20+ Million Readerbase
Indexed In
  • RefSeek
  • Hamdard University
  • EBSCO A-Z
  • Publons
  • Google Scholar
Share This Page
Recommended Webinars & Conferences

4th Annual Summit on Cardiology and Heart Diseases

Toronto, Canada

2nd International Congress on Heart

Singapore, Singapore
Journal Flyer
Flyer image

Research Article - (2016) Volume 2, Issue 4

Association of the C3435T Multi-Drug Resistance Gene-1 (MDR-1) Polymorphism with Clopidogrel Resistance among Moroccan Acute Coronary Syndromes (ACS) Patients

Hind Hassani Idrissi1*, Wiam Hmimech1, Nada El Khorb2, Hafid Akoudad2, Rachida Habbal3 and Sellama Nadifi1
1Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, Casablanca, Morocco
2Department of Cardiology, University Hospital Center Hassan II, Fes, Morocco
3Department of Cardiology, University Hospital Center Ibn Rochd, Casablanca, Morocco
*Corresponding Author: Hind Hassani Idrissi, Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, Casablanca, Morocco, Tel: 212617786544 Email:

Abstract

Background: Antiplatelet drugs are recommended as one of the main medications used in Acute Coronary Syndromes (ACS) patients. However, an inter-individual variability in platelet response to Clopidogrel has been found in a substantial group of them. Genetic is known be the dominant influencing factor for individual and interethnic variations in drug responses. Polymorphisms in the genes involved in Clopidogrel absorption, biotransformation to the active metabolite, or platelet response to adenosine diphosphate (ADP) have been associated to this impaired response. Interestingly, Multi-Drug Resistance gene-1 (MDR-1) polymorphism influences oral bioavailability of clopidogrel and prognosis of ACS patients. The objectives of our study are, first, to determine the frequency of the C3435T MDR1 polymorphism among Moroccan ACS patients compared to healthy subjects; and second, to assess its effect on Clopidogrel response in a sample of Moroccan ASC patients.
Methods and results: 40 ACS patients were recruited and compared to 99 healthy controls. Extracted DNA samples were genotyped by PCR-RFLP method using MboI restriction enzyme. The VerifyNow assay was used to evaluate platelet function among ACS patients. Our results showed that HTA, Smoking, Creatinine and Sex were statistically associated to Clopidogrel resistance (P=0.05; P=0.05; P=0.05 and P=0.04 respectively). 63.64% of the ST (+) patients were carrying the mutant allele, 54.5% of them having the heterozygous genotype and 36.4% the homozygous mutant one, compared to 9.1% having the homozygous wild type genotype. 62.5% of the resistant group was carrying the mutant allele (50% of them had the TT mutant genotype, 25% CT and 25% CC profiles). Among cases, 42.5% were homozygous mutant TT, 35% CC and 22.5% CT, compared to 39.4% CC, 51.5% CT and 9.1% TT among healthy controls. This polymorphism was positively correlated to ACS risk of development in the CT genotype and the additive transmission model (OR [95% CI]=0.49 [0.16-0.99], P=0.002; OR [95% CI]=2.17 [0.94-2.72], P=0.02), increasing-thus- the association of this polymorphism with the risk of pathology occurrence.
Conclusion: To the best of our knowledge, our study is the first in Morocco to assess the effect of the C3435T MDR1 polymorphism on Clopidogrel response in a sample of Moroccan ACS patients; we also tried to explore the frequency of this polymorphism among Moroccan ACS patients and compare them to healthy individuals. The distribution of the mutant allele in Clopidogrel resistance groups, among ACS sub-types, and also in cases compared to controls and there correlations, suggest a potential association of this variant with Clopidogrel resistance and ACS occurrence risk in our population. Understanding the functional and clinical consequences of this MDR1 variant and others may provide a basis for treating patients more effectively. If this variability in response could be assigned to a mutation in the MDR1 gene, patients could be screened and appropriate dose adjustments could be made on the basis of their MDR1 genotype.

Keywords: ACS, Clopidogrel resistance, C3435T MDR1 polymorphism, Moroccan population

Introduction

Clopidogrel is an oral thienopyridine prodrug, usually prescribed for ACS patients as dual anti-platelet therapy in combination with aspirin. The efficacy of its metabolite active to decrease platelet aggregation by selectively and irreversibly blocking the P2Y12 receptor located on the surface of platelets, make of it the therapy of choice [1]. Once the P2Y12 receptor is inactivated, the association G-protein can no more inhibit adenylyl cyclase, inducing –thus- increased cAMP, and inhibiting the phosphoinositide 3-kinase (PI3K), as a result, the expression of glycoprotein IIb/IIIa subsequently decreases [2].

In the normal case, the glycoprotein IIb/IIIa mediates fibrinogen binding, leading to platelet aggregation and formation of a blood clot (Thrombus). This is what makes the prevention of this pathway by Clopidogrel and other ADP receptors inhibitors very essential in the treatment of thromboembolic events by decreasing platelet aggregation [1].

An inter-individual variability of response to Clopidogrel has been widely described; not all patients reach the same degree of benefit from the given drug, they also still experience thrombotic events even after receiving the treatment [3]. The most part of this heterogeneity in response remains unclear, while some clinical factors that we know may explain just a small fraction of it (Smoking, use of PPIs (Proton Pump Inhibitors), lipophilic statins, calcium channel blockers, high pretreatment platelet reactivity…) [4,5-7]. Genetics may also explain another fraction of this variability in response to Clopidogrel [8,9].

ABCB1 gene

The physiology of drug transport is a key determinant of drug absorption, bioavailability, renal and biliary excretion, and its penetration into brain and other tissues. These specific functions may be modulated by several factors, and importantly by genomic variation [10]. Human ATP-binding cassette (ABC) transporters, especially ABCB1 (ATP-binding cassette, sub-family B, member 1), have been for a long time ago the focus of studies trying to access their effect on mediating drug resistance in a multitude of human pathologies, such as ACS [11]. This protein ensures the absorption of the pro-drug of Clopidogrel from the gastro-intestinal tract into the bloodstream, and transports it back into the intestinal lumen inhibiting thus the absorption [12,13]. Human multidrug resistance 1 (MDR1 ) gene, also called ABCB1, is located on chromosome 7q21.1, and is composed of 28 exons [14,15]. The product of this gene is a protein of 170 KD (1280 amino acids), called P-gp (P-glycoprotein), an energy-dependent efflux pump, involved in extrusion of a big variety of toxins as well as dietary and environmental carcinogens, and drugs [16-20].

Its activity consists of pumping out of cells, numerous compounds, such as drugs, using the energy derived from ATP hydrolysis. This activity has important pharmacokinetic and pharmacodynamic consequences [10]. The P-gp is expressed in several human tissues (gut, liver, kidney) [21]. In the apical membranes of intestinal, renal and hepatic epithelial cells, it acts on absorption and elimination of its substrates; within the apical membranes of capillary endothelial cells of the brain, it acts by limiting the penetration of drugs to the CNS [10]. The MDR1 gene is highly polymorphic and more than 100 polymorphic sites with a minor allele frequency higher than 5% were discovered [22]. One of the most widely described as having an association with intestinal P-gp expression and activity in vivo is the C3435T (rs 1045642) [23]. Localized in exon 26, this polymorphism consists of a C-to-T exchange at cDNA 3435 position of the MDR1 gene; it has no consequences on the amino acid sequence of the P-gp, but affects its function [23].

This synonymous variant of the MDR1 gene alters gene expression, protein activity and stability of its RNAm, and substrate concentration and specificity [24-26]. It results in over-expression of the P-gp, associated to decrease Clopidogrel active metabolite, increased ontreatment platelet reactivity, and increased cardiovascular events [27-29]. Thus, carriers of the homozygous mutant genotype TT have lower intestinal P-gp expression, compared to carriers of the wild type genotypic profile CC [30,31]. The mechanism whereby such synonymous SNP exerts this effect still remain uncertain; it may be a marker for a functional SNP or conceivably be functional; it may also alter translation process [24]. Several studies have explored the association of this polymorphism with ACS risk, among different populations, but have found inconsistent and confusing results. The objectives of our study are, first, to determine the frequency of the C3435T MDR1 polymorphism among Moroccan ACS and healthy subjects; and second, to assess its effect on Clopidogrel response in a sample of Moroccan ASC patients.

Materials and methods

Study population

Recruited patients were those having documented antiplatelet therapy (Clopidogrel), a VerifyNow P2Y12 platelet function test, and no more heparin in their blood. Excluded ones are those having incomplete clinical data or no platelet function test results. A baseline P2Y12 platelet function test was performed to all patients in this study, to evaluate their platelet function state. Blood samples were collected from 40 unrelated ACS Moroccan patients and compared to 99 apparently healthy subjects whose C3435T MDR1 genetic results were previously reported in the study of Kassogue et al. [31], and showing no symptoms of coronary artery diseases. Clinical data concerning risk factors, biological parameters and the Verify-Now test results were collected; an informed consent, approved by the Ethical Committee of the University of Hassan II, School of Medecine, Casablanca, was signed by each patient and control before entering the study. The study protocol we followed was previously described in our paper in press [32,33].

DNA Extraction

Venous blood from all patients was collected in EDTA tubes. Samples were manipulated directly or stored at -20°C until extraction of DNA. Genomic DNA was extracted from blood leukocytes using the standard method of salting out [34-37].

Genotype Determination

We used PCR-RFLP to genotype samples for the C3435T MDR1 polymorphism, as previously described by Tanabe et al. [38]. Genotyping of this variant was performed by amplification from 50 to 100 ng of genomic DNA, followed by digestion using 10 units of MboI restriction enzyme.

The digestion gave rise to three profiles: homozygous wild type CC (Two fragments of 130 and 76 pb), heterozygous TC (three fragments of 206, 130 and 76 bp), and homozygous mutated TT (one fragment of 206 bp). The digested product was separated on 3% agarose gel electrophoresis stained with 0.5 μg/mL ethidium bromide (BET), and visualized with UV rayons.

Statistical Analysis

Statistical analysis was performed using SPSS 21.0 software. Chi square test (χ2) was used to determine statistical significance of association/non-association between genotypes and classical risk factors. Hardy–Weinberg Equilibrium test (HWE) was performed for cases and controls groups. Odds ratio (OR) were calculated to estimate the association between genotypes and ACS risk, using a Confidence Interval (CI) of 95%. Significance was approved at P value less than 0.05.

Results

Characteristics of the study population

The distribution of C3435T-MDR1 polymorphism was in HardyeWeinberg equilibrium (HWE) for controls group but not for cases one (Table 1). The average age was 57.23 ± 8.49 for patients vs. 31.78 ± 12.94 for healthy controls. There was a predominance of male among cases (52.5%) and female among controls (68%) (Table 2). (Table 3) describes the routine pathology data for our SCA patients; 56.75% of these patients were under IPP, when 43.25% were not.

  EHW Cases   EHW Controls  
  X2 square P-value>0.05 X2 square P-value>0.05
C3435T MDR1 6 <0.05 1.3 0.25*

Table 1: Hardey-Weinberg Equilibrium (HWE) for cases and controls groups.

  SCA patients (N=40) Controls (N=100)
Age (years) 57.23 ± 8.49 31.78 ± 12.94
Age of  disease occurrence (years) 54.16 ± 8.31  
Sex    
Male 21 (52.5%) 32 (32%)
Female 19 (47.5%) 68 (68%)
Ethnicity    
Arab 32 (80%) 71 (71%)
Berber 8 (20%) 29 (29%)

Table 2: Description of ACS study population.

Parameters SCA patients
Total Cholesterol (g/l) 1.91 ± 0.67
HDL (g/l) 1.82 ± 6.58
LDL (g/l) 1.34 ± 0.77
Triglycerides TG (g/l) 1.39 ± 0.56
Glucose (g/l) 1.47 ± 0.89
Creatinine (mg/l) 11.9 ± 10.87
Fibrinogene 3.72 ± 1.2
HB (g/dl) 14.15 ± 3.26
GB (elts/mm3) 10397.78 ± 14972.03
Pq (elts/mm3) 235934.57 ± 114017.04
BMI (kg/m²) 26.86 ± 3.7
IPP  
(+) 21 (56.75%)
(-) 16 (43.25%)

Table 3: Routine pathology data of our ACS patients.

Verify now results vs . Risk factors

Patients were placed into resistant and non-resistant groups, based on their platelet function test results, and the baseline characteristics of these patients correlated to resistance groups are shown in (Table 4): HTA, Smoking, Creatinine and Sex were the risk factors that showed statistically significant association with Clopidogrel resistance (P=0.05; P=0.05; P=0.05 and P=0.04 respectively).

  Non Resistants % Resistants % P-value
Age 57 ± 8.48 59.25 ± 8.01 0.4
Sex     0.04*
Male 21 (58.33%) 0 (0%)  
Female 15 (41.67%) 4 (100%)  
SCA TYPE     0.3
ST (+) 9 (25%) 2 (50%)  
ST (-) 27 (75%) 2 (50%)  
familial ACD     NC
presence 0 (0%) 0 (0%)  
abscence 36 (100%) 4 (100%)  
personal ACD     0.5
presence 21 (58.3%) 2 (50%)  
abscence 15 (41.7%) 2 (50%)  
Diabete     0.6
presence 18 (50%) 2 (50%)  
abscence 18 (50%) 2 (50%)  
HTA     0.05*
presence 19 (52.78%) 4 (100%)  
abscence 17 (47.22%) 0 (0%)  
Dyslipidemia     0.7
presence 10 (27.77%) 2 (50%)  
abscence 26 (72.23%) 2 (50%)  
Smoking     0.05*
presence 17 (47.22%) 0 (0%)  
abscence 19 (52.78%) 4 (100%)  
Creatinine (mg/l) 11.41 ± 10.87 19 ± 4.34 0.05*
Fibrinogene 3.59 ± 1.2 5.88 ± 1.21 0.3
Pq 245937.5 ± 194017.04 129236.7 ± 84770.54 0.5
IPP                0.5
used 19 (52.78%) 2 (50%)  
non used 17 (47.22%) 2 (50%)  

Table 4: Baseline characteristics of our SCA patients vs. Verify now test results.

Allelic Frequencies

When correlating the C3435T MDR1 genotypes to the classical risk factors of the pathology, only one statistically significant association was found with personal antecedent, among SCA patients; no association was detected with the other risk factors.

Table 6 shows the distribution of patients ACS type (ST (+) and ST (-)) according to the C3435T MDR1 genotypic profiles: The majority of ST (+) patients were carrying the mutant allele (63.64%) in its heterozygous and mutant forms (54.5% and 36.4%), compared to a small fraction of them (9.1%) that were carrying the wild type allele in its homozygous form. For ST (-) group, equal distribution of the wild type and mutant alleles was found (50% for each one), with 44.8% that were CC or TT, and only 10.3% that were CT.

Distribution of resistant and non-resistant patients according to the C3435T MDR1 genotypes is reported in (Table 7): The most part of the resistant group of patients were carrying the mutant allele (62.5%), 50% were carrying the homozygous mutant genotypic profile, 25% were heterozygous and 25% homozygous wild type. For the nonresistant patients, 41% were homozygous mutant, 22.2% heterozygous and 36.1 % homozygous wild type.

Allelic and genotypic frequencies among cases and controls are reported in (Table 8): 42.5% of the cases were carrying the homozygous mutant genotype TT, when only 9.1% were TT in the controls group. Wild type and heterozygous frequencies were 35% and 22.2% among cases vs. 39.4% and 51.5% among controls respectively. A statistically significant association was found with the heterozygous genotype TC (OR [95% CI]=0.49 [0.16-0.99], P=0.002). There was also a positive correlation with the additive transmission model, but not the dominant and recessive models (OR [95% CI], P=2.17 [0.94-2.72], P=0.02), increasing-thus- the association of this polymorphism with the risk of pathology occurrence.

Discussion

Acute Coronary Syndrome (ACS) in one of the most associated pathologies to a risk of mortality and morbidity, in all its forms; which makes of it a big challenge to clinicians. The treatment of ACS typically includes antithrombotic and antiplatelet agents, in addition to percutaneous coronary or surgical interventions [38,39]. Clopidogrel is currently used in routine combined to aspirin as dual antiplatelet therapy, and has shown important efficacy [40-44]. Despite all its benefits, a significant inter-individual variability in response to Clopidogrel has been widely described. As consequences to this variability in response to treatment, some patients show nonresponsiveness or decreased inhibition of platelet aggregation after being treated by Clopidogrel, which has been associated with increased risk of cardiovascular events [45,46]. One of the first causes of this variability lies in the pharmacokinetics of Clopidogrel. This prodrug needs to be –first- transported by the P-gp protein (ABCB1 gene product), and metabolized –after- by the Cyp 450 system in the liver; the active metabolite then irreversibly inhibits the P2Y12 , the direct receptor of Clopidogrel [47]. Genetic variations in these genes (ABCB1, CYP3A4, CYP2B6, P2Y12 ) have been associated with this inter-individual variability in response to Clopidogrel, and may – thusexplain an important fraction of it [39,48].

Interestingly, Multi-Drug Resistance gene-1 (MDR-1) polymorphism has been found to influence oral bioavailability of clopidogrel and prognosis of ACS patients. Taking these backgrounds into account, the purpose of the present study – as being the first to do it in Morocco- was to determine the frequency of C3435T MDR1 polymorphism among Moroccan ACS patients; and to evaluate the correlation between Clopidogrel resistance and genetic testing represented by the C3435T MDR1 polymorphism, in a sample of Moroccan ACS patients.

Our study population was for the most part of it composed of male (52.5%), when a predominance of female was noted among healthy controls (68%) (Table 2). Similar distribution was reported in the study of Ayaz et al. [48], investigating the association of MDR1 C3435T and G2677T/A polymorphisms with plasma platelet-activating factor levels and coronary artery disease risk in Turkish population. The average age was 57.23 ± 8.49 and 31.78 ± 12.94 in cases and controls groups respectively.

56.75% of our patients were under PPIs vs . 43.25% that were not (Table 3). Proton-pump inhibitors (PPI) are known to potentially affect the Clopidogrel platelet inhibition relationship [49,50]. Several studies have reported an inhibitory effect of PPIs (especially omeprazole) on the antiplatelet efficacy of Clopidogrel [51-55]; when others did not report any interaction between PPIs and Clopidogrel [56-58]. In 2010, The Committee on Human Medicinal Products (CHMP) has reported that the concomitant use of clopidogrel and omeprazole or esomeprazole in clinical practice was discouraged, and that, in patients under PPIs, use of pantoprazole in place of omeprazole or lansoprazole is recommended; this come from the fact that pantoprazole does not appear to have the significant inhibitory effect on the cytochrome enzyme CYP2C19 that other PPIs such us omeprazole have [59].

Our patients were placed into resistants and non-resistants, according to their platelet function test results, and were then correlated to the baseline characteristics of the studied population (Table 4). HTA, Smoking, Creatinine and Sex were the risk factors that showed statistically significant association with Clopidogrel resistance (P=0.05; P=0.05; P=0.05 and P=0.04 respectively). 58.33% of the nonresistant patients were male, when the whole resistant group was female (Table 4). Jun-Feng et al. have found similar results concerning the predominance of female among resistant and male among nonresistant patients, and the positive correlation of HTA and Creatinine with Clopidogrel resistance, but not for the rest of risk factors [60]. Other studies report a as well a significant association of smoking with clopidogrel resistance [61,62]. Several studies have suggested that women do not accrue equal therapeutic benefit of antithrombotic therapy [63,64]. Although multiple contributing factors have been described (Differences in vessel wall biology between men and women; the direct influence of sex hormones (oestrogens, progesterone or androgens) on platelets and their indirect effect on the vasculature), the physiological mechanism behind this gender disparity remains unclear [65].

A statistical comparison was held between distribution of C3435T MDR1 polymorphism among ACS patients and traditional risk factors; we found a statistically significant association with personal antecedent (P=0.02); no other significant association was found with the other risk factors (Table 5). Correlation between this polymorphism and ACS sub-groups (ST+ and ST-) showed that the majority of ST(+) patients were carrying the mutant allele (63.64%) in its heterozygous and mutant forms (54.5% CT and 36.4% TT), when only a small fraction of them was carrying the wild type allele in its homozygous form (9.1% CC). ST (-) group showed equal distribution of the wild type and mutant alleles (50% for each one), with 44.8% of ST (-) patients having the CC or TT genotypes, and only 10.3% that were CT. This distribution showed statistically signification association of the C3435T polymorphism with the ACS sub-types (P=0.007) (Table 6).

Risk factor   SCA patients   P-value<0.05
  N % Hz % muté %  
Familial ACD       NC
presence 0 0 0  
absence 14 (35%) 9 (22.5%) 17 (42.5%)  
personal ACD       0.02*
presence 9 (39.1%) 8 (34.8%) 6 (26.1%)  
absence 5 (29.4%) 1 (5.9%) 11 (64.7%)  
HTA       0.2
presence 9 (39.1%) 3 (13.04%) 11 (47.8%)  
absence 5 (29.4%) 6 (35.3%) 6 (35.3%)  
Smoking       0.2
presence 5 (29.4%) 6 (35.3%) 6 (35.3%)  
absence 9 (39.16%) 3 (13.04%) 11 (47.8%)  
Diabetes       0.6
presence 6 (30%) 4 (20%) 10 (50%)  
absence 8 (40%) 5 (25%) 7 (35%)  
Dyslipidaemia       0.3
presence 3 (30%) 1 (10%) 6 (60%)  
absence 11 (36.7%) 8 (26.7%) 11 (36.7%)  

Table 5: C3435T MDR1 genotypes distribution vs. risk factors.

    SCA patients       P-value<0.05
  N % Hz % mutant% wild type allele mutant allele 0.007*
ST (+) 1 (9.1%) 6 (54.5%) 4 (36.4%) 36.36% 63.64%  
ST (-) 13 (44.8%) 3 (10.3%) 13 (44.8%) 50% 50%  

Table 6: C3435T MDR1 genotypes distribution vs. ACS sub-groups.

Distribution of resistant and non-resistant patients according to the C3435T MDR1 genotypes showed that the majority of the resistant group were carrying the mutant allele (62.5%), 50% were carrying the homozygous mutant form TT, 25% were heterozygous CT and 25% homozygous wild type CC. For the non-resistant group, 41% were homozygous mutant TT, 22.2% heterozygous CT and 36.1 % homozygous wild type CC (Table 7). Allelic and genotypic frequencies among cases and controls reported in (Table 8) show that: 42.5% of the cases were carrying the homozygous mutant genotype TT, when only 9.1% were TT in the controls group. A statistically significant association was found with the heterozygous genotype TC (OR [95%CI], P=0.49 [0.16-0.99], P=0.002). There was also a positive correlation with the additive transmission model, but not the dominant and recessive ones (OR [95%CI], P=2.17 [0.94-2.72], P=0.02), increasing-thus- the association of this polymorphism with the risk of pathology occurrence. The study performed by Jaitner et al. trying to assess the association of ABCB1 C3435T genotypes with the antiplatelet efficacy of clopidogrel and the risk of stent thrombosis (ST) in large cohorts of clopidogrel-treated patients undergoing percutaneous coronary intervention, reported that the C3435T MDR1 polymorphism did not influence the risk of ST or the response to Clopidogrel in Clopidogrel treated patients [66]. Association between C3435T MDR1 and Clopidogrel resistance is not as well established, and studies have reported inconsistent and conflicting results. Spiewak et al. have found a significant correlation between this polymorphism and Clopidogrel resistance and the availability of its active metabolite [67]. This may be explained by the decreased intestinal absorption of Clopidogrel in patients carrying the TT mutant genotype, compared to those carrying the CC wild type one. Several other studies reported similar findings, when others reported contracting results. Another study performed by Wallentin et al. has found that the ex-vivo addition of the active metabolite of Clopidogrel can overcome its resistance, and induce higher platelet inhibition regardless of the initial platelet reactivity after 600 mg loading dose and 75 mg maintenance dose of Clopidogrel. This suggests that variability in antiplatelet response to clopidogrel depends more on plasma levels of its active metabolite (that can be affected by ABCB1 and Cytochrome P450 genetic variations) than on the platelet receptor properties [67,68].

    SCA patients P-value<0.05
  N % Hz % mutant % wild type allele mutant allele 0.5
Non-Resistant 13 (36.1%) 8 (22.2%) 15 (41.7%) 47.2% 52.8%  
Resistant 1 (25%) 1 (25%) 2 (50%) 37.5% 62.5%  

Table 7: C3435T MDR1 genotypes distribution vs verify now test results.

  Genotypes/alleles Cases (%) Controls (%) OR (95% CI) P-value
C3435T MDR1 CC 14 (35%) 39 (39.4%) 1  
  CT 9 (22.5%) 51 (51.5%) 0.49 [0.16-0.99] 0.002*
  TT 17 (42.5%) 9 (9.1%) 5.26 [1.28-9.87] 0.5
           
  CC+CT (b) 23 (57.5%) 90 (90.9%) 1  
  TT 17 (42.5%) 9 (9.1%) 7.39 [2.09-13.74] 0.5
           
  CC (c) 14 (35%) 39 (39.4%) 1  
  CT+TT 26 (65%) 60 (61.6%) 1.21 [0.42-1.85] 0.5
           
  C (d) 37 (46.25%) 129 (65.85%) 1  
  T 43 (53.75%) 69 (34.15%) 2.17 [0.94-2.72] 0.002*

Table 8: Allelic and genotypic frequencies of C3435T MDR1 polymorphism among cases and controls.

Conclusion

As causing no change on the protein sequence and being located in a non-regulatory region of the MDR1 gene, it may be conceivable that the C3435T polymorphism is not causative for differences in P-gp expression. It may be linked to other changes in regions that control expression of the MDR1 gene (the promoter/enhancer region) or regions that are relevant for mRNA processing. Nevertheless, this SNP appears to allow the differentiation of alleles with distinct MDR-1 expression and activity. Therefore, understanding the functional and clinical consequences of this MDR1 variant and others may provide a basis for treating patients more effectively. If this variability in response could be assigned to a mutation in the MDR1 gene, patients could be screened and appropriate dose adjustments could be made on the basis of their MDR1 genotype. Since we found significant associations between some patient characteristics and Clopidogrel resistance, these characteristics should be considered prior to the administration of Clopidogrel. This would help distinguish patients that are likely to respond to the drug from those that would likely be resistant. While this study is promising, it is partly limited by the small sample size, and should be followed up by multi-center clinical trials with a greater number of patients in order to confirm and verify our findings.

References

  1. Perry CG, Shuldiner AR (2013) Pharmacogenomics of Anti-platelet Therapy: How much evidence is enough for clinical implementation?. J Hum Genet 58: 339–345.
  2. Cattaneo M (2011) The platelet P2Y12 receptor for adenosine diphosphate: congenital and druginduced defects. Blood 117: 2102-2112.
  3. Gurbel PA, Bliden KP, Hiatt BL, O’Connor CM (2003) Clopidogrel for coronary stenting: response variability, drug resistance, and the effect of pretreatment platelet reactivity. Circulation 107: 2908-2913.
  4. Marín F, González-Conejero R, Capranzano P, Bass TA, Roldán V, et al. (2009) Pharmacogenetics in Cardiovascular Antithrombotic Therapy. J Am Coll Cardiol 54: 1041-1057.
  5. Gilard M, Arnaud B, Le Gal G, Abgrall JF, Boschat J (2006) Influence of omeprazol on the antiplatelet action of clopidogrel associated to aspirin. J Thromb Haemost 4: 2508-2509.
  6. Lau WC, Gurbel PA, Watkins PB, Neer CJ, Hopp AS, et al. (2004) Contribution of hepatic cytochrome P450 3A4 metabolic activity to the phenomenon of clopidogrel resistance. Circulation 109: 166–171.
  7. Siller-Matula JM, Lang I, Christ G, Jilma B (2008) Calcium-channel blockers reduce the antiplatelet effect of clopidogrel. J Am Coll Cardiol 52: 1557-1563.
  8. Cayla G, Hulot JS, O'Connor SA, Pathak A, Scott SA, et al. (2011) Clinical, angiographic, and genetic factors associated with early coronary stent thrombosis. JAMA 306: 1765-1774.
  9. Evans WE, McLeod HL (2003) Pharmacogenomics-drug disposition, drug targets, and side effects. N Engl J Med 348: 538-549.
  10. Roden DM (2013) Cardiovascular Pharmacogenomics: The Future of Cardiovascular Therapeutics? Can J Cardiol 29: 58-66.
  11. Heestermans AA, van Werkum JW, Taubert D, Seesing TH, von Beckerath N, et al. (2008) Impaired bioavailability of clopidogrel in patients with a ST-segment elevation myocardial infarction. Thromb Res 122: 776-781.
  12. Mega JL, Close SL, Wiviott SD, Shen L, Walker JR, et al. (2010) Genetic variants in ABCB1 and CYP2C19 and cardiovascular outcomes after treatment with clopidogrel and prasugrel in the TRITON-TIMI 38 trial: a pharmacogenetic analysis. Lancet 376: 1312-1319.
  13. Fisch AS, Perry CG, Stephens SH, Horenstein RB, Shuldiner AR (2013) Pharmacogenomics of Anti-platelet and Anti-coagulation Therapy. Current Cardiology Reports 15: 381.
  14. Efferth T (2001) The human ATP-binding cassette transporter genes: from the bench to the bedside. Curr Mol Med 1: 45–65.
  15. Burger H, Foekens JA, Look MP, Meijer-van GME, Klijn JG, et al. (2003) RNA expression of breast cancer resistance protein, lung resistance-related protein, multidrug resistance-associated proteins 1 and 2, and multidrug resistance gene 1 in breast cancer: correlation with chemotherapeutic response. Clin Cancer Res 9: 827–836.
  16. Lin JH (2003) Drug-drug interaction mediated by inhibition and induction of P-glycoprotein. Adv Drug Deliv Rev 55: 53-81.
  17. Almond LM, Edirisinghe D, Dalton M, Bonington A, Back DJ, et al. (2005) Intracellular and plasma pharmacokinetics of nevirapine in human immunodeficiency virus-infected individuals. Clin Pharmacol Ther 78: 132-142.
  18. Ambudkar SV, Kimchi-Sarfaty C, Sauna ZE, Gottesman MM (2003) P-glycoprotein: from genomics to mechanism. Oncogene 22: 7468-7485.
  19. Kerb R (2006) Implications of genetic polymorphisms in drug transporters for pharmacotherapy. Cancer Lett 234: 4-33.
  20. Ashariati A (2008) Polymorphism C3435T of the MDR-1 gene predicts response to preoperative chemotherapy in locally advanced breast cancer with her2/neu expression. Acta Med Indones 44: 187-191.
  21. Leslie EM, Deeley RG, Cole SP (2005) Multidrug resistance proteins: role of P-glycoprotein, MRP1, MRP2, and BCRP (ABCG2) in tissue defense. Toxicol Appl Pharmacol 204: 216–237.
  22. Fromm MF (2002) Genetically determined differences in P-glycoprotein function: implications for disease risk. Toxicology 181-182: 299-303.
  23. Hoffmeyer S, Burk O, von Richter O, Arnold HP, Brockmöller J, et al. (2000) Functional polymorphisms of the human multidrug resistance gene: Multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci U S A 97: 3473–3478.
  24. Kimchi-Sarfaty C, Oh JM, Kim IW, Sauna ZE, Calcagno AM, et al. (2007) A ‘silent’ polymorphism in the MDR1 gene changes substrate specificity. Science 315: 525-528.
  25. Simon T, Verstuyft C, Mary-Krause M, Quteineh L, Drouet E, et al. (2009) Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med 360: 363-375.
  26. Hemauer SJ, Nanovskaya TN, Abdel-Rahman SZ, Patrikeeva SL, Hankins GD, et al. (2010) Modulation of human placental P-glycoprotein expression and activity by MDR1 gene polymorphisms. Biochem Pharmacol 79: 921-925.
  27. Taubert D, von Beckerath N, Grimberg G, Lazar A, Jung N, et al. (2006) Impact of P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther 80: 486–501.
  28. Wang XD, Zhang DF, Liu XB, Lai Y, Qi WG, et al. (2012) Modified clopidogrel loading dose according to platelet reactivity monitoring in patients carrying ABCB1 variant alleles in patients with clopidogrel resistance. Eur J Intern Med 23: 48-53.
  29. Su J, Xu J, Li X, Zhang H, Hu J, et al. (2012) ABCB1 C3435T Polymorphism and Response to Clopidogrel Treatment in Coronary Artery Disease (CAD) Patients: A Meta-Analysis. PLoS One 7: e46366.
  30. Sills GJ, Mohanraj R, Butler E, McCrindle S, Collier L, et al. (2005) Lack of association between the C3435T polymorphism in the human multidrug resistance (MDR1) gene and response to antiepileptic drug treatment. Epilepsia 46: 643-647.
  31. Kassogue Y, Dehbi H, Nassereddine S, Quachouh M, Nadifi S (2013) Genotype variability and haplotype frequency of MDR1 (ABCB1) gene polymorphism in Morocco. DNA and cell biology 32: 582-588.
  32. Idrissi HH, Hmimech W, el Khorb N, Akoudad H, Habbal R, et al. Does i-T744C P2Y12 polymorphism modulate Clopidogrel response among Moroccan Acute Coronary Syndromes (ACS) patients? Genetics International Journal. Hindawi Publications. Paper in press.
  33. Malinin A, Pokov A, Spergling M, Defranco A, Schwartz K, et al. (2007) Monitoring platelet inhibition after Clopidogrel with the VerifyNow-P2Y12 (R) rapid analyzer: the VERIfy Thrombosis risk Assessment (VERITAS) study. Thromb Res 119: 277-284.
  34. Prabhakaran S, Wells KR, Lee VH, Flaherty CA, Lopes DK (2008) Prevalence and risk factors for aspirin and Clopidogrel resistance in cerebrovascular stenting. AJNR Am J Neuroradiol 29: 281-285.
  35. Gasparyan AY (2010) Aspirin and Clopidogrel resistance: methodological challenges and opportunities. Vasc Health Risk Manag 6: 109–112.
  36. Miller SA, Dykes DD, Polesky HF (1988) A simple salting out procedure for extracting DNA from human nucleated cells.  Nucleic Acids Res 16: 1215.
  37. Tanabe M, Leiri I, Nagata N, Inoune K, Ito S, et al. (2001) Expression of P-gly6coprotein in human placenta: relation to genetic poly-morphism of the multifrug resistance (MDR)-1 gene. J Pharmacol Exp Ther 297: 1137-1143.
  38. Showkathali R, Natarajan A (2012) Antiplatelet and Antithrombin Strategies in Acute Coronary Syndrome: State-Of-The-Art Review. Curr Cardiol Rev 8: 239-249.
  39. Yusuf S, Zhao F, Mehta S R, Chrolavicius S, Tognoni G, et al. (2001) Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med 345: 494-502.
  40. Mehta SR, Yusuf S, Peters RJ, Bertrand ME, Lewis BS, et al. (2001) Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCI-CURE study. Lancet 358: 527-533.
  41. Steinhubl SR, Berger PB, Mann JT, Fry ET, DeLago A, et al. (2002) Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. JAMA 288: 2411-2420.
  42. Chen ZM, Jiang LX, Chen YP, Xie JX, Pan HC, et al. (2005) Addition of clopidogrel to aspirin in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 366: 1607-1621.
  43. Sabatine MS, Cannon CP, Gibson CM, López-Sendón JL, Montalescot G, et al. (2005) Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with ST-segment elevation. N Engl J Med 352: 1179-1189.
  44. Gurbel PA, Becker RC, Mann KG, Steinhubl SR, Michelson AD (2007) Platelet function monitoring in patients with coronary artery disease. J Am Coll Cardiol 50: 1822-1834.
  45. Price MJ (2009) Bedside evaluation of thienopyridine antiplatelet therapy. Circulation 119: 2625-2632.
  46. Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, Ramírez C, Cavallari U, et al. (2005) Variability in platelet aggregation following sustained aspirin and clopidogrel treatment in patients with coronary heart disease and influence of the 807 C/T polymorphism of the glycoprotein Ia gene. Am J Cardiol 96: 1095-1099.
  47. Aventis S, Bristol-Myers S (2009) Plavix (clopidogrel) package insert. Bridgewater, NJ.
  48. Ayaz G, Batar B, Kanigur G, Guven M, Onaran I, et al. The association of MDR1 C3435T and G2677T/A polymorphisms with plasma platelet-activating factor levels and coronary artery disease risk in Turkish population. Gene 527: 301–305.
  49. Nordeen JD, Patel AV, Darracott RM, Johns GS, Taussky P, et al. (2013) Clopidogrel Resistance by P2Y12 Platelet Function Testing in Patients Undergoing Neuroendovascular Procedures: Incidence of Ischemic and Hemorrhagic Complications J Vasc Interv Neurol 6: 26-34.
  50. Gilard M, Arnaud B, Cornily JC, Le Gal G, Lacut K, et al. (2008) Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole CLopidogrel Aspirin) study. J Am Coll Cardiol 51: 256-260.
  51. Ho PM, Maddox TM, Wang L, Fihn SD, Jesse RL, et al. (2009) Risk of adverse outcomes associated with concomitant use of clopidogrel and proton pump inhibitors following acute coronary syndrome. JAMA 301: 937-944.
  52. Juurlink DN, Gomes T, Ko DT, Szmitko PE, Austin PC, et al. (2009) A population-based study of the drug interaction between proton pump inhibitors and clopidogrel. CMAJ 180: 713-718.
  53. Sibbing D, Morath T, Stegherr J, Braun S, Vogt W, et al. (2009) Impact of proton pump inhibitors on the antiplatelet effects of clopidogrel. Thromb Haemost 101: 714-719.
  54. Small DS, Farid NA, Payne CD, Weerakkody GJ, Li YG, et al. (2008) Effects of the proton pump inhibitor lansoprazole on the pharmacokinetics and pharmacodynamics of prasugrel and clopidogrel. J Clin Pharmacol 48: 475-484.
  55. Depta JP, Bhatt DL (2010) Omeprazole and clopidogrel: Should clinicians be worried? Cleve Clin J Med 77: 113-116.
  56. O'Donoghue ML, Braunwald E, Antman EM, Murphy SA, Bates ER, et al. (2009) Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomized trials. Lancet 374: 989-997.
  57. Bhatt DL, Cryer BL, Contant CF, Cohen M, Lanas A, et al. (2010) Clopidogrel with or without omeprazole in coronary artery disease. N Engl J Med 363: 1909-1917.
  58. (2010) Interaction between clopidogrel and proton-pump inhibitors. CHMP updates warning for clopidogrel-containing medicines.
  59. Su JF, Hu XH2, Li CY (2015) Risk factors for clopidogrel resistance in patients with ischemic cerebral infarction and the correlation with ABCB1 gene rs1045642 polymorphism. Exp Ther Med 9: 267-271.
  60. Bliden KP, Dichiara J, Lawal L, Singla A, Antonino MJ, et al. (2008) The association of cigarette smoking with enhanced platelet inhibition by clopidogrel. J Am Coll Cardiol 52: 531-533.
  61. Gurbel PA, Nolin TD, Tantry US (2012) Clopidogrel efficacy and cigarette smoking status. JAMA 307: 2495-2496.
  62. Berger JS, Roncaglioni MC, Avanzini F, Pangrazzi I, Tognoni G, et al. (2006) Aspirin for the primary prevention of cardiovascular events in women and men: a sex-specific meta-analysis of randomized controlled trials. JAMA 295: 306-313.
  63. Boersma E, Harrington RA, Moliterno DJ, White H, Théroux P, et al. (2002) Platelet glycoprotein IIb/IIIa inhibitors in acute coronary syndromes: a meta-analysis of all major randomised clinical trials. Lancet 359: 189-198.
  64. Jochmann N, Stangl K, Garbe E, Baumann G, Stangl V, et al. (2005) Female-specific aspects in the pharmacotherapy of chronic cardiovascular diseases. Eur Heart J 26: 1585-1595.
  65. Jaitner J, Morath T, Byrne RA, Braun S, Gebhard D et al. No Association Of Abcb1 C3435t Genotype With Clopidogrel Response Or Risk Of Stent Thrombosis In Patients Undergoing Coronary Stenting. Circ Cardiovasc Interv 5: 82-88.
  66. Spiewak M, Malek LA, Kostrzewa G, Kisiel B, Serafin A, et al. (2009) Influence of C3435T multidrug resistance gene-1 (MDR-1) polymorphism on platelet reactivity and prognosis in patients with acute coronary syndromes. Kardiol Pol 67: 827-834.
  67. Wallentin L, James S, Storey RF, Armstrong M, Barratt BJ, et al. (2010) Effect of CYP2C19 and ABCB1 single nucleotide polymorphisms on outcomes of treatment with ticagrelor versusclopidogrel for acute coronary syndromes: a genetic substudy of the PLATO trial. Lancet 376: 1320-1328.
  68. Wallentin L, Varenhorst C, James S, Erlinge D, Braun OO, et al. (2008) Prasugrel achieves greater and faster P2Y12receptor-mediated platelet inhibition than clopidogrel due to more efficient generation of its active metabolite in aspirin-treated patients with coronary artery disease. Eur Heart J 29: 21-30.
Citation: Idrissi HH, Hmimech W, El Khorb N, Akoudad H, Habbal R, et al. (2016) Association of the C3435T Multi-Drug Resistance Gene-1 (MDR-1) Polymorphism with Clopidogrel Resistance among Moroccan Acute Coronary Syndromes (ACS) Patients. J Thrombo Cir 2:115.

Copyright: © 2016 Idrissi HH, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
bellicon