Back to Journals » Pharmacogenomics and Personalized Medicine » Volume 11

Pharmacogenetic association study on clopidogrel response in Puerto Rican Hispanics with cardiovascular disease: a novel characterization of a Caribbean population

Authors Hernandez-Suarez DF , Botton MR , Scott SA , Tomey MI, Garcia MJ, Wiley J, Villablanca PA, Melin K , Lopez-Candales A, Renta JY , Duconge J 

Received 17 February 2018

Accepted for publication 29 March 2018

Published 8 June 2018 Volume 2018:11 Pages 95—106

DOI https://doi.org/10.2147/PGPM.S165805

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Martin Bluth



Dagmar F Hernandez-Suarez,1 Mariana R Botton,2 Stuart A Scott,2 Matthew I Tomey,3 Mario J Garcia,4 Jose Wiley,4 Pedro A Villablanca,5 Kyle Melin,6 Angel Lopez-Candales,7 Jessicca Y Renta,8 Jorge Duconge9

1Cardiovascular Medicine Division, Department of Medicine, University of Puerto Rico School of Medicine, San Juan, PR, USA; 2Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; 3Cardiovascular Medicine Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; 4Division of Cardiovascular Diseases, Department of Medicine, Montefiore Medical Center/Albert Einstein College of Medicine New York, NY, USA; 5Division of Cardiology, Department of Medicine, New York University Langone Medical Center, New York, NY, USA; 6Department of Pharmacy Practice, University of Puerto Rico Medical Sciences Campus, San Juan, PR, USA; 7Cardiovascular Medicine Division, Department of Medicine, University of Puerto Rico School of Medicine, San Juan, PR, USA; 8Department of Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, PR, USA; 9Pharmaceutical Sciences Department, University of Puerto Rico Medical Sciences Campus, San Juan, PR, USA

Introduction: High on-treatment platelet reactivity (HTPR) to clopidogrel imparts an increased risk for ischemic events in adults with coronary artery disease. Platelet reactivity varies with ethnicity and is influenced by both clinical and genetic variables; however, no clopidogrel pharmacogenetic studies with Puerto Rican patients have been reported. Therefore, we sought to identify clinical and genetic determinants of on-treatment platelet reactivity in a cohort of Puerto Rican patients with cardiovascular disease.
Methods: We performed a retrospective study of 111 patients on 75 mg/day maintenance dose of clopidogrel. Patients were allocated into 2 groups: Group I, without HTPR; and Group II, with HTPR. Platelet function was measured ex vivo using the VerifyNow® P2Y12 assay and HTPR was defined as P2Y12 reaction units (PRU) ≥230. Genotyping testing was performed using Taqman® Genotyping Assays.
Results: The mean PRU across the cohort was 203±61 PRU (range 8–324), and 42 (38%) patients had HTPR. Multiple logistic regression showed that 27% of the total variation in PRU was explained by a history of diabetes mellitus, hematocrit, CYP2C19*2, and PON1 p.Q192R. Body mass index (odds ratio [OR]=1.15; 95% CI: 1.03–1.27), diabetes mellitus (OR=3.46; 95% CI: 1.05–11.43), hematocrit (OR=0.75; 95% CI: 0.65–0.87), and CYP2C19*2 (OR=4.44; 95% CI: 1.21–16.20) were the only independent predictors of HTPR.
Conclusion: Moreover, we propose a predictive model to determine PRU values as measured by VerifyNow P2Y12 assay for the Puerto Rican Hispanic population. This model has the potential to identify Hispanic patients at higher risk for adverse events on clopidogrel.

Keywords: clopidogrel, platelet reactivity, genotyping, Hispanics, Puerto Rico

Introduction

Clopidogrel is a platelet adenosine diphosphate (ADP) receptor inhibitor commonly used to prevent thrombotic events in patients with acute coronary syndrome (ACS), ischemic stroke, carotid artery stenosis (CAS), and peripheral artery disease (PAD). Clopidogrel remains one of the most widely prescribed ADP receptor blockers, used by up to 40,000,000 people and with previous reports of up to 9 billion dollars a year in global sales.1,2 However, significant variability in clinical response and platelet inhibition has been observed, which can lead to decreased antithrombotic effect for some patients.3 Multiple clinical, cellular, genetic, and pharmacokinetic factors have been suggested as determinants of this variability;4 however, the evidence supporting these variables and their effect on platelet reactivity among patients on clopidogrel is still conflicting.5 In addition, the importance of ancestry and ethnicity on clopidogrel responsiveness among patients of underrepresented minority populations is still uncertain and warrants further research.6

To date, most clopidogrel responsiveness studies have focused on the association between CYP2C19 variant alleles (e.g., CYP2C19*2 and *3) and both high on-treatment platelet reactivity (HTPR) and adverse cardiovascular events.7,8 In addition, PON1 p.Q192R, ABCB1 c.3435C>T, and the P2RY12 H2 haplotype have also been proposed as contributors to adverse outcome risk on clopidogrel.9 In contrast, the CYP2C19*17 allele increases expression and enzyme activity, leading to enhanced platelet inhibition and a higher risk of bleeding.10 However, only ~12% of the variability in clopidogrel response has been explained by CYP2C19 alone, suggesting that other important clinical, genetic, or environmental factors have yet to be identified.1114

Hispanics have a higher prevalence of cardiovascular risk factors, recurrence rate of thrombotic events, and worse cardiovascular outcomes when compared with non-Hispanic Caucasians; however, there is a paucity of antiplatelet studies reported on this group. As such, greater efforts should be directed to include this population in cardiovascular pharmacogenetic studies. Interestingly, among all Hispanic sub-groups living within the USA and its territories, Puerto Ricans are the only one with higher age-adjusted death rates when compared with non-Hispanic Whites.15 Additionally, among Hispanic adults of diverse backgrounds, the prevalence of adverse cardiovascular disease risk profiles is higher among Puerto Ricans.16 Moreover, our preliminary findings in Puerto Ricans suggest that interindividual variation in ancestral contributions have significant implications on clopidogrel responsiveness.17,18 Hence, we hypothesized that due to the trihybrid admixture and high prevalence of cardiovascular risk factors, Puerto Ricans might have a unique clinical and genetic contribution to clopidogrel responsiveness that could change our current approach to antiplatelet therapy in this population.

Furthermore, clopidogrel is preferred among ADP receptor blockers in Puerto Rico, largely because of its availability as a generic drug and lower cost. Since genetic determinants of impaired response to clopidogrel are not currently known in Caribbean Hispanics, we sought to determine pharmacogenetic variants associated with platelet reactivity to clopidogrel in Puerto Rican Hispanic patients.

Methods

Study design and ethics

This was a multicenter case–control study of Puerto Rican Hispanics patients receiving antiplatelet therapy who were recruited between January and February 2017. The Human Research Subjects Protection Office (HRSPO) approved this study (Protocol No. A4070416). HRSPO serves as the administrative office for the University of Puerto Rico Medical Science Campus Institutional Review Boards (assurance #FWA00005561). The protocol was also conducted in accordance with the Declaration of Helsinki and in compliance with Good Clinical Practice. All patients provided written informed consent.

Patient population and data collection

A total of 111 patients of Hispanic Puerto Rican descent on clopidogrel therapy were consecutively recruited at the University District Hospital and the Cardiovascular Center of Puerto Rico and the Caribbean, in San Juan, PR. Past medical history and preadmission laboratory data were obtained from medical record. Puerto Rican Hispanics aged ≥21 years on 75 mg/day maintenance dose of clopidogrel for at least 7 consecutive days were included in the study. Exclusion criteria included the use of any oral anticoagulant or glycoprotein IIb/IIIa receptor inhibitors, administration of other ADP receptor blocker other than clopidogrel within 2 weeks of enrollment, hematocrit (Hct) ≤25%, platelet count <100×109/L, blood urine nitrogen (BUN)/creatinine >30/1.5 mg/dL, known platelet function disorder, or active hepatic disease. Patients were allocated into 2 groups based on P2Y12 reaction units (PRU) cutoff values:7,19 Group I without HTPR (PRU<230) and Group II with HTPR (PRU≥230).

Platelet function testing

Blood samples for platelet function and genetic testing were collected within 2 days of a scheduled vascular/cardiac minimally invasive procedure during the preadmission evaluation. Whole blood was drawn from a peripheral vein through a 22-gauge needle. An initial 2 mL sample was collected from each participant and saved for other laboratory tests as part of the preadmission process. A second tube containing 3.2% sodium citrate was obtained with 2 mL blood for platelet function testing. Platelet function was measured ex vivo using the US Food and Drug Administration-approved point-of-care VerifyNow® P2Y12 analyzer following the manufacturer’s instructions (Accumetrics, Inc., San Diego, CA, USA).

Since the definition of HTPR in the Hispanic population is unknown, we used the upper tertile PRU value (≥230) previously obtained by our group in a pilot study of Puerto Rican patients on clopidogrel therapy to define HTPR.20 This value has also been used to define HTPR in other studies7,19 and is similar to other cutoffs reported in literature.21,22 In addition, a PRU value of 230 has been recognized by the manufacturer as the cutoff for samples with no platelet inhibition.

DNA extraction and genotyping

Genomic DNA was extracted using the QIAamp DNA Blood Maxi Kit (spin protocol; QIAGEN Inc., Venlo, Limburg) according to the manufacturer’s instructions and genotyped for a panel of candidate variants using TaqMan® SNP Genotyping Assay Reagent kits (Applied Biosystems, Carlsbad, CA, USA): CYP2C19*2, *3, *4 and *17; ABCB1 c.3435C>T; PON1 p.Q192R; P2RY12 H2 haplotype; B4GALT2 c.909C>T, and c.366G>C variants; CES1 c.428G>A and PEAR1 rs12041331 and rs2768759.23

Statistical analysis

Continuous variables were compared using the 2-tailed Student’s t-test while Chi-square or Fisher’s exact tests were used for categorical data as appropriate. The Hardy–Weinberg equilibrium (HWE) test was applied as a quality control for genotyping; deviation from HWE was estimated using a χ2 goodness-of-fit test with 1° of freedom. Comparison of minor allele frequencies (MAFs) between our cohort and reference populations were performed using a Z-test for population proportions. Simple linear regression analysis was performed to determine the association between all measurements and platelet reactivity. A multivariate linear regression was used to identify the contribution of all significant variables to platelet reactivity variability, and a multiple logistic regression was performed to determine predictors of HTPR. The corresponding odds ratio (OR) and 95% CI were calculated. Statistical analyses were performed using SAS software version 9.4 (SAS Institute, Cary, NC, USA), and p-values<0.05 were considered statistically significant.

Results

Study population

All the study participants (n=111) underwent platelet function testing and genotyping. Enrolled individuals were distributed into the non-HTPR (n=69, 62%) and HTPR (n=42, 38%) groups. The indications for the patient cohort included coronary artery disease (CAD; 77%), PAD (34%), CAS (7%), cerebral artery aneurysm (2%), and stroke (2%). All patients were on 75 mg/day maintenance dose of clopidogrel for >7 days. The patient baseline characteristics are detailed in Table 1. HTPR patients had higher body mass index (BMI), prevalence of diabetes mellitus (DM), use of proton-pump inhibitors (PPI), and calcium channel blockers (CCB), but lower hemoglobin and Hct levels compared with non-HTPR patients.

Table 1 Baseline clinical characteristics of the study patients according to on-treatment platelet reactivity

Note: aValues are mean ± SD or n (%).

Abbreviations: BMI, Body mass index; BUN, blood urine nitrogen; Hct, hematocrit; Hgb, hemoglobin; HTPR, high on-treatment platelet reactivity; WBC, white blood count.

Genotyping

The genotyping results are illustrated in Tables 2 and 3. Approximately 25% of patients carried at least 1 copy of the CYP2C19*2 allele, which was the only variant with different allele frequencies between groups. Due to the low frequency of homozygous variant allele carriers for most of the tested genes (e.g., CYP2C19*2/*2), carriers of at least 1 variant allele were compared with wild-type individuals (Table 3). Notably, the non-HTPR group had a higher proportion of patients with at least 1 copy of the PON1 p.Q192R variant.

Table 2 Genotype frequencies of all polymorphisms of interest in the studied groups and their corresponding p-values after comparison between HTPR and non-HTPR

Notes: Departure from the HWE is also shown (p-values in this column correspond to the χ2 goodness-of-fit test, with 1° of freedom). aValues are represented in n (%). Alleles CYP2C19*3 and CYP2C19*4 were not observed in the study population.

Abbreviations: HTPR, high on-treatment platelet reactivity, HWE, Hardy–Weinberg equilibrium.

Table 3 Genotype frequency of carriers of at least 1 variant allele

Notes: aValues are represented in n (%). bCarrier includes genotypes with at least 1 copy of the mutant allele. Alleles CYP2C19*3 and CYP2C19*4 were not observed in the study population. P2RY12, B4GALT2, and CES1 variants were not included as no homozygotes for the mutant allele were reported.

Abbreviation: HTPR, high on-treatment platelet reactivity.

The MAFs of CYP2C19*2, CYP2C19*17, ABCB1 3435C>T, P2RY12 H2, B4GALT2 909C>T, B4GALT2 366G>C, PEAR1 rs12041331, and PON1 p.Q192R in the study cohort were 15%, 14%, 37%, 8%, 6%, 3%, 20%, and 47%, respectively. We also compared all the MAFs observed in the study population with those from reference populations (data taken from the 1000 Genomes Phase I project).24 We found no significant differences in MAFs between our cohort and the Puerto Ricans sample included in the 1000 Genomes Phase I project (p>0.05). No significant departure from HWE was detected for the tested variants except for CYP2C19*2 (Table 2), which was attributed to the relatively small sample size of the cohort.

Determinants of platelet reactivity

The distribution of platelet reactivity in the study cohort is illustrated in Figure 1. The mean platelet reactivity to clopidogrel was 203±61 PRU (range: 8–324). Five clinical factors (age, history of DM, Hct, BUN, use of CCB) and 2 genetic variants (CYP2C19*2, PON1 p.Q192R) were associated with platelet reactivity by linear regression (Table 4). Notably, Hct and PON1 p.Q192R were negatively correlated with platelet reactivity. Although not significant, smoking had a tendency to decrease PRU values while the use of PPI had the opposite effect (Figure 2). All the factors associated with platelet reactivity, as well as BMI and PPI (Table 4), were included in a multiple linear regression analysis. DM, Hct, CYP2C19*2, and PON1 p.Q192R were the only independent predictors of platelet reactivity after adjusting for all possible confounders and interactions. The contribution of the studied genetic variants to platelet reactivity was ~6%; however, the model increased to 34% when clinical variables were added (Table 5).

Figure 1 Distribution of platelet reactivity as measured by PRU.

Abbreviations: HTPR, high on-treatment platelet reactivity; PRU, P2Y12 reaction units.

Table 4 Simple linear regression between important variables and platelet reactivity

Note: aCES1 was not included due to the observed low frequency (n=2).

Abbreviations: BMI, body mass index; BUN, blood urine nitrogen; CCB, calcium channel blockers; Hct, hematocrit; PPI, proton-pump inhibitors.

Figure 2 Association between important clinical and genetic characteristics with platelet reactivity.

Abbreviations: CCB, calcium channel blockers; PPI, proton-pump inhibitors; PRU, P2Y12 reaction units.

Table 5 Contribution of study variables to the interindividual variability in on-treatment platelet reactivity

Notes: aCYP2C19*2 and PON1 p.Q192R. b7 clinical variables (5 variables associated with platelet reactivity in Table 4, body mass index and proton-pump inhibitor).

Development of a predictive model for platelet reactivity

An initial predictive model to estimate platelet reactivity was obtained from the 9 variables included in the multiple linear regression analysis. This initial model estimates PRU by the following equation:

Initial model (R2=34%, p<0.0001):

where age is in years, BMI in kg/m2, Hct in %, BUN in mg/dL, DM, PPI, and CCB are coded as 0 if absent and 1 if present. The CYP2C19*2 and PON1 p.Q192R variants are coded as 0 for homozygous wild-type genotypes and 1 if at least 1 copy of the variant allele is carried.

Since clinical genotyping may not be feasible in some medical settings, particularly in low-resource institutions, we designed a clinical model to be considered in the case of unavailable genetic data. To obtain this model, a multiple linear regression analysis was first performed excluding the genetic data. BMI, DM, and Hct were the only clinical variables that predicted platelet reactivity and, therefore, were included in the following model:

Clinical model (R2=23%, p<0.0001):

Finally, a final model was obtained from the 4 clinical and genetic variables that independently predicted platelet reactivity:

Final model (R2=27%, p<0.0001):

Model diagnostic analysis is shown in Figure 3. As seen, the final model has a normal probability plot for residuals and a good visual correlation between the observed versus fitted PRU values.

Figure 3 Model diagnostics analysis.

Notes: (A) Q-Q plot for normality analysis of residuals showing a normal distribution. (B) Observed versus fitted PRU values showing a strong visual correlation.

Abbreviation: PRU, P2Y12 reaction units.

Predictors of HTPR phenotype

Clinical and genetic characteristics associated with HTPR are presented in Table 6. DM, BMI, Hct, BUN, PPI, CCB, CYP2C19*2, and PON1 p.Q192R were all correlated with HTPR. Interestingly, Hct and PON1 p.Q192R had a protective effect on determining HTPR. When all these variables were included in a multivariable logistic regression analysis, we found that BMI (OR=1.15; 95% CI: 1.03–1.27), DM (OR=3.46; 95% CI: 1.05–11.43), Hct (OR=0.75; 95% CI: 0.65–0.87), and CYP2C19*2 (OR=4.44; 95% CI: 1.21–16.20) were the only factors independently correlated with PRU≥230.

Table 6 Stepwise logistic regression analysis to determine the best predictor of high on-treatment platelet reactivity

Abbreviations: BMI, body mass index; CCB, calcium channel blockers; Hct, hematocrit; OR, odd ratios; PPI, proton-pump inhibitors.

Discussion

The paucity of reports on antiplatelet response in the Hispanic population and recent reports linking ethnicity with platelet reactivity25 prompted our study to identify clinical and genetic determinants of on-treatment platelet reactivity among cardiovascular Puerto Rican patients on clopidogrel. To the best of our knowledge, no studies on clopidogrel pharmacogenetics in Caribbean Hispanics have previously been reported. Our study determined that DM, Hct, CYP2C19*2, and PON1 p.Q192R were predictors of platelet reactivity on clopidogrel in Puerto Rican patients. In addition, we developed a novel multivariable model to predict PRU (measured by VerifyNow-P2PY12) in the Hispanic population.

In the present study, 38% of our patient population had HTPR defined as a PRU≥230, which was generally consistent with data from Mallouk et al, indicating that between 16% and 50% of patients treated with clopidogrel had a poor biological response.26 However, comparisons of HTPR prevalence across different populations is challenging as HTPR depends on the cutoff value used. Furthermore, on-treatment platelet reactivity is multifactorial, as it is influenced by clinical and genetic factors, time and method of platelet reactivity assessment, antiplatelet indication, ancestry, and potentially other unknown variables.10,27

Puerto Rican Hispanics are unique as a result of their 500-year history of genomic admixture,28 yet little is known about the frequencies of important pharmacogenetic variants in this population. Prior reports have determined the frequencies of CYP2C19*2 and *3 in Puerto Ricans;29,30 however, no information regarding other CYP2C19 polymorphisms, clopidogrel pharmacogenes and their haplotype structure is currently known. We report the MAFs of 6 important clopidogrel pharmacogenes (CYP2C19, ABCB1, PON1, P2RY12, B4GALT2, CES1, and PEAR1) in a cohort of cardiovascular patients from the Commonwealth of Puerto Rico.

A recent review of CYP2C19 studies worldwide estimated a CYP2C19*2 frequency between 9% in North Africans and 61% in Native Oceanians.31 American admixed population had a frequency of 12%, which is similar to the 15% found in our study. Furthermore, we found no significant difference when the CYP2C19*2 frequency in our cohort was compared with that in the Puerto Rican population included in the 1000 Genomes Project (13%).24 In contrast, neither the CYP2C19*3 nor the CYP2C19*4 variants were identified in our cohort. This is consistent with previous reports in the Puerto Rican population.29,30 In addition, the CYP2C19*4 variant is very rare in the admixed American population.31

The CYP2C19, PON1, P2RY12, ABCB1, CES1, PEAR1, and, most recently, B4GALT2 genes have been previously linked to clopidogrel response.8,10,3234 To date, most studies have focused on the association between CYP2C19 and both HTPR and major adverse cardiovascular events.3537 Likewise, PON1 p.Q192R, ABCB1 c.3435C>T, P2RY12 H2 haplotype, and PEAR1 rs12041331 have been proposed to be related to the same outcomes. In contrast, the increased function CYP2C19*17 allele results in greater enzyme expression and activity, leading to an enhanced platelet inhibition and increased risk of bleeding. Furthermore, low on-treatment platelet reactivity, defined as PRU<50 has been recently associated with the B4GALT2 c.909C>T and c.366G>C variants in populations of European descendent.10 In our study, no significant associations were found between these B4GALT2 variants and platelet reactivity. The MAFs observed in the present cohort are lower than the frequencies previously reported in a cohort of individuals with mostly European ancestry (0.06 versus 0.10 for 909C>T and 0.03 versus 0.07 for 366G>C).10 Also, B4GALT2 909C>T and 366G>C polymorphisms were in strong linkage disequilibrium (p<0.001; D’=1.0; r2=0.523), which is expected for younger populations. The unique genetic background of Puerto Ricans might have contributed to this apparent discrepancy in the results. Thus, our data suggest that the association between these B4GALT2 variants with on-treatment platelet reactivity is likely population-specific.

Only ~12% of the variability in clopidogrel response is explained by CYP2C19 alone;38 however, heritability estimates suggest that genetics can account for up to 73% of such variance.11 In addition, these genes are involved in the bioactivation and metabolism of a large number of prescription drugs other than clopidogrel as well as drug classes, including antidepressants, benzodiazepines, mephenytoin, and PPI. Hence, the importance to study these genetic polymorphisms is not limited to antiplatelet drugs. Moreover, several non-genetic factors have been identified as possible determinants of poor biological response to clopidogrel, such as BMI, DM, concomitant use of some drugs, smoking status, Hct, and ethnicity.13,25,39,40

In the Caribbean population, the effects of clinical and genetic factors on clopidogrel responsiveness have not been fully elucidated. In our study, we found that DM, Hct, CYP2C19*2, and PON1 p.Q192R were the only independent predictors of interindividual variability in platelet reactivity as measure by VerifyNow. Together, these variables explained 27% of PRU variability. In a similar study, Larsen et al, reported 21% of PRU variability determined by >10 clinical and genetic factors for a New Zealand ACS population.41 Interestingly, both Hct and PON1 p.Q192R were inversely correlated with platelet reactivity to clopidogrel in our cohort. The negative relationship between Hct levels and clopidogrel response has been previously demonstrated;42 however, a correction of PRU for Hct lacks of clinical validity and does not improve the prediction of adverse cardiovascular events.43 The particular case of PON1 p.Q192R is more controversial. In 2011, Bouman et al, tested the clinical relevance of PON1 p.Q192R in patients undergoing percutaneous coronary intervention on clopidogrel treatment.44 They found that p.192Q was associated with a reduced conversion of 2-oxo-clopidogrel to the active thiol metabolite when compared with the PON1 p.192R variant. Consequently, p.Q192R resulted in a more efficient clopidogrel bioactivation. Moreover, p.192QQ homozygous individuals were at higher risk of stent thrombosis. Park et al, reported similar results in the CROSS-VERIFY Cohort study, where 1336 patients were genotyped and p.192Q was found to be an independent predictor of worse cardiovascular outcome and significantly associated with higher levels of small dense low-density lipoprotein cholesterol.45 However, subsequent studies have failed to demonstrate a clear association between PON1 and clopidogrel response variability.46 Our study supports a positive effect of PON1 on clopidogrel response in Puerto Ricans. In this population, patients with at least 1 copy of p.192R had a better response to clopidogrel, which underscores the importance of ancestry and admixture in determine the ultimate effect of pharmacogenetic variants.

Patients with DM have increased platelet reactivity and a higher percent of circulating immature platelets,13,47 which could reduce the inhibitory effect of clopidogrel on platelet aggregation. Moreover, the loss of responsiveness to insulin in DM patients could lead to a reduced response to antiplatelet drugs, resulting in heightened platelet reactivity.48 In fact, a recent study found an association between insulin receptor substrate-1 variants and HTPR with clopidogrel therapy in CAD patients with DM.49 In Puerto Ricans, the effect of this interaction may be more prominent as the prevalence of DM is much higher when compared with other ethnic groups.50,51

The other genotyped variants included in our study (CYP2C19*17, ABCB1, P2RY12, B4GALT2, CES1, and PEAR1) were not found to be significantly associated with platelet reactivity in the Puerto Rican population. Similarly, BMI, use of PPI and CCB, or being a smoker did not predict platelet reactivity after multivariate analysis was performed. Current evidence is inconsistent about whether these factors determine clopidogrel responsiveness.9,32,5257

HTPR is an objective measure of clopidogrel responsiveness and it has been associated with increased risk of adverse cardiovascular events.7,35 In our sample, BMI, DM, Hct, and CYP2C19*2 were predictors of PRU ≥230. Interestingly, we failed to find a significant association between PON1 p.Q192R and HTPR after multivariate analysis. Given a lack of consensus regarding the best cutoff PRU value to define HTPR and since a PRU≥230 might not be the true threshold value for Puerto Ricans, we constructed a PRU predictive model able to provide an objective quantitative measurement of platelet reactivity to clopidogrel. Although other authors have proposed similar models,42 none has been developed particularly for Hispanics and very few have considered patients with diagnosis other than coronary disease. Our study is novel as it offers clinicians a valuable clinical decision-support tool by using clinical and genetic information or clinical data alone, so they can identify patients at higher risk of having poor response to clopidogrel and thus facilitate prompt therapy optimization to minimize adverse cardiovascular events. Yet, further validation remains pending and the benefit of adding new variables still needs to be assessed. However, the proposed model is useful for a wide range of cardiovascular entities.

Some limitations of this study need to be highlighted. First, despite the relatively small sample, the study was adequately powered to discover strong risk predictors of HTPR with OR ≥3.0 for the homozygous risk genotype. However, weaker predictors conferring smaller risk increases may have remained undiscovered. Yet, this study was primarily designed to perform preliminary assessments on clopidogrel response in Puerto Rican Hispanics as a way to set the appropriate context for further pharmacogenetic studies in this population at large. Second, medication compliance was self-reported and, therefore, it is possible that some patients were not adherent to clopidogrel therapy. Finally, PRU was measured at a single time in each participant. Hence, intraindividual variability was not previously assessed. This could be important since repeat PRU measurements might detect important changes in clopidogrel responsiveness and help differentiate patients with and without adverse clinical events.58,59

Conclusion

In a representative sample of Puerto Rican patients with cardiovascular disease, DM, Hct, CYP2C19*2, and PON1 p.Q192R were associated with on-treatment platelet reactivity. Additionally, we developed a predictive model to determine PRU values as measured by VerifyNow P2Y12 assay for the Puerto Rican Hispanic population. This model may be critical in identifying Hispanic patients at higher risk for adverse events on clopidogrel.

Acknowledgments

We want to acknowledge first the patients for voluntarily participating in this study. Also, we would like to acknowledge the staff members of the Preadmission/Admission Department and Public Relationships Office at the Cardiovascular Center of Puerto Rico and the Caribbean, as well as the Endovascular Department of the University of Puerto Rico School of Medicine for their support during the development of this study. This publication was partially supported by the National Institute on Minority Health and Health Disparities (NIMHD) of the National Institutes of Health (NIH) Award Numbers CCTRECD-R25MD007607, TL1TR001434, U54MD007587 and the Research Minority Institutions (RCMI) award # 8G12 MD007600. JD is supported in part by the SC1 grant # HL123911 from the National Heart, Lung, and Blood Institute (NHLBI) and the Research Minority Institutions (RCMI) award # 2U54MD00760031. SAS is supported in part by the NIGMS of the NIH, through grant K23GM104401.

Disclosure

The authors report no conflicts of interest in this work.

References

1.

Kitzmiller JP, Groen DK, Phelps MA, Sadee W. Pharmacogenomic testing: relevance in medical practice: why drugs work in some patients but not in others. Cleve Clin J Med. 2011;78:243–257.

2.

Debnath B, Al-Mawsawi LQ, Neamati N. Are we living in the end of the blockbuster drug era? Drug New Perspect. 2010;23:670–684.

3.

Krishna V, Diamond GA, Kaul S. Do platelet function testing and genotyping improve outcome in patients treated with antithrombotic agents? The role of platelet reactivity and genotype testing in the prevention of atherothrombotic cardiovascular events remains unproven. Circulation. 2012;125:1288–1303.

4.

Geisler T, Gawaz M. Individualized antiplatelet therapy: what can a clinical score contribute? Hamostaseologie. 2009;29(4):360–367.

5.

Karathanos A, Geisler T. Monitoring aspirin and clopidogrel response: testing controversies and recommendations. Mol Diagn Ther. 2013;17:123–137.

6.

Zhang T, Tsang W, Wijeysundera HC, Ko DT. Reporting and representation of ethnic minorities in cardiovascular trials: a systematic review. Am Heart J. 2013;166:52–57.

7.

Price MJ, Berger PB, Teirstein PS, et al. Standard- vs high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention: the GRAVITAS randomized trial. JAMA. 2011;305:1097–1105.

8.

Scott SA, Sangkuhl K, Stein CM, et al. Clinical pharmacogenetics implementation consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update. Clin Pharmacol Ther. 2013;94:317–323.

9.

Wallentin L, James S, Storey RF, et al. Effect of CYP2C19 and ABCB1 single nucleotide polymorphisms on outcomes of treatment with ticagrelor versus clopidogrel for acute coronary syndromes: a genetic substudy of the PLATO trial. Lancet. 2010;376:1320–1328.

10.

Scott SA, Collet JP, Baber U, et al. Exome sequencing of extreme clopidogrel response phenotypes identifies B4GALT2 as a determinant of on-treatment platelet reactivity. Clin Pharmacol Ther. 2016;100:287–294.

11.

Shuldiner AR, O’Connell JR, Bliden KP, et al. Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA. 2009;302:849–857.

12.

Mega JL, Hochholzer W, Frelinger AL 3rd, et al. Dosing clopidogrel based on CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovascular disease. JAMA. 2011;306(20):2221–2228.

13.

Angiolillo DJ, Jakubowski JA, Ferreiro JL, et al. Impaired responsiveness to the platelet P2Y12 receptor antagonist clopidogrel in patients with type 2 diabetes and coronary artery disease. J Am Coll Cardiol. 2014;64(10):1005–1014.

14.

Samant S, Jiang XL, Peletier LA, et al. Identifying clinically relevant sources of variability: the clopidogrel challenge. Clin Pharmacol Ther. 2017;101(2):264–273.

15.

Escarce JJ, Morales LS, Rumbaut RG [webage on the Internet]. The Health Status and Health Behaviors of Hispanics. In: National Research Council (US) Panel on Hispanics in the United States; Tienda M, Mitchell F, editors. Hispanics and the Future of America. Washington, DC: National Academies Press; 2006. Available from: https://www.ncbi.nlm.nih.gov/books/NBK19899/. Accessed January 16, 2018.

16.

Daviglus ML, Talavera GA, Avilés-Santa ML, et al. Prevalence of major cardiovascular risk factors and cardiovascular diseases among Hispanic/Latino individuals of diverse backgrounds in the United States. JAMA. 2012;308:1775–1784.

17.

Duconge J, Ruaño G. Emerging role of admixture in the pharmacogenetics of Puerto Rican Hispanics. J Pharmacogenom Pharmacoproteomics. 2010;1(101):1000101.

18.

Duconge J, Escalera O, Korchela M, Ruaño G [webpage on the Internet]. Clinical implications of genetic admixture in Hispanic Puerto Ricans: impact on the pharmacogenetics of CYP2C19 and PON1. Chapter 7, In: Sanoudou D, editor. Clinical Applications of Pharmacogenetics. InTech, 2012:151–164. Available from: http://www.intechopen.com/books/clinical-applications-of-pharmacogenetics/clinical-implications-of-genetic-admixture-in-hispanic-puerto-ricans-impact-on-the-pharmacogenetics-. Accessed January 16, 2018.

19.

Price MJ, Berger PB, Angiolillo DJ, et al. Evaluation of individualized clopidogrel therapy after drug-eluting stent implantation in patients with high residual platelet reactivity: design and rationale of the GRAVITAS trial. Am Heart J. 2009;157(5):818–824.

20.

Hernandez-Suarez DF, Núñez-Medina H, Scott SA, et al. Effect of cilostazol on platelet reactivity among patients with peripheral artery disease on clopidogrel therapy. Drug Metab Pers Ther. 2018;33(1):49–55.

21.

Price MJ, Endemann S, Gollapudi RR, et al. Prognostic significance of post-clopidogrel platelet reactivity assessed by a point-of-care assay on thrombotic events after drug-eluting stent implantation. Eur Heart J. 2008;29(8):992–1000.

22.

Patti G, Nusca A, Mangiacapra F, Gatto L, D’Ambrosio A, Di Sciascio G. Point-of-care measurement of clopidogrel responsiveness predicts clinical outcome in patients undergoing percutaneous coronary intervention results of the ARMYDA-PRO (Antiplatelet therapy for Reduction of MYocardial Damage during Angioplasty-Platelet Reactivity Predicts Outcome) study. J Am Coll Cardiol. 2008;52(14):1128–1133.

23.

Livak KJ. Allelic discrimination using fluorogenic probes and the 5’ nuclease assay. Genet Anal-Biomol Eng. 1999;14:143–149.

24.

Amigo J, Salas A, Phillips C, Carracedo A. SPSmart: adapting population based SNP genotype databases for fast and comprehensive web access. BMC Bioinformatics. 2008;9:428.

25.

Pendyala LK, Torguson R, Loh JP, et al. Racial disparity with on-treatment platelet reactivity in patients undergoing percutaneous coronary intervention. Am Heart J. 2013;166:266–272.

26.

Mallouk N, Labruyère C, Reny JL, et al. Prevalence of poor biological response to clopidogrel: a systematic review. Thromb Haemost. 2012;107:494–506.

27.

Johnson JA, Roden DM, Lesko LJ, Ashley E, Klein TE, Shuldiner AR. Clopidogrel: a case for indication-specific pharmacogenetics. Clin Pharmacol Ther. 2012;91:774–776.

28.

Bryc K, Velez C, Karafet T, et al. Colloquium paper: genome-wide patterns of population structure and admixture among Hispanic/Latino populations. Proc Natl Acad Sci USA. 2010;107:8954–8961.

29.

Duconge J, Cadilla CL, Renta JY, et al. Prevalence of CYP2C19 gene polymorphisms in the Puerto Rican population: a preliminary report. P R Health Sci J. 2008;27:357–358.

30.

Orengo-Mercado C, Nieves B, López L, et al. Frequencies of functional polymorphisms in three pharmacokinetic genes of clinical interest within the admixed Puerto Rican population. J Pharmacogenomics Pharmacoproteomics. 2013;4(113):1000113.

31.

Fricke-Galindo I, Céspedes-Garro C, Rodrigues-Soares F, et al. Interethnic variation of CYP2C19 alleles, ‘predicted’ phenotypes and ‘measured’ metabolic phenotypes across world populations. Pharmacogenomics J. 2016;16:113–123.

32.

Calderón-Cruz B, Rodríguez-Galván K, Manzo-Francisco LA, et al. C3435T polymorphism of the ABCB1 gene is associated with poor clopidogrel responsiveness in a Mexican population undergoing percutaneous coronary intervention. Thromb Res. 2015;136:894–898.

33.

Lewis JP, Horenstein RB, Ryan K, et al. The functional G143E variant of carboxylesterase 1 is associated with 
increased clopidogrel active metabolite levels and greater clopidogrel response. Pharmacogenet Genomics. 2013;23:1–8.

34.

Lewis JP, Ryan K, O’Connell JR, et al. Genetic variation in PEAR1 is associated with platelet aggregation and cardiovascular outcomes. Circ Cardiovasc Genet. 2013;6:184–192.

35.

Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 polymorphisms and response to clopidogrel. N Engl J Med. 2009;360:354–362.

36.

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;304:1821–1830.

37.

Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med. 2009;360:363–375.

38.

Cuisset T, Morange PE, Alessi MC. Recent advances in the pharmacogenetics of clopidogrel. Hum Genet. 2012;131:653–664.

39.

Gagne JJ, Bykov K, Choudhry NK, Toomey TJ, Connolly JG, Avorn J. Effect of smoking on comparative efficacy of antiplatelet agents: systematic review, meta-analysis, and indirect comparison. BMJ. 2013;347:f5307.

40.

Frelinger AL 3rd, Bhatt DL, Lee RD, et al. Clopidogrel pharmacokinetics and pharmacodynamics vary widely despite exclusion or control of polymorphisms (CYP2C19, ABCB1, PON1), noncompliance, diet, smoking, co-medications (including proton pump inhibitors), and pre-existent variability in platelet function. J Am Coll Cardiol. 2013;61:872–879.

41.

Larsen PD, Johnston LR, Holley A, et al. Prevalence and significance of CYP2C19*2 and CYP2C19*17 alleles in a New Zealand acute coronary syndrome population. Intern Med J. 2015;45:537–545.

42.

Miura G, Ariyoshi N, Sato Y, et al. Genetic and non-genetic factors responsible for antiplatelet effects of clopidogrel in Japanese patients undergoing coronary stent implantation: an algorithm to predict on-clopidogrel platelet reactivity. Thromb Res. 2014;134:877–883.

43.

Janssen PW, Bergmeijer TO, Godschalk TC, et al. The effect of correcting VerifyNow P2Y12 assay results for hematocrit in patients undergoing percutaneous coronary interventions. J Thromb Haemost. 2017;15:618–623.

44.

Bouman HJ, Schömig E, Van Werkum JW, et al. Paraoxonase-1 is a major determinant of clopidogrel efficacy. Nat Med. 2011;17:110–116.

45.

Park KW, Park JJ, Kang J, et al. Paraoxonase 1 gene polymorphism does not affect clopidogrel response variability but is associated with clinical outcome after PCI. PLoS One. 2013;8:e52779.

46.

Mega JL, Close SL, Wiviott SD, et al. PON1 Q192R genetic variant and response to clopidogrel and prasugrel: pharmacokinetics, pharmacodynamics, and a meta-analysis of clinical outcomes. J Thromb Thrombolysis. 2016;41:374–383.

47.

Grove EL, Hvas AM, Kristensen SD. Immature platelets in patients with acute coronary syndromes. Thromb Haemost. 2009;101:151–156.

48.

Angiolillo DJ, Bernardo E, Ramirez C, et al. Insulin therapy is associated with platelet dysfunction in patients with type 2 diabetes mellitus on dual oral antiplatelet treatment. J Am Coll Cardiol. 2006;48:298–304.

49.

Zhang D, Zhang X, Liu D, et al. Association between insulin receptor substrate-1 polymorphisms and high platelet reactivity with clopidogrel therapy in coronary artery disease patients with type 2 diabetes mellitus. Cardiovasc Diabetol. 2016;15:50.

50.

Pérez CM, Soto-Salgado M, Suárez E, Guzmán M, Ortiz AP. High prevalence of diabetes and prediabetes and their coexistence with cardiovascular risk factors in a hispanic community. J Immigr Minor Health. 2015;17:1002–1009.

51.

Behavioral Risk Factor Surveillance System (BRFSS) Database [webpage on the Internet]. Available from: http://www.cdc.gov/brfss/brfssprevalence/index.html. Accessed in October 2016.

52.

Fontana P, Dupont A, Gandrille S, et al. Adenosine diphosphate-induced platelet aggregation is associated with 
P2Y12 gene sequence variations in healthy subjects. Circulation. 2003;108:989–995.

53.

Yang WY, Petit T, Cauwenberghs N, et al. PEAR1 is not a major susceptibility gene for cardiovascular disease in a Flemish population. BMC Med Genet. 2017;18:45.

54.

Nardin M, Verdoia M, Sartori C, et al. Body mass index and platelet reactivity during dual antiplatelet therapy with clopidogrel or ticagrelor. J Cardiovasc Pharmacol. 2015;66:364–370.

55.

Kwok CS, Loke YK. Effects of proton pump inhibitors on platelet function in patients receiving clopidogrel: a systematic review. Drug Saf. 2012;35:127–139.

56.

Olesen JB, Gislason GH, Charlot MG, et al. Calcium-channel blockers do not alter the clinical efficacy of clopidogrel after myocardial infarction: a nationwide cohort study. J Am Coll Cardiol. 2011;57:409–417.

57.

Gurbel PA, Bliden KP, Logan DK, et al. The influence of smoking status on the pharmacokinetics and pharmacodynamics of clopidogrel and prasugrel: the PARADOX study. J Am Coll Cardiol. 2013;62:505–512.

58.

Arméro S, Camoin Jau L, Omar Aït Mokhtar O, et al. Intra-individual variability in clopidogrel responsiveness in coronary artery disease patients under long term therapy. Platelets. 2010;21:503–507.

59.

Ahn SG, Lee SH, Yoon JH, et al. Different prognostic significance of high on-treatment platelet reactivity as assessed by the VerifyNow P2Y12 assay after coronary stenting in patients with and without acute myocardial infarction. JACC Cardiovasc Interv. 2012;5:259–267.

Creative Commons License © 2018 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.