):455-461
  61. 61. Assimes TL et al. Lack of association between the Trp719Arg polymorphism in kinesin-like protein-6 and coronary artery disease in 19 case-control studies. Journal of the American College of Cardiology. 2010;56(19):1552-1563
  62. 62. Barber MJ et al. Genome-wide association of lipid-lowering response to statins in combined study populations. PLoS One. 2010;5(3):e9763
  63. 63. Zintzaras E et al. APOE gene polymorphisms and response to statin therapy. The Pharmacogenomics Journal. 2009;9(4):248-257
  64. 64. Boekholdt SM et al. Cholesteryl ester transfer protein TaqIB variant, high-density lipoprotein cholesterol levels, cardiovascular risk, and efficacy of pravastatin treatment: Individual patient meta-analysis of 13,677 subjects. Circulation. 2005;111(3):278-287
  65. 65. Regieli JJ et al. CETP genotype predicts increased mortality in statin-treated men with proven cardiovascular disease: An adverse pharmacogenetic interaction. European Heart Journal. 2008;29(22):2792-2799
  66. 66. Cooper-DeHoff RM et al. The clinical pharmacogenetics implementation consortium guideline for SLCO1B1, ABCG2, and CYP2C9 genotypes and statin-associated musculoskeletal symptoms. Clinical Pharmacology and Therapeutics. 2022;111(5):1007-1021
  67. 67. 23andMe Releases New FDA-Cleared Genetic Report on Simvastatin, a Commonly Prescribed Statin. 2023. Available from: https://investors.23andme.com/news-releases/news-release-details/23andme-releases-new-fda-cleared-genetic-report-simvastatin
  68. 68. Brodde OE. Beta1- and beta2-adrenoceptor polymorphisms and cardiovascular diseases. Fundamental & Clinical Pharmacology. 2008;22(2):107-125
  69. 69. Magnusson Y et al. Ser49Gly of beta1-adrenergic receptor is associated with effective beta-blocker dose in dilated cardiomyopathy. Clinical Pharmacology and Therapeutics. 2005;78(3):221-231
  70. 70. Rathz DA et al. Amino acid 49 polymorphisms of the human beta1-adrenergic receptor affect agonist-promoted trafficking. Journal of Cardiovascular Pharmacology. 2002;39(2):155-160
  71. 71. Chen L et al. Arg389Gly-beta1-adrenergic receptors determine improvement in left ventricular systolic function in nonischemic cardiomyopathy patients with heart failure after chronic treatment with carvedilol. Pharmacogenetics and Genomics. 2007;17(11):941-949
  72. 72. Bristow MR et al. An alpha2C-adrenergic receptor polymorphism alters the norepinephrine-lowering effects and therapeutic response of the beta-blocker bucindolol in chronic heart failure. Circulation. Heart Failure. 2010;3(1):21-28
  73. 73. Ma ST et al. Association between beta1 adrenergic receptor gene Arg389Gly polymorphism and risk of heart failure: A meta-analysis. Genetics and Molecular Research. 2015;14(2):5922-5929
  74. 74. Li YJ et al. Polymorphisms of Arg389Gly of beta1-adrenergic receptor gene and essential hypertension risk: A meta analysis. Zhonghua Yi Xue Za Zhi. 2011;91(44):3115-3119
  75. 75. Al-Balushi K et al. Frequencies of the Arg16Gly, Gln27Glu and Thr164Ile adrenoceptor beta2 polymorphisms among Omanis. Sultan Qaboos University Medical Journal. 2015;15(4):e486-e490
  76. 76. de Groote P et al. Association between beta-1 and beta-2 adrenergic receptor gene polymorphisms and the response to beta-blockade in patients with stable congestive heart failure. Pharmacogenetics and Genomics. 2005;15(3):137-142
  77. 77. Liu J et al. beta1-adrenergic receptor polymorphisms influence the response to metoprolol monotherapy in patients with essential hypertension. Clinical Pharmacology and Therapeutics. 2006;80(1):23-32
  78. 78. Kardia SL et al. Multiple interactions between the alpha 2C- and beta1-adrenergic receptors influence heart failure survival. BMC Medical Genetics. 2008;9:93
  79. 79. Zisaki A, Miskovic L, Hatzimanikatis V. Antihypertensive drugs metabolism: An update to pharmacokinetic profiles and computational approaches. Current Pharmaceutical Design. 2015;21(6):806-822
  80. 80. Bijl MJ et al. Genetic variation in the CYP2D6 gene is associated with a lower heart rate and blood pressure in beta-blocker users. Clinical Pharmacology and Therapeutics. 2009;85(1):45-50
  81. 81. Luzum JA et al. CYP2D6 genetic variation and Beta-blocker maintenance dose in patients with heart failure. Pharmaceutical Research. 2017;34(8):1615-1625
  82. 82. Liggett SB et al. A GRK5 polymorphism that inhibits beta-adrenergic receptor signaling is protective in heart failure. Nature Medicine. 2008;14(5):510-517
  83. 83. Lobmeyer MT et al. Polymorphisms in genes coding for GRK2 and GRK5 and response differences in antihypertensive-treated patients. Pharmacogenetics and Genomics. 2011;21(1):42-49
  84. 84. Petersen M et al. Association of beta-adrenergic receptor polymorphisms and mortality in carvedilol-treated chronic heart-failure patients. British Journal of Clinical Pharmacology. 2011;71(4):556-565
  85. 85. LOPRESSOR (metoprolol tartrate). 2020. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/018704s027,028lbl.pdf
  86. 86. Spinasse LB et al. Different phenotypes of the NAT2 gene influences hydralazine antihypertensive response in patients with resistant hypertension. Pharmacogenomics. 2014;15(2):169-178
  87. 87. Han LW et al. Effect of N-acetyltransferase 2 genotype on the pharmacokinetics of hydralazine during pregnancy. Journal of Clinical Pharmacology. 2019;59(12):1678-1689
  88. 88. Schoonen WM et al. Do selected drugs increase the risk of lupus? A matched case-control study. British Journal of Clinical Pharmacology. 2010;70(4):588-596
  89. 89. Brugts JJ et al. Genetic determinants of treatment benefit of the angiotensin-converting enzyme-inhibitor perindopril in patients with stable coronary artery disease. European Heart Journal. 2010;31(15):1854-1864
  90. 90. Nishio K et al. Angiotensin-converting enzyme and bradykinin gene polymorphisms and cough: A meta-analysis. World Journal of Cardiology. 2011;3(10):329-336
  91. 91. Schelleman H et al. Angiotensinogen M235T polymorphism and the risk of myocardial infarction and stroke among hypertensive patients on ACE-inhibitors or beta-blockers. European Journal of Human Genetics. 2007;15(4):478-484
  92. 92. Liu Y et al. Association of AGTR1 A1166C and CYP2C9 *3 gene polymorphisms with the antihypertensive effect of valsartan. International Journal of Hypertension. 2022;2022:7677252
  93. 93. Pare G et al. Genetic variants associated with angiotensin-converting enzyme inhibitor-associated angioedema. Pharmacogenetics and Genomics. 2013;23(9):470-478
  94. 94. Wang R et al. Association between alpha-adducin gene polymorphism (Gly460Trp) and genetic predisposition to salt sensitivity: A meta-analysis. Journal of Applied Genetics. 2010;51(1):87-94
  95. 95. Psaty BM et al. Diuretic therapy, the alpha-adducin gene variant, and the risk of myocardial infarction or stroke in persons with treated hypertension. JAMA. 2002;287(13):1680-1689
  96. 96. Lynch AI et al. Pharmacogenetic association of the NPPA T2238C genetic variant with cardiovascular disease outcomes in patients with hypertension. JAMA. 2008;299(3):296-307
  97. 97. Turner ST et al. Genomic association analysis of common variants influencing antihypertensive response to hydrochlorothiazide. Hypertension. 2013;62(2):391-397
  98. 98. McDonough CW et al. Association of variants in NEDD4L with blood pressure response and adverse cardiovascular outcomes in hypertensive patients treated with thiazide diuretics. Journal of Hypertension. 2013;31(4):698-704
  99. 99. Duarte JD et al. Association of chromosome 12 locus with antihypertensive response to hydrochlorothiazide may involve differential YEATS4 expression. The Pharmacogenomics Journal. 2013;13(3):257-263
  100. 100. Kaab S et al. A large candidate gene survey identifies the KCNE1 D85N polymorphism as a possible modulator of drug-induced torsades de pointes. Circulation. Cardiovascular Genetics. 2012;5(1):91-99
  101. 101. Paulussen AD et al. Genetic variations of KCNQ1, KCNH2, SCN5A, KCNE1, and KCNE2 in drug-induced long QT syndrome patients. Journal of Molecular Medicine (Berlin, Germany). 2004;82(3):182-188
  102. 102. Jamshidi Y et al. Common variation in the NOS1AP gene is associated with drug-induced QT prolongation and ventricular arrhythmia. Journal of the American College of Cardiology. 2012;60(9):841-850
  103. 103. Parvez B et al. Common genetic polymorphism at 4q25 locus predicts atrial fibrillation recurrence after successful cardioversion. Heart Rhythm. 2013;10(6):849-855
  104. 104. Parvez B et al. A common beta1-adrenergic receptor polymorphism predicts favorable response to rate-control therapy in atrial fibrillation. Journal of the American College of Cardiology. 2012;59(1):49-56
  105. 105. Rouini MR, Afshar M. Effect of CYP2D6 polymorphisms on the pharmacokinetics of propafenone and its two main metabolites. Thérapie. 2017;72(3):373-382
  106. 106. Doki K et al. Serum flecainide S/R ratio reflects the CYP2D6 genotype and changes in CYP2D6 activity. Drug Metabolism and Pharmacokinetics. 2015;30(4):257-262
  107. 107. Strauss DG et al. Common genetic variant risk score is associated with drug-induced QT prolongation and torsade de pointes risk: A pilot study. Circulation. 2017;135(14):1300-1310
  108. 108. Infante T et al. Network medicine: A clinical approach for precision medicine and personalized therapy in coronary heart disease. Journal of Atherosclerosis and Thrombosis. 2020;27(4):279-302
  109. 109. Segar MW et al. Phenomapping of patients with heart failure with preserved ejection fraction using machine learning-based unsupervised cluster analysis. European Journal of Heart Failure. 2020;22(1):148-158

Written By

Georges Nemer and Nagham Nafiz Hendi

Submitted: 07 September 2023 Reviewed: 18 September 2023 Published: 14 October 2023