Back to Journals » The Application of Clinical Genetics » Volume 17

The Diversity of CYP2C19 Polymorphisms in the Thai Population: Implications for Precision Medicine

Authors Nakhonsri V, John S , Panumasmontol H, Jantorn M, Chanthot P, Hanpramukkun N, Meelarp S, Sukasem C, Tongsima S, Hasatsri S , Prawang A, Thaingtamtanha T , Vanwong N , Atasilp C, Chamnanphon M, Jinda P, Satapornpong P

Received 24 March 2024

Accepted for publication 20 June 2024

Published 2 July 2024 Volume 2024:17 Pages 95—105

DOI https://doi.org/10.2147/TACG.S463965

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Martin Maurer



Vorthunju Nakhonsri,1 Shobana John,2,3 Hathaichanok Panumasmontol,4,5 Manassanan Jantorn,4,5 Pongpipat Chanthot,4,5 Nuntachai Hanpramukkun,6 Supaporn Meelarp,7 Chonlaphat Sukasem,2,3 Sissades Tongsima,1 Sukhontha Hasatsri,4 Abhisit Prawang,8 Thanawat Thaingtamtanha,9,10 Natchaya Vanwong,11,12 Chalirmporn Atasilp,13 Monpat Chamnanphon,14 Pimonpan Jinda,2,3 Patompong Satapornpong4,5

1National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand; 2Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; 3Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi Hospital, Bangkok, Thailand; 4Division of General Pharmacy Practice, Department of Pharmaceutical Care, College of Pharmacy, Rangsit University, Pathum Thani, Thailand; 5Excellence Pharmacogenomics and Precision Medicine Centre, College of Pharmacy, Rangsit University, Pathum Thani, Thailand; 6Division of Pharmaceutical Technology, Department of Industrial Pharmacy, College of Pharmacy, Rangsit University, Pathum Thani, Thailand; 7Ounjai Medical Clinic, Bangsue, Bangkok, Thailand; 8Division of Pharmacy Practice, Department of Pharmaceutical Care, College of Pharmacy, Rangsit University, Pathum Thani, Thailand; 9Department of Chemistry and Biology, University of Siegen, Siegen, Germany; 10Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada; 11Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand; 12Department of Clinical Chemistry, SYstems Neuroscience of Autism & PSychiatric Disorders (SYNAPS) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand; 13Chulabhorn International College of Medicine, Thammasat University, Pathumthani, Thailand; 14Department of Pathology, Faculty of Medicine, Srinakharinwirot University, Nakornnayok, Thailand

Correspondence: Patompong Satapornpong, Director of Excellence Pharmacogenomics and Precision Medicine Centre, College of Pharmacy, Rangsit University, Pathum Thani, Thailand, Tel +66- 2-791-6000 1420, Fax +66- 2-791-6000 1403, Email [email protected]

Introduction: CYP2C19 plays a major role in the metabolism of various drugs. The most common genetic variants were the CYP2C19*2 and *3 alleles (rs4244285 and rs4986893, non-functional variants). In previous studies, we found that genetic polymorphisms in CYP2C19 variants influenced the active metabolites of clopidogrel and caused major adverse cardiovascular and cerebrovascular effects. However, the distribution of CYP2C19 varies among ethnic groups and according to adverse drug reactions. This study aimed to investigate the frequency of CYP2C19 genetic polymorphisms in the Thai population and analyze the differences in the frequency of CYP2C19 genetic polymorphisms between Thai and other populations.
Methods: This study enrolled 211 unrelated healthy Thai individuals in total. We performed a real-time polymerase chain reaction to genotype CYP2C19*2 (681G > A) and CYP2C19*3 (636G > A).
Results: In the Thai population, the CYP2C19*1 allele was the most prevalent at 70.14%, while the CYP2C19*2 and *3 alleles were found at frequencies of 25.36% and 4.50%, respectively. Conversely, the CYP2C19*3 allele was not detected in Caucasian, Hispanic, African, Italian, Macedonian, Tanzanian, or North Indian populations. The phenotypic profile of this gene revealed that the frequency of intermediate metabolizers (IMs) is nearly equal to that of extensive metabolizers (EMs), at 42.65% and 48.82% respectively, with genotypes *1/*2 (36.02%) and *1/*3 (6.63%). Likewise, poor metabolizers (PMs) with genotypes *2/*2 (6.16%), *2/*3 (2.37%), and *3/*3 (< 1%) are more prevalent in our population as well.
Conclusion: The distribution of CYP2C19 genotype and phenotype influenced by non-functional alleles has potential as a pharmacogenomics biomarker for precision medicine and is dependent on an ethnic-specific genetic variation database.

Keywords: CYP2C19 gene, genetic diversity, Thai population, interethnic differences

Introduction

Cytochrome P450 (CYP) proteins form a superfamily of enzymes that are involved in the metabolism of drugs, fatty acids, steroids, and xenobiotics.1,2 Research has demonstrated that the metabolic activity of CYP enzymes is influenced by genetic polymorphisms.3,4 These polymorphisms in drug-metabolizing enzyme genes contribute to variations in pharmacological responses and the risk of adverse drug events among individuals and across different ethnic groups.5 The human CYP2C subfamily includes four members: CYP2C8, CYP2C9, CYP2C18, and CYP2C19.6

The CYP2C19 gene is located on chromosome 10 (10q24.1-q24.3) and is responsible for approximately 10% of drug metabolism in clinical practice.7,8 CYP2C19 plays a crucial role in metabolizing various therapeutic drugs, including clopidogrel, phenytoin, omeprazole, proguanil, diazepam, citalopram, imipramine, amitriptyline, and clomipramine.9 The most common genetic variant of CYP2C19 is the CYP2C19*2 allele (rs4244285, c. G681A), a single base pair mutation in exon 5 that results in a splicing defect, impairing enzyme function.10 Another significant variant, CYP2C19*3 (rs4986893), is a G636A mutation in exon 4, producing a premature stop codon.11 CYP2C19 genotypes and phenotypes are categorized as follows: *1/*1 (extensive metabolizers, EMs, two functional alleles); *1/*2 and *1/*3 (intermediate metabolizers, IM, one null allele and one functional allele); and *2/*2, *2/*3, and *3/*3 (poor metabolizers, PM, two non-functional alleles).12–14 Numerous studies have shown a correlation between CYP2C19 polymorphisms and enzyme activity in patients treated with relevant drugs.15–17

Clopidogrel is commonly used to treat myocardial infarction (MI), stroke, acute coronary syndrome (ACS), and atherosclerotic vascular disease.18 Notably, CYP2C19 plays a crucial role in converting clopidogrel into its active metabolite, clopi-H4.19 This active metabolite inhibits the adenosine diphosphate P2Y12 receptor, thereby reducing platelet activation. According to the Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for clopidogrel, alleles associated with the CYP2C19 poor metabolizers phenotype (*2/*2, *2/*3, and *3/*3) are significantly linked to lower levels of active clopidogrel metabolites and an increased risk of major adverse cardiovascular and cerebrovascular events.20 Furthermore, the CYP2C19 intermediate metabolizers (IMs) phenotype also reduces the effectiveness of clopidogrel.

The distribution of poor metabolizers (PMs) with genotypes *2/*2, *2/*3, and *3/*3 vary among different populations. The highest prevalence is observed in East Asians (12.97%) and Asians (8.15%), followed by African Americans (4.05%) and Europeans (2.38%). A similar pattern is seen with intermediate metabolizers (IMs), with East Asians having the highest frequency (45.92%), followed by Asians (40.80%), African-Americans (31.39%), and Europeans (26.10%).21 These genetic variations in CYP2C19 across different ethnicities are crucial for understanding individual differences in treatment effectiveness and safety, especially in the context of pharmacogenomics-based dosing guidelines. Consequently, this study aimed to investigate the prevalence of CYP2C19 genetic variations in the Thai population and compare them with those in other populations.

Materials and Methods

Thai Subjects

This cross-sectional study recruited 211 unrelated, healthy individuals of Thai ethnicity, with clinical data obtained from the Excellence Pharmacogenomics and Precision Medicine Centre, College of Pharmacy, Rangsit University, between November 2021 and February 2023 (Supplement 1). All participants were native Thais who were lived in the location of Thailand and had no history of adverse drug reactions (ADRs). We used self-identified race/ethnicity (SIRE) method to confirm the ethnicity, specifically three generations were confirmed as Thais. Thailand is centrally located in Mainland Southeast Asia, bordered by Myanmar to the west, Laos to the northeast, Cambodia to the east, and Malaysia to the south. The study received approval from the Ethics Review Board of Rangsit University (COA. No. RSUERB2021-104), and according to the Declaration of Helsinki with a written informed consent was obtained from all participants.

CYP2C19 Genotyping

Genomic DNA was extracted from EDTA whole blood using a Geneaid DNA Isolation Kit (Geneaid Biotech Ltd., New Taipei City, Taiwan). The quality and quantity of the genomic DNA were measured using an ultraviolet-visible spectrophotometer (Zhengzhou, Henan, China). Genotyping of CYP2C19*2 (681G > A, rs4244285, non-functional variant) and CYP2C19*3 (636G > A, rs4986893, non-functional variant) was performed using TaqMan assays (C__30634128_10 and C__27861809_10 respectively; ABI, Foster City, CA, USA). The PCR cycling conditions included 50 cycles of denaturation at 92°C for 15 seconds, and annealing and extension at 60°C for 1.30 minutes. CYP2C19 variants were identified using a QIAGEN QIAquant 96 real-time PCR system (Qiagen, Hilden, Germany).

CYP2C19 Phenotype

According to the Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline, CYP2C19*1/*1 is categorized as an extensive metabolizers (EMs) with two normal-function alleles. Intermediate metabolizers (IMs) have one normal and one non-functional allele (CYP2C19*1/*2 and CYP2C19*1/*3). Poor metabolizers (PMs) carry two non-functional alleles (CYP2C19*2/*2, CYP2C19*2/*3, and CYP2C19*3/*3).

Statistical Analysis

The frequencies of the two common CYP2C19 variants were analyzed using the Arlequin program version 3.1 for Hardy–Weinberg equilibrium testing. Allele and phenotype frequencies of CYP2C19 in the Thai population were compared with those in other populations using R Statistical Software (v4.1.2; R Core Team 2021). Odds ratios for all comparisons were illustrated in a forest plot using the Meta for R package (v4.2.0; Viechtbauer 2010). A significant difference was identified for each population compared with the Thai population if the p-value was less than 0.05.

Results

The Frequency of CYP2C19 Alleles in Thai Population

A total of 211 unrelated healthy Thai participants were included in this study, comprising fifty-five males (26.07%) and one hundred fifty-six females (73.93%). The mean age of the participants was 38.3 years, ranging from 19 to 75 years. We determined the allele frequency distribution of CYP2C19 in the Thai population (Table 1). The CYP2C19*1 allele was the most common, with a frequency of 70.14%. The frequencies of the CYP2C19*2 and CYP2C19*3 alleles were 25.36% (n = 107) and 4.50% (n = 19), respectively. Among the healthy Thai participants, the CYP2C19 genotypes were distributed as follows: 103 (48.82%) were extensive metabolizers (EMs, CYP2C19*1/*1), 76 (36.02%) were intermediate metabolizers (IMs, *1/*2 and *1/*3), and 14 (6.63%), 13 (6.16%), and 5 (2.37%) were poor metabolizers (PMs, *2/*2 and *2/*3). The frequency of the CYP2C19*3/*3 (PMs) genotype was less than 1.00% of the total Thai population. The allele frequencies of CYP2C19*2 and *3 were tested for Hardy–Weinberg equilibrium (p-value < 0.05), showing no significant deviation in the Thai population.

Table 1 Alleles and Genotyping Frequencies for CYP2C19 Genes in the Thai Population (n = 211)

Comparative of CYP2C19 Genotype and Phenotype Frequency Between the Thai and Other Populations

The frequencies of the CYP2C19*2 and *3 alleles were evaluated in Thai and various other populations, as shown in Tables 2 and 3 and Figure 1A and B. This study found that the CYP2C19*2 allele was particularly frequent. The highest frequencies of the CYP2C19*2 allele were observed in Indians (37.90%),30 Native Japanese (34.50%),33 and Bhutanese (30.14%),34 followed by North Indians (29.75%),29 Koreans (28.40%),33 Thais (25.36%), Han Chinese (24.67%),31 Vietnamese (20.50%),35 and Africans (20.20%)26 (Table 2 and Figure 1A). Conversely, the Bhutanese population had the highest observed frequency of the CYP2C19*3 allele (15.69%)34 (Table 3 and Figure 1B). High distributions of the CYP2C19*3 allele were also observed in Koreans (10.10%),33 Native Japanese (9.00%),32 Malaysians (6.50%),36 and Thais (4.50%), while the CYP2C19*3 allele was not found in Caucasian, Hispanic, African, Italian, Macedonian, Tanzanian, and North Indian populations.22–26,28,29

Table 2 The Allele Frequencies of CYP2C19*2 in Many Populations

Table 3 The Allele Frequencies of CYP2C19*3 in Many Populations

Figure 1 Forest plot displaying the odds ratios for the frequencies of CYP2C19*2 (A) and CYP2C19*3 (B) alleles across multiple populations in comparison to the Thai population allele frequency.

A comparison of CYP2C19*2 and *3 frequencies between the Thai population and other populations is presented in Tables 2 and 3 and Figure 1A and B. The distribution of CYP2C19 variations was similar among the Thai, Han Chinese, Vietnamese, and Venezuelan populations (p-value > 0.05). However, the CYP2C19*2 allele showed significant differences between the Thai population and others, such as Italian (p-value = 1.21×10−5), Macedonian (p-value = 0.0056), Caucasian (p-value = 0.0176), Hispanic (p = 6.04×10−5), Indian (p-value = 0.0298), and Malaysian populations (p-value = 0.0007). A strong significant difference was also observed for CYP2C19*3 frequencies when comparing Thai with Bhutanese and Hispanic populations (p-value = 8.94×10−6 and 1.58 x 10−5, respectively).

Phenotyping of CYP2C19*1/*1 (extensive metabolizers, EMs) was highly prevalent in several populations (Table 4 and Figures 2 and 3). The highest frequencies of CYP2C19*1/*1 were observed in the Italian population (79.44%) compared to the Thai population (p-value = 8.22×10−14).22 Intermediate metabolizers (CYP2C19 *1/*2 and *1/*3) showed the following phenotype distribution: Italian (18.89%), Macedonian (19.02%), Caucasian (18.52%), Hispanic (18.00%), African (19.19%), African-American (23.60%), and Malaysian (14.52%) populations.22–26,36 Significant differences in IMs prevalence were observed when compared with Thais (p-value < 0.05), with the highest frequency detected in the Bhutanese population (79.01%; OR = 5.05, 95% CI = 3.49–7.34, p-value = 7.83×10−20).34

Table 4 Phenotyping Frequencies of CYP2C19*2 and *3 Polymorphism Among Different Ethnicities Compared with Thai Population

Figure 2 Forest plot displaying the odds ratios for the frequencies of CYP2C19 intermediate metabolizers (IMs; *1/2 + *1/3) diplotypes (A) and CYP2C19 poor metabolizers (PMs; *2/*2 + *2/*3 + *3/*3) diplotypes (B) across multiple populations in comparison to the Thai population diplotype frequency.

Figure 3 Distribution of CYP2C19 polymorphisms in Thais and ethnic groups.

The distribution of poor metabolizers (PMs) of CYP2C19 (*2/*2, *2/*3, and *3/*3) was 19.00% in Native Japanese,32 14.02% in Koreans,33 10.42% in Han Chinese,31 9.82% in Indians,30 8.53% in Thais, 7.44% in North Indians,29 6.32% in Bhutanese,34 6.06% in Africans,26 4.84% in Malaysians,36 4.63% in Venezuelans,27 4.40% in African-Americans,25 4.17% in Caucasians,24 4.00% in Vietnamese,35 3.77% in Tanzanians,28 2.72% in Macedonians,23 1.67% in Italians,22 and 1.60% in Hispanics.25 The PMs prevalence was significantly higher in Thai patients compared to Italian (p-value = 0.0001), Macedonian (p-value = 0.0168), and Hispanic (p-value = 0.0006) patients. Conversely, the PMs prevalence in the Thai population was lower than in the Native Japanese population (OR = 2.51, 95% CI = 1.34–4.86, p-value = 0.0024). The odds ratios for these comparisons are illustrated in Figure 2A and B using forest plots.

Discussion

According to the Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines, the CYP2C19 gene is a significant pharmacogenomic polymorphism in various populations.20,37 In clinical settings, CYP2C19 plays a pivotal role in metabolizing approximately 8–10% of commonly used medications, including proton pump inhibitors, anticonvulsants, antiplatelet agents, and antidepressants.14,24 Notably, patients carrying homozygous alleles (PMs) and heterozygous alleles (IMs) of CYP2C19*2 and *3 variants are reported to have approximately 1.8-fold and 1.5-fold increased risk, respectively, of developing severe adverse cardiovascular events (e.g.death, myocardial infarction, stroke) following clopidogrel treatment.38,39 Thus, non-functional CYP2C19 variants significantly impact the dosage requirements for clopidogrel therapy in individual patients.

In our study, the most prevalent non-functional variant was the CYP2C19*2 allele, found in approximately 25.36% of our sample, consistent with previous reports in various populations, including Asian populations (Han Chinese, Native Japanese, Korean, Bhutanese, Vietnamese, and North Indians), Africans, African-Americans, Venezuelans, and Tanzanians.25–28,31–36 In contrast, Caucasian, Hispanic, Macedonian, and Italian populations exhibited lower frequencies of the CYP2C19*2 allele compared to the Thai population.22–25 However, these variations were attributed to the distribution of non-functional CYP2C19 alleles in each population and their impact on clopidogrel efficacy. Further investigations are necessary to confirm the influence of CYP2C19*2 distribution across ethnicities and the effects on clinically relevant medications.

The allele frequency of CYP2C19*3 (c C806T) in our study was approximately 4.50%, similar to earlier reports in the Thai population.40 Additionally, the frequency of CYP2C19*3 in Han Chinese was 3.27%, consistent with other Asian populations, as shown in Table 3 and Figure 1B. Interestingly, the distribution of CYP2C19*3 in these Asian populations, including Thai, closely resembled each other, except for Bhutanese, Korean, and North Indian populations. Specifically, we observed a higher distribution of CYP2C19*3 in the Bhutanese population compared to the Thai population (p-value = 8.94×10−6). However, the prevalence of the CYP2C19*3 allele was absent in Italian, Macedonian, Caucasian, Hispanic, African, Tanzanian, and North Indian populations.22–26,28,29 This genetic variability across racial and ethnic groups underscores the importance of CYP2C19*3 allele frequency as a pharmacogenomics marker for screening different ethnicities to prevent therapeutic failures in individual patients, particularly in the Asian population.

None of the Thai participants in our study carried the homozygous CYP2C19*3/*3 allele. However, PMs phenotypes based on CYP2C19*2/*2 and *2/*3 genotypes were present in approximately 6.16% and 2.37% of the sample, respectively. Our findings align with previous research indicating a PMs distribution of approximately 19% in Asian populations compared to around 2% in Caucasians.41 Notably, substantial variation in the distribution of these CYP2C19 genotypes was observed across populations, with less than 1% occurrence of these variants (*1/*3, *2/*3, and *3/*3) in European and African American populations, contrasting with frequencies of *1/*3 (7.34%), *2/*3 (3.33%), and *3/*3 (0.44%) in East Asian populations.42 Given the genetic diversity of CYP2C19 variants across ethnicities, determining the CYP2C19*3 genotype and phenotype before treatment initiation is particularly advantageous for Asian populations. Moreover, the understanding of CYP2C19 polymorphisms can significantly impact patient prognosis and outcomes, particularly in the context of drugs like clopidogrel, where genetic variations affect drug metabolism and efficacy. However, the relationship between CYP2C19 phenotype frequency and drug efficacy or adverse drug reactions (ADRs) warrants further investigation in other populations.

Conversely, the CYP2C19*17 variant (c. −806C>T) demonstrates ultra-rapid CYP2C19 activity and holds a significant role in the metabolic pathways of therapeutic drugs, which could heighten the risk of treatment failure with this substrate. From a clinical standpoint, the CYP2C19*17 variant strongly correlates with an increased risk of clopidogrel-induced bleeding in patients with cardiovascular and cerebrovascular diseases (OR = 1.89, 95% CI: 1.09–3.25, p-value = 0.02). This association is attributed to the heightened transcriptional activity of the CYP2C19 enzyme and its exaggerated response to clopidogrel’s prodrug.43 Conversely, this variant exhibits a notable association with a reduced risk of major adverse cardiovascular and cerebrovascular events in patients with coronary artery disease (OR = 0.76, 95% CI: 0.60–0.98, p = 0.03).43 Furthermore, the CYP2C19*17 genotype has been observed at higher frequencies among European and African populations, approximately 15.0–25.0%, whereas the Asian population exhibits a much lower frequency, approximately 4%.44 Given these trends, further exploration of the frequency of CYP2C19*17 variants and their association with adverse drug reactions is warranted across various populations.

One limitation of our study stems from the genotyping method employed for CYP2C19. We utilized real-time PCR to detect two single nucleotide polymorphisms (SNPs), rs4244285 (*2) and rs4986893 (*3). In cases where individuals did not carry these two variants, we presumed the genotype to be *1. However, this assumption may not accurately represent the true *1 status, as other untested variants could be present. This methodological approach has the potential to affect the precision of our genotyping Results and subsequent interpretations of CYP2C19 metabolic phenotypes. While real-time PCR is a highly reliable technique, the importance of validation cannot be overstated. To mitigate this concern, future studies should consider validating genotyping results using DNA sequencing methods, which offer a more comprehensive analysis of CYP2C19 variants and minimize potential errors. By integrating such validation steps, we can enhance the accuracy of genotyping and ensure a more dependable identification of metabolic phenotypes, thereby enhancing the relevance of our findings in clinical practice. In future research, we plan to include clinical data to investigate how these genetic variations impact drug response and adverse reactions in real-world scenarios.

In conclusion, the distribution of the CYP2C19 gene via non-functional alleles holds promise for implementation in clinical practice and relies on an ethnic-specific genetic variation database. Specifically, the genotype and phenotype of CYP2C19*3 could serve as a pharmacogenomics biomarker in Thai and Asian populations for screening, mitigating genetic associations with adverse drug reactions induced by certain medications.

Acknowledgments

This study was supported by grants from the (1) College of Pharmacy, Rangsit University; (2) Research Institute of Rangsit University (64/2563); (3) Doctor Kasem Foundation (049/2565); and (4) Thailand Center of Excellence for Life Sciences (TCELS) (TC 24/63), Thailand. The authors thank the study participants and staff of the Excellence of Pharmacogenomics and Precision Medicine Centre, College of Pharmacy, Rangsit University, National Biobank of Thailand (NBT) and Pharmacogenomics and Personalized Medicine of Ramathibodi Hospital, Mahidol University.

Disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest or financial conflict with the subject matter or materials discussed in this manuscript.

References

1. Reichhart DW, Feyereisen R. Cytochromes P450: a success story. Genome Biol. 2000;1(6):1–7.

2. Sim SC, Kacevska M, Ingelman-Sundberg M. Pharmacogenomics of drug-metabolizing enzymes: a recent update on clinical implications and endogenous effects. Pharmacogenomics J. 2013;13(1):1–11. doi:10.1038/tpj.2012.45

3. Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome P450 enzymes and its clinical impact. Drug Metab Rev. 2009;41(2):89–295. doi:10.1080/03602530902843483

4. Trenaman SC, Bowles SK, Andrew MK, Goralski K. The role of sex, age and genetic polymorphisms of CYP enzymes on the pharmacokinetics of anticholinergic drugs. Pharmacol Res Perspect. 2021;9(3):1–33. doi:10.1002/prp2.775

5. Zhou L, Sharma P, Yeo KR, et al. Assessing pharmacokinetic differences in Caucasian and East Asian (Japanese, Chinese and Korean) populations driven by CYP2C19 polymorphism using physiologically-based pharmacokinetic modelling. Eur J Pharm Sci. 2019;139:1–10.105061. doi:10.1016/j.ejps.2019.105061

6. Goldstein JA. Clinical relevance of genetic polymorphisms in the human CYP2C subfamily. Br J Clin Pharmacol. 2001;52(4):349–355. doi:10.1046/j.0306-5251.2001.01499.x

7. Wan-Po AL, Girard T, Farndon P, Cooley C, Lithgow J. Pharmacogenetics of CYP2C19: functional and clinical implications of a new variant CYP2C19*17. Br J Clin Pharmacol. 2010;69(3):222–230. doi:10.1111/j.1365-2125.2009.03578.x

8. Ellithi M, Baye J, Wilke RA. CYP2C19 genotype-guided antiplatelet therapy: promises and pitfalls. Pharmacogenomics. 2020;21(12):889–897. doi:10.2217/pgs-2020-0046

9. Lo C, Nguyen S, Yang C, et al. Pharmacogenomics in Asian subpopulations and impacts on commonly prescribed medications. Clin Transl Sci. 2020;13(5):861–870. doi:10.1111/cts.12771

10. Santos PC, Soares RA, Santos DB, et al. CYP2C19 and ABCB1 gene polymorphisms are differently distributed according to ethnicity in the Brazilian general population. BMC Med Genet. 2011;12:13.1–7. doi:10.1186/1471-2350-12-13

11. Ghodke Y, Joshi K, Arya Y, et al. Genetic polymorphism of CYP2C19 in Maharashtrian population. Eur J Epidemiol. 2007;22(12):907–915. doi:10.1007/s10654-007-9196-0

12. Sun H, Qu Q, Chen ZF, et al. Impact of CYP2C19 variants on clinical efficacy of clopidogrel and 1-year clinical outcomes in coronary heart patients undergoing percutaneous coronary intervention. Front Pharmacol. 2016;7:1–9. eCollection 2016. doi:10.3389/fphar.2016.00453

13. Yadav AK, Chakkumkollath AK, Helna A, et al. Substantiation of a clopidogrel metabolism-associated gene (CYP2C19) variation among healthy individuals. Indian Heart J. 2023;S0019–4832(23). doi:10.1016/j.ihj.2023.05.005

14. Desta Z, Zhao X, Shin JG, Flockhart DA. Clinical significance of the cytochrome P450 2C19 genetic polymorphism. Clin Pharmacokinet. 2002;41(12):913–958. doi:10.2165/00003088-200241120-00002

15. Bahar MA, Setiawan D, Hak E, Wilffert B. Pharmacogenetics of drug-drug interaction and drug-drug-gene interaction: a systematic review on CYP2C9, CYP2C19 and CYP2D6. Pharmacogenomics. 2017;18(7):701–739. doi:10.2217/pgs-2017-0194

16. Chaudhry SR, Muhammad S, Eidens M, et al. Pharmacogenetic prediction of individual variability in drug response based on CYP2D6, CYP2C9 and CYP2C19 genetic polymorphisms. Curr Drug Metab. 2014;15(7):711–718. doi:10.2174/1389200215666141125121952

17. Lee SJ. Clinical Application of CYP2C19 pharmacogenetics toward more personalized medicine. Front Genet. 2012;3:1–7. doi:10.3389/fgene.2012.00318

18. Patti G, Micieli G, Cimminiello C, Bolognese L. The Role of Clopidogrel in 2020: a Reappraisal. Cardiovasc Ther. 2020;2020:1–12. doi:10.1155/2020/8703627

19. Brown SA, Pereira N. Pharmacogenomic Impact of CYP2C19 Variation on Clopidogrel Therapy in Precision Cardiovascular Medicine. J Pers Med. 2018;8(1):1–31. doi:10.3390/jpm8010008

20. Lee CR, Luzum JA, Sangkuhl K, et al. Clinical Pharmacogenetics implementation consortium guideline for CYP2C19 genotype and clopidogrel therapy: 2022 update. Clin Pharmacol Ther. 2022;112(5):959–967. doi:10.1002/cpt.2526

21. CYP2C19 frequency table; Cited 29 Sept 2022. Available from: https://files.cpicpgx.org/data/report/current/frequency/CYP2C19_frequency_table.xlsx. Accessed June 27, 2024.

22. Serpe L, Canaparo R, Scordo MG, Spina E. Pharmacogenetics of drug-metabolizing enzymes in Italian populations. Drug Metab Pers Ther. 2015;30(2):107–120. doi:10.1515/dmdi-2014-0028

23. Jakovski K, Nestorovska AK, Labacevski N, Dimovski AJ. Characterization of the most common CYP2C9 and CYP2C19 allelic variants in the population from the Republic of Macedonia. Pharmazie. 2013;68(11):893–898.

24. Vidović S, Škrbić R, Stojiljković MP, et al. Prevalence of five pharmacologically most important CYP2C9 and CYP2C19 allelic variants in the population from the Republic of Srpska in Bosnia and Herzegovina. Arh Hig Rada Toksikol. 2021;72(3):129–134. doi:10.2478/aiht-2021-72-3499

25. Martis S, Peter I, Hulot JS, Kornreich R, Desnick RJ, Scott SA. Multi-ethnic distribution of clinically relevant CYP2C genotypes and haplotypes. Pharmacogenomics J. 2013;13(4):369–377. doi:10.1038/tpj.2012.10

26. Santos PCJL, Soares RAG, Santos DBG, et al. CYP2C19 and ABCB1 gene polymorphisms are differently distributed according to ethnicity in the Brazilian general population. BMC Med Genet. 2011;12:1–7. doi:10.1186/1471-2350-12-13

27. de Guerra DC, Flores S, Izaguirre MH. Distribution of CYP2C19*2 and CYP2C19*3 polymorphisms in Venezuelan populations with different admixture. Ann Hum Biol. 2013;40(2):197–200. doi:10.3109/03014460.2012.749946

28. Dandara C, Masimirembwa CM, Magimba A, et al. Genetic polymorphism of CYP2D6 and CYP2C19 in east- and Southern African populations including psychiatric patients. Eur J Clin Pharmacol. 2001;57(1):11–17. doi:10.1007/s002280100282

29. Lamba JK, Dhiman RK, Kohli KK. CYP2C19 genetic mutations in North Indians. Clin Pharmacol Ther. 2000;68(3):328–335. doi:10.1067/mcp.2000.109365

30. Adithan C, Gerard N, Vasu S, Rosemary J, Shashindran CH, Krishnamoorthy R. Allele and genotype frequency of CYP2C19 in a Tamilian population. Br J Clin Pharmacol. 2003;56(3):331–333. doi:10.1046/j.1365-2125.2003.01883.x

31. Chen L, Qin S, Xie J, et al. Genetic polymorphism analysis of CYP2C19 in Chinese Han populations from different geographic areas of mainland China. Pharmacogenomics. 2008;9(6):691–702. doi:10.2217/14622416.9.6.691

32. Myrand SP, Sekiguchi K, Man MZ, et al. Pharmacokinetics/genotype associations for major cytochrome P450 enzymes in native and first- and third-generation Japanese populations: comparison with Korean, Chinese, and Caucasian populations. Clin Pharmacol Ther. 2008;84(3):347–361. doi:10.1038/sj.clpt.6100482

33. Kim KA, Song WK, Kim KR, Park JY. Assessment of CYP2C19 genetic polymorphisms in a Korean population using a simultaneous multiplex pyrosequencing method to simultaneously detect the CYP2C19*2, CYP2C19*3, and CYP2C19*17 alleles. J Clin Pharm Ther. 2010;35(6):697–703. doi:10.1111/j.1365-2710.2009.01069.x

34. Dorji PW, Wangchuk S, Boonprasert K, Tarasuk M, Bangchang KN. Pharmacogenetic relevant polymorphisms of CYP2C9, CYP2C19, CYP2D6, and CYP3A5 in Bhutanese population. Drug Metab Pers Ther. 2019;34(4):1–14. doi:10.1515/dmpt-2019-0020

35. Vu NP, Thanh Nguyen HT, Bich Tran NT, et al. CYP2C19 genetic polymorphism in the Vietnamese population. Ann Hum Biol. 2019;46(6):491–497. doi:10.1080/03014460.2019.1687750

36. Ang GY, Yu CY, Subramaniam V, et al. Detection of CYP2C19 genetic variants in Malaysian orang asli from massively parallel sequencing data. PLoS One. 2016;11(10):1–12. doi:10.1371/journal.pone.0164169

37. Galindo IF, Garro CC, Soares FR, et al. Interethnic variation of CYP2C19 alleles, ‘predicted’ phenotypes and ‘measured’ metabolic phenotypes across world populations. Pharmacogenomics J. 2016;16(2):113–123. doi:10.1038/tpj.2015.70

38. Mega JL, Simon T, Collet JP, et al. Reduced-function CYP2C19 genotype and risk of adverse clinical outcomes among patients treated with clopidogrel predominantly for PCI: a meta-analysis. JAMA. 2010;304(16):1821–1830. doi:10.1001/jama.2010.1543

39. Xi Z, Fang F, Wang J, AlHelal J, Zhou Y, Liu W. CYP2C19 genotype and adverse cardiovascular outcomes after stent implantation in clopidogrel-treated Asian populations: a systematic review and meta-analysis. Platelets. 2019;30(2):229–240. doi:10.1080/09537104.2017.1413178

40. Sukasem C, Tunthong R, Chamnanphon M, et al. CYP2C19 polymorphisms in the Thai population and the clinical response to clopidogrel in patients with atherothrombotic-risk factors. Pharmgenomics Pers Med. 2013;6:85–91. doi:10.2147/PGPM.S42332

41. Mizutani T. PM frequencies of major CYPs in Asians and Caucasians. Drug Metab Rev. 2003;35(2–3):99–106. doi:10.1081/dmr-120023681

42. Ionova Y, Ashenhurst J, Zhan J, 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. Clin Transl Sci. 2020;13(6):1298–1306. doi:10.1111/cts.12830

43. Huang B, Cui DJ, Ren Y, Han B, Yang DP, Zhao X. Effect of cytochrome P450 2C19*17 allelic variant on cardiovascular and cerebrovascular outcomes in clopidogrel-treated patients: a systematic review and meta-analysis. J Res Med Sci. 2017;22:1–10. doi:10.4103/jrms.JRMS_590_16

44. Nestorovska AK, Jakovski K, Naumovska Z, et al. Distribution of the most common genetic variants associated with a variable drug response in the population of the Republic of Macedonia. Balkan J Med Genet. 2014;17(2):5–14. doi:10.2478/bjmg-2014-0069

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.