Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-06-08T04:37:13.663Z Has data issue: false hasContentIssue false

Genetic Analysis of CYP2C9 with Reference to Drug Response in Epilepsy Patients of Pakistan

Published online by Cambridge University Press:  01 January 2024

Hafsa Maqbool
Affiliation:
Institute of Zoology, University of the Punjab Lahore, Lahore, Pakistan
Tayyaba Saleem
Affiliation:
Institute of Zoology, University of the Punjab Lahore, Lahore, Pakistan Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
Nadeem Sheikh*
Affiliation:
Institute of Zoology, University of the Punjab Lahore, Lahore, Pakistan
Aqsa Ashfaq
Affiliation:
Institute of Zoology, University of the Punjab Lahore, Lahore, Pakistan
*
Correspondence should be addressed to Nadeem Sheikh; nadeem.zool@pu.edu.pk

Abstract

Epilepsy is a major global issue. Epilepsy patients are treated with AED (antiepileptic drugs). Interindividual variability in drug response has been documented in several studies. The resistance to drug response may be attributed to genetic polymorphism. The current study was undertaken to investigate the CYP2C9 gene polymorphism associated with antiepileptic drug (AED) resistance in the Pakistani population. The current study included 337 individuals including 100 control subjects, 110 drug-resistant subjects, and 127 drug responders. Genomic DNA was isolated from blood, and amplification of rs1799853 (430C > T) and rs1057910 was carried out by polymerase chain reaction. Genotypes of CYP2C9 SNPs were determined by Sanger’s sequencing. Astounding results were observed in the current study that none of the well-known reported SNPs of CYP2C9 was found in our Pakistani cohorts. However, a novel missense variant (c.374G > A) was found only in drug-resistant patients of the current study. According to the in silico analysis performed by PolyPhen-2, it was observed that this nonsynonymous substitution is likely to be pathogenic. The results of our study demonstrated that rs1799853 and rs1057910 may be involved in drug resistance in the Pakistani population. However, some other variants on CYP2C9 may play a critical role in AED resistance that needs to be explored.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © 2022 Hafsa Maqbool et al.

1. Introduction

Epilepsy is one of the highest incidence rate CNS illnesses, characterized by repeated seizures with a variety of symptomatology [Reference Manford1, Reference Udani2]. Uncontrolled epileptic seizures reduce patients’ quality of life by raising the risk of physical injury and having a detrimental impact on their physical and psychological well-being. Although the possible risk factors are well understood, it is still unclear why the efficacy of antiepileptic medicine administered to two people with the same kind of epilepsy or seizures may be so drastically different [Reference Kozera-Kępniak, Jastrzębski and Klimek3]. Genetic variables that alter the pharmacodynamic and pharmacokinetic characteristics of given medications are among the causes being considered. Currently, the thought under consideration by many scientists is that investigation of the mechanisms underlying drug resistance may lead to the development of novel treatment techniques. Recent research has increasingly shown that genetic variables, such as polymorphisms in the genes of microsomal enzymes involved in drug metabolism (CYP), have a role in the development of drug resistance in epilepsy [Reference Lazarowski and Czornyj4Reference Ke, Cheng, Yu and Di7]. The P450 cytochrome enzyme family is involved in the metabolism of the majority of medicines. The CYP2 family is in charge of the initial stage of exogenous particle metabolism. Many enzymes encoded by polymorphic genes are members of the CYP2 family. In terms of clinical significance, the enzymes CYP2C9, CYP2C19, and CYP2D6 are the most significant. CYP genetic variations were found in many cultures throughout the world, and the variations are responsible for the particular activity of drug-metabolizing enzymes [Reference Lin and Lu8]. Drugs, including antiepileptic medications, are biotransformed and eliminated by oxidation processes catalyzed by P450 cytochrome enzymes, as well as glucuronidation, in which CYP2C9 and CYP2C19 play a key role. The genetic polymorphisms of CYP2C9 and CYP2C19 impact the rate of drug metabolism, resulting in variable levels of sensitivity to prescribed therapeutic doses [Reference Pierzchała9]. So far, 13 alleles of the CYP2C9 genes have been found, with CYP2C9∗2 and CYP2C9∗3 resulting from single-amino-acid substitutions in the CYP29C∗1 coding region, R144C, and I359L, respectively. It is linked to a drop in the phenytoin-metabolizing enzyme’s activity [Reference Brandolese, Scordo, Spina, Gusella and Padrini10]. The CYP2C9 polymorphism encompasses the presence of the three alleles CYP2C9∗1, CYP2C9∗2, and CYP2C9∗3, which are all characteristic of the Caucasian population. The prevalence frequencies of the ∗2 (Arg144Cys) and ∗3 (Ile359Leu) alleles in the Caucasian population are 8–12 percent and 3–8 percent, respectively. These are the alleles that cause a significant drop in isoenzyme activity. The proportion of ∗2 and ∗3 alleles is significantly lower in oriental and Afro-American groups, whereas various additional variants of the CYP2C9 isoenzyme producing gene (∗4, ∗5, ∗6) have been identified as being distinctive of these people [Reference Scordo, Aklillu, Yasar, Dahl, Spina and Ingelman-Sundberg11]. Many medicines, including nonsteroidal anti-inflammatory medicines, antiepileptic medications, angiotensin II receptor antagonists, and antidiabetic and antithrombotic medicines, are biotransformed by the CYP2C9 isoenzyme. The genotype of a patient may aid in the selection and administration of antiepileptic medications. Literature evidence suggests a link between the CYP2C9 polymorphism and drug-resistant epilepsy; however, this link appears to be skewed by ethnicity [Reference Franco and Perucca12Reference Chan, Sellors, Chiew and Buckley15]. The goal of the study was to identify a relationship between the SNP rs1799853 and rs1057910 of CYP2C9 gene in the Pakistani cohort.

2. Materials and Methods

2.1. Subjects

This study was approved by the Bioethics Committee of the Institute of Zoology University of Punjab, Lahore, Pakistan (Bioethics/133). Written informed consent was taken from every patient/guardian. All patients were recruited through the hospitals of Punjab, with standard, established clinical epilepsy diagnosis. Patients were eligible if they had drug-resistant or drug-responsive epilepsy, as described by the ILAE (International League Against Epilepsy) criteria and had been taking an AED for at least a year [Reference Kwan, Arzimanoglou and Berg16]. The patients were excluded if they have substantial psychiatric comorbidity, uncertain record of seizure frequency, experienced pseudoseizures, inconsistent AED therapy, drug addiction, and occurrence of neurodegenerative disorders. The study included 337subjects of which 110 were drug resistant, 127 were drug-responsive pediatric patients, and 100 were normal healthy individuals as a control group. Drug resistance was defined as consistent seizure frequency despite treatment with the maximum tolerance dose of two established AED. Drug responsiveness was defined as complete freedom from seizures for at least two years after treatment with AED. The participants of the study (patients/controls) were recruited from the same facility, shared similar ethnicity, and were unrelated. A structured questionnaire was used to collect demographic information as well as information on seizure forms and frequency, past medical history, AED history, and related family history. For DNA extraction and genotyping, a 3cc venous blood sample was taken. To ensure blind genotyping, a code was used for the identification of subject information and genotype data.

2.2. Genotyping

The genomic DNA was isolated from peripheral blood lymphocytes by the modified organic extraction method [Reference Green and Sambrook17] and diluted to a final concentration of 2 0 ng/μL with diethyl pyrocarbonate (DEPC) water. The selected SNPs were amplified using previously reported primers. The rs1799853 was amplified using CACTGGCTGAAAGAGCTAACAGAG as the forward primer and AGGAAGAGATTGAACGTGTGA as the reverse primer, while for rs1057910, GTGGGTGACCCTACTCCATATCAC was used as the forward primer and TTGACCTTCTCCCCACCAGCCTGC as the reverse primer [Reference Al-Mohizea, Alkharfy and Bagulb18]. The annealing temperature for rs1799853 and rs1057910 was 65°C and 60°C, respectively. Thermal cycling conditions were 5 min of initial denaturation at 95°C followed by 35 cycles of denaturation at 95°C for 45 s, annealing for 45 s at the abovementioned temperatures for each SNP, 45 s of initial extension at 72°C, and final extension at 72°C for 10 min. The confirmation of PCR products was made by resolving amplicon of 375 bp (rs1799853) and 130 bp (rs1057910) on 2% agarose gel. Genotyping was performed from a commercial source (1st BASE, Singapore).

2.3. In Silico Analysis

The sequences were visualized using BioEdit 7.0 software. BLAST was performed to compare all the sequences with the reference sequence (Homo sapiens Taxid 9606). Wild and mutant DNA sequences were translated and aligned by MEGA X software. Prediction of the clinical significance of the identified variants was made by PolyPhen-2software. Changes in the protein stability consequent to the identified mutations were accessed using mCSM software, and functional protein association analysis was performed by the online available tool STRING.

3. Results

The mean age of the control group including 68 males and 32 females was 75 ± 21.89 months. The demographic attributes and seizure type for both groups, i.e., drug resistant and drug responsive, are presented in Table 1. None of the SNPs deviated from Hardy–Weinberg equilibrium in control subjects. We did not find any of the targeted mutations either in drug-responsive or drug-resistant epilepsy patients. We identified a novel missense variant at the c.374G > A position in CYP2C9 as shown in Figure 1(b). Genotype count for the identified SNP is given in Table 2.

TABLE 1: Clinical and demographic attributes of drug-resistant and drug-responsive subjects.

FIGURE 1: (a) The pathogenicity score for c.374G > A, (b) electropherogram of wild and heterozygous variant c.374G > A, (c) interaction of CYP2C9 with other functional nodes, and (d) predicted transmembrane topology with the highlighted variant position.

TABLE 2: CYP2C9 variant identified by sequencing.

3.1. In Silico Analysis

The alignment of mutated and reference sequence indicated a change in amino acid from arginine to histidine at the 125 position in CYP2C9 protein. The pathogenicity score for c.374G > A was 0.998, indicating this substitution to be likely pathogenic as presented in Figure 1(a). The position of the detected variant in polypeptide was determined by mCSM (Figure 1(d)). The predicted stability score (ΔΔG) was 0.44 kcal/mol. The interaction of the CYP2C9 gene with other function nodes is shown in Figure 1(c).

4. Discussion

Genetic and biomarker studies are still needed to identify epileptic patients at risk of developing medication resistance. Genetic variability has been demonstrated to contribute both in susceptibility and occurrence of disease and heterogeneity in therapy response [Reference Coe, Girirajan and Eichler19]. Grover et al. reported about 20–95% of the clinical variation in drug effects and disposition linked to genetics. On average, AED monotherapy is successful in 60–70% of newly diagnosed epileptic patients, with a switch to another AED being successful in up to 50% of patients who fail the first AED treatment [Reference Grover, Gourie-Devi and Baghel20]. CYP2C9 is responsible for up to 90% of phenytoin hydroxylation [Reference Kumari, Lakhan, Garg, Kalita, Misra and Mittal21]. The identification and characterization of genetic variants of the CYP2C9 gene might contribute to the optimization of antiepileptic drugs being administered. To the best of our knowledge, no studies have examined the relationship between CYP2C9 polymorphisms and drug-resistant epilepsy in Pakistani children. According to the global distribution analysis of CYP2C9 polymorphism, CYP2C9∗2 and CYP2C9∗3 variations are more common in European populations [Reference Sánchez-Diz, Estany-Gestal and Aguirre22]. Asians have no CYP2C9∗2 mutation and have a very low CYP2C9∗3 frequency, ranging from 1.132 to 5.418. Both the CYP2C9∗2 (4 percent) and CYP2C9∗3 (2 percent) alleles were found to be rare among Asians and Africans [Reference Soga, Nishimura and Ohtsuka23, Reference Hamdy, Hiratsuka and Narahara24]. Two CYP2C9 alleles associated with impaired metabolism are found in 11 and 8% of Americans, but just 3 and 0.8 percent of Africans. The rs1799853 and rs1057910 polymorphisms of the CYP2C9 gene were evaluated genetically in order to determine their involvement in the development of drug-resistant epilepsy in children in the current investigation. These polymorphisms were chosen based on the previously cited research findings, which imply that they are linked to drug-resistant epilepsy. We could not find any of these well-known SNPs in the current study; however, a novel variant c.375G > A was found in the drug-resistant patients. This may confer the drug-resistant phenotype in epilepsy patients. According to a study conducted in a north Indian population, the CYP2C9 mutant allele has a protective effect against the development of AED resistance [Reference Kumari, Lakhan, Garg, Kalita, Misra and Mittal21]. Previous research has found a substantial link between the CYP2C9 genotype and phenytoin maintenance dosage requirements [Reference van der Weide, Steijns, van Weelden and de Haan25]. van der Weide et al. discovered that phenytoin dose requirements are affected by CYP2C9 allelic variations; for patients with at least one mutant CYP2C9 allele, the mean phenytoin dosage required to produce a therapeutic blood concentration was roughly 37% lower than for wild-type people [Reference van der Weide, Steijns, van Weelden and de Haan25]. The mutant CYP2C9 alleles are linked to poor drug metabolism and function but on the other hand act as a barrier to the development of AED resistance. A report from Southern India showed the major role of CYP2C9 variants in phenytoin hydroxylation [Reference Rosemary, Surendiran, Rajan, Shashindran and Adithan26]. The polymorphic alleles change the activity of these isoenzymes, causing metabolism to be absent, diminished, or elevated. The findings of Lakhan et al. investigations revealed that CYP2C9 genetic variations have a substantial role in biasing epilepsy medication, demonstrating the importance of CYP2C9 mutants in preventing medication unresponsiveness in epileptic patients [Reference Lakhan, Kumari, Singh, Kalita, Misra and Mittal27]. The rs1799853 polymorphism in the CYP2C9 gene has been linked to the incidence of T allele, which four times increases the likelihood of medication resistance in individuals with epilepsy [Reference Makowska, Smolarz, Bryś, Forma and Romanowicz28]. The FDA (Food and drug Administration authority) has updated the clinical pharmacology part of the phenytoin label to include a warning concerning the risk of abnormally high levels in individuals who have specific CYP2C9 allelic variations [Reference Calderon-Ospina, Galvez and López-Cabra29]. Another study found a link between CYP2C9 polymorphisms and lower phenytoin metabolism and improved medication responsiveness. Nonresponders had a lower frequency of the heterozygous CYP2C9∗3 allele, according to the research by Lakhan et al. [Reference Lakhan, Kumari, Singh, Kalita, Misra and Mittal27, Reference Ufer, Mosyagin and Muhle30]. In the current study, no differences in genotypes of drug-resistant, drug-responsive, and control groups were observed which may be attributed to various factors. Our study has some limitations. Firstly, the study’s power may be insufficient as our sample size may not be large enough to establish a meaningful link. Perhaps in a larger investigation, it might be demonstrated. Secondly, some polymorphisms, such as those in the CYP2C9, CYP2C19, and UGT2B7 genes, are linked to the metabolism of some AEDs but not others. Our research did not aim to look at the link between polymorphisms and medication resistance in individuals on a specific AED. Additionally, drug resistance is a complicated trait that is influenced by several genes. The expression of multidrug transporters, in addition to drug-metabolizing enzymes, modulates phenytoin and carbamazepine disposition and may explain interindividual pharmacokinetic heterogeneity.

4.1. Conclusions

The patterns of genetic variations of the drug-metabolizing gene (CYP2C9) in different ethnicities translate into variation in drug response. Understanding CYP2C9 variability would improve rational drug usage and has public health implications. This study was designed to screen out the possible genetic variants of the CYP2C9 gene associated with AED resistance in Pakistani pediatric epilepsy patients. Our findings indicated an overall similar distribution of wild-type alleles in AED-resistant, AED-responsive, and control subjects. The wild-type alleles are reported to be associated with a higher rate of metabolism and early elimination of drug, thus requiring higher dose of AED to attain seizure freedom. The higher dose of AED is likely to be a contributing factor towards AED resistance. We also identified a novel variant. We analyzed only two SNPs of the CYP2C9 gene which were reported to be the hotspot for altering drug response. The pronouncement of CYP2C9 polymorphic contribution to drug response entails the screening of the whole gene on a larger scale in Pakistani pediatric epilepsy patients. The role of CYP2C9 polymorphism influence on drug response remains to be determined. Early detection of CYP2C9 polymorphism may help to predict and personalize the drug dose requirement. Further research is needed to validate the novel polymorphism found and those previously reported. It is noteworthy that multifactorial disorders such as epilepsy are influenced by a variety of factors that may be taken into account in the future.

Data Availability

The data used to support the findings of this study are available upon request from the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest regarding the publication of this paper.

Acknowledgments

The authors are thankful to all the participants of this study. This work was supported by the HigherEducation Commission, Pakistan (No. 8369/Punjab/NRPU/R&D/HEC/2017) under the National Research Program for Universities.

References

Manford, M., “Recent advances in epilepsy,Journal of Neurology, vol. 264, no. 8, pp. 18111824, 2017.CrossRefGoogle ScholarPubMed
Udani, V., “Pediatric epilepsy-an Indian perspective,Indian Journal of Pediatrics, vol. 72, no. 4, pp. 309313, 2005.CrossRefGoogle ScholarPubMed
Kozera-Kępniak, A., Jastrzębski, K., and Klimek, A., “Pharmacogenetic determinants of drug resistance in epilepsy,Aktualn Neurol, vol. 13, pp. 96102, 2013.Google Scholar
Lazarowski, A. and Czornyj, L., “Potential role of multidrug resistant proteins in refractory epilepsy and antiepileptic drugs interactions,Drug Metabolism and Drug Interactions, vol. 26, no. 1, pp. 21–6, 2011.CrossRefGoogle ScholarPubMed
Emich-Widera, E., Likus, W., Kazek, B. et al., “CYP3A5∗3 and C3435∗T MDR1 polymorphisms in prognostication of drug-resistant epilepsy in children and adolescents,BioMed Research International, vol. 2013, Article ID 526837, 7 pages, 2013.CrossRefGoogle Scholar
Ghosh, C., Hossain, M., Solanki, J., Najm, I. M., Marchi, N., and Janigro, D., “Overexpression of pregnane X and glucocorticoid receptors and the regulation of cytochrome P450 in human epileptic brain endothelial cells,Epilepsia, vol. 58, no. 4, pp. 576585, 2017.CrossRefGoogle ScholarPubMed
Ke, X.-J., Cheng, Y.-F., Yu, N., and Di, Q., “Effects of carbamazepine on the P-gp and CYP3A expression correlated with PXR or NF-κB activity in the bEnd.3 cells,Neuroscience Letters, vol. 690, pp. 4855, 2019.CrossRefGoogle ScholarPubMed
Lin, J. H. and Lu, A. Y., “Role of pharmacokinetics and metabolism in drug discovery and development,Pharmacological Reviews, vol. 49, no. 4, pp. 403449, 1997.Google ScholarPubMed
Pierzchała, K., “Padaczka oporna na leczenie–epidemiologia i aktualny stan badań,Neurologia I Neurochirurgia Polska, vol. 44, no. 3, pp. 285290, 2010.CrossRefGoogle Scholar
Brandolese, R., Scordo, M., Spina, E., Gusella, M., and Padrini, R., “Severe phenytoin intoxication in a subject homozygous for CYP2C93,Clinical Pharmacology & Therapeutics, vol. 70, no. 4, pp. 391394, 2001.Google Scholar
Scordo, M. G., Aklillu, E., Yasar, U., Dahl, M.-L., Spina, E., and Ingelman-Sundberg, M., “Genetic polymorphism of cytochrome P4502C9 in a Caucasian and a black African population,British Journal of Clinical Pharmacology, vol. 52, no. 4, pp. 447450, 2001.CrossRefGoogle Scholar
Franco, V. and Perucca, E., “CYP2C9 polymorphisms and phenytoin metabolism: implications for adverse effects,Expert Opinion on Drug Metabolism and Toxicology, vol. 11, no. 8, pp. 12691279, 2015.CrossRefGoogle ScholarPubMed
Suvichapanich, S., Jittikoon, J., Wichukchinda, N. et al., “Association analysis of CYP2C9∗3 and phenytoin-induced severe cutaneous adverse reactions (SCARs) in Thai epilepsy children,Journal of Human Genetics, vol. 60, no. 8, pp. 413417, 2015.CrossRefGoogle Scholar
Kousar, S., Wafai, Z. A., Wani, M. A., Jan, T. R., and Andrabi, K. I., “Clinical relevance of genetic polymorphism in CYP2C9 gene to pharmacodynamics and pharmacokinetics of phenytoin in epileptic patients: validatory pharmacogenomic approach to pharmacovigilance,International Journal of Clinical Pharmacology & Therapeutics, vol. 53, no. 7, pp. 504516, 2015.CrossRefGoogle ScholarPubMed
Chan, B. S. H., Sellors, K., Chiew, A. L., and Buckley, N. A., “Use of multi-dose activated charcoal in phenytoin toxicity secondary to genetic polymorphism,Clinical Toxicology, vol. 53, no. 2, pp. 131133, 2015.CrossRefGoogle ScholarPubMed
Kwan, P., Arzimanoglou, A., Berg, A. T. et al., “Definition of drug resistant epilepsy: consensus proposal by the ad hoc Task Force of the ILAE Commission on Therapeutic Strategies,Epilepsia, vol. 51, no. 6, pp. 1069–77, 2010.CrossRefGoogle Scholar
Green, M. R. and Sambrook, J., “Isolation of high-molecular-weight DNA using organic solvents,Cold Spring Harbour Protocols, vol. 2017, no. 4, Article ID prot093450, 2017.Google ScholarPubMed
Al-Mohizea, A. M., Alkharfy, K. M., Bagulb, K. M. et al., “Genetic variability and haplotype profile of MDR1 in Saudi Arabian males,Molecular Biology Reports, vol. 39, no. 12, pp. 1029310301, 2012.CrossRefGoogle ScholarPubMed
Coe, B. P., Girirajan, S., and Eichler, E. E., “The genetic variability and commonality of neurodevelopmental disease,American Journal of Medical Genetics Part C: Seminars in Medical Genetics, vol. 160C, no. 2, 2012.CrossRefGoogle ScholarPubMed
Grover, S., Gourie-Devi, M., Baghel, R. et al., “Genetic profile of patients with epilepsy on first-line antiepileptic drugs and potential directions for personalized treatment,Pharmacogenomics, vol. 11, no. 7, pp. 927941, 2010.CrossRefGoogle ScholarPubMed
Kumari, R., Lakhan, R., Garg, R. K., Kalita, J., Misra, U. K., and Mittal, B., “Pharmacogenomic association study on the role of drug metabolizing, drug transporters and drug target gene polymorphisms in drugresistant epilepsy in a north Indian population,Indian Journal of Human Genetics, vol. 17, pp. S32S40, 2011.Google Scholar
Sánchez-Diz, P., Estany-Gestal, A., Aguirre, C. et al., “Prevalence of CYP2C9 polymorphisms in the south of Europe,The Pharmacogenomics Journal, vol. 10, no. 3, Article ID 243, 2010.CrossRefGoogle Scholar
Soga, Y., Nishimura, F., Ohtsuka, Y. et al., “CYP2C polymorphisms, phenytoin metabolism and gingival overgrowth in epileptic subjects,Life Sciences, vol. 74, no. 7, pp. 827834, 2004.CrossRefGoogle ScholarPubMed
Hamdy, S. I., Hiratsuka, M., Narahara, K. et al., “Allele and genotype frequencies of polymorphic cytochromes P450 (CYP2C9, CYP2C19, CYP2E1) and dihydropyrimidine dehydrogenase (DPYD) in the Egyptian population,British Journal of Clinical Pharmacology, vol. 53, no. 6, pp. 596603, 2002.CrossRefGoogle Scholar
van der Weide, J., Steijns, L. S. W., van Weelden, M. J. M., and de Haan, K., “The effect of genetic polymorphism of cytochrome P450 CYP2C9 on phenytoin dose requirement,Pharmacogenetics, vol. 11, no. 4, pp. 287291, 2001.CrossRefGoogle ScholarPubMed
Rosemary, J., Surendiran, A., Rajan, S., Shashindran, C., and Adithan, C., “Influence of the CYP2CP & CYP2C19 polymorphisms on phenytoin hydroxylation in healthy individuals from south India,Indian Journal of Medical Research, vol. 123, no. 5, Article ID 665, 2006.Google Scholar
Lakhan, R., Kumari, R., Singh, K., Kalita, J., Misra, U. K., and Mittal, B., “Possible role of CYP2C9 & CYP2C19 single nucleotide polymorphisms in drug refractory epilepsy,Indian Journal of Medical Research, vol. 134, no. 3, pp. 295301, 2011.Google ScholarPubMed
Makowska, M., Smolarz, B., Bryś, M., Forma, E., and Romanowicz, H., “An association between the rs1799853 and rs1057910 polymorphisms of CYP2C9, the rs4244285 polymorphism of CYP2C19 and the prevalence rates of drug-resistant epilepsy in children,International Journal of Neuroscience, vol. 131, no. 12, pp. 18, 2020.Google ScholarPubMed
Calderon-Ospina, C. A., Galvez, J. M., López-Cabra, C. et al., “Possible genetic determinants of response to phenytoin in a group of Colombian patients with epilepsy,Frontiers in Pharmacology, vol. 11, Article ID 555, 2020.CrossRefGoogle Scholar
Ufer, M., Mosyagin, I., Muhle, H. et al., “Non-response to antiepileptic pharmacotherapy is associated with the ABCC2 −24C>T polymorphism in young and adult patients with epilepsy,Pharmacogenetics and Genomics, vol. 19, no. 5, pp. 353362, 2009.CrossRefGoogle Scholar
Figure 0

TABLE 1: Clinical and demographic attributes of drug-resistant and drug-responsive subjects.

Figure 1

FIGURE 1: (a) The pathogenicity score for c.374G > A, (b) electropherogram of wild and heterozygous variant c.374G > A, (c) interaction of CYP2C9 with other functional nodes, and (d) predicted transmembrane topology with the highlighted variant position.

Figure 2

TABLE 2: CYP2C9 variant identified by sequencing.