Back to Journals » Pharmacogenomics and Personalized Medicine » Volume 13

Genetic Association of Epilepsy and Anti-Epileptic Drugs Treatment in Jordanian Patients

Authors AL-Eitan LN , Al-Dalala IM, Elshammari AK, Khreisat WH , Nimiri AF, Alnaamneh AH, Aljamal HA , Alghamdi MA 

Received 21 July 2020

Accepted for publication 6 October 2020

Published 16 October 2020 Volume 2020:13 Pages 503—510

DOI https://doi.org/10.2147/PGPM.S273125

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr Martin H Bluth



Laith N AL-Eitan,1,2 Islam M Al-Dalala,3 Afrah K Elshammari,4 Wael H Khreisat,4 Aseel F Nimiri,4 Adan H Alnaamneh,1 Hanan A Aljamal,1 Mansour A Alghamdi5,6

1Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, Jordan; 2Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan; 3Department of Blood Banking, King Hussein Medical Centre, Royal Medical Services, Amman, Jordan; 4Queen Rania Hospital for Children, King Hussein Medical Center, Royal Medical Services, Amman, Jordan; 5Department of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia; 6Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia

Correspondence: Laith N AL-Eitan
Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
Tel +962-2-7201000 Ext.:23464
Fax +962-2-7201071
Email [email protected]

Purpose: The aim of this study was to investigate the possible effects of single-nucleotide polymorphisms (SNPs) within SLC1A1, SLC6A1, FAM131B, GPLD1, F2, GABRG2, GABRA1, and CACNG5 genes on response to anti-epileptic drugs (AEDs) and the genetic predisposition of epilepsy in Jordanian patients.
Patients and Methods: A total of 299 healthy individuals and 296 pediatric patients from the Jordanian population were recruited. Blood samples are collected, and genotyping was performed using a custom platform array analysis.
Results: The SLC1A1 rs10815018 and FAM131B rs4236482 polymorphisms found to be associated with epilepsy susceptibility. Moreover, SLC1A1 rs10815018 and GPLD1 rs1126617 polymorphisms were associated with generalized epilepsy (GE), while FAM131B rs4236482 is associated with the focal phenotype. Regarding the therapeutic response, the genetic polymorphisms of FAM131B rs4236482, GABRA1 rs2279020, and CACNG5 rs740805 are conferred poor response (resistance) to AEDs. There was no linkage of GLPD1 haplotypes to epilepsy, its subtypes, and treatment responsiveness.
Conclusion: Our findings suggested that SLC1A1, FAM131B, and GPLD1 polymorphisms increasing the risk of generating epilepsy, while FAM131B, GABRA1, and CACNG5 variants may play a role in predicting drug response in patients with epilepsy (PWE).

Keywords: pharmacogenetics, anti-epileptic drugs, generalized epilepsy, focal epilepsy, pharmacotherapy, Jordan

Introduction

Epilepsy is a common neurological condition that affects people of all ages.1 Epilepsy is affecting more than70 million individuals worldwide, wherein the number of patients is growing in developing countries nowadays.1 It also has a substantial impact on patients’ morbidity and mortality.2 Although the etiology of epilepsy remains unsettled, different evidences suggested that epilepsy could be generated due to genetic variants. It is believed that more than 70% of patients with epilepsy (PWE) have a genetic predisposition.3 Genetic abnormalities can alter the electrical impulses, affect channels function, modify neuronal excitability, and may also disrupt pharmacokinetics of AEDs, and therefore affect treatment efficacy.4

Epilepsy have many different syndromes and types which have studied extensively. However, information regarding detailed genetic factors and AED selection for the individual patient is insufficient; hence, the importance of pharmacogenomic studies in this area rises considerably.5 To date, there is a limited number of studies investigating the genetic and pharmacogenetic basis of common diseases, including epilepsy among the Jordanian population.611 Recent studies found that epilepsy is associated with several chromosomal regions, where mutations in these regions cause neurological dysfunction.12 The solute carrier gene, SLC1A1, is a high-affinity glutamate transporter that is associated with neurodevelopmental phenotypes, such as myoclonic-atonic epilepsy and schizophrenia.13,14 The other family member, the SLC6A1 gene, encodes a voltage-dependent gamma-aminobutyric acid (GABA) transporter GAT-1, which is accounts for GABA reuptake from the synapse. The latter of which is an essential inhibitory neurotransmitter that stabilizes the brain’s neuronal excitation.15 Several mutations within the SLC6A1gene were reported in patients with myoclonic-atonic seizures.15 The FAM131B gene is not well characterized yet; however, it is identified in patients with brain neoplasia such as astrocytoma and glioma.16 The glycosylphosphatidylinositol phospholipase, D1GPLD1 is encoded as a plasma protein.17 This gene is found mainly in the liver and brain, which may have a role in epilepsy pathology and pharmacology.17 For the F2 gene, especially the rs1799963 SNP, it showed to has some linkage with vascular and thrombotic diseases.18 Variants in the F2 gene are associated with increased production of prothrombin, which can lead to multiple neurological conditions.19 GABRG2 gene is encoding GABA, one of the brain’s main inhibitory neurotransmitters. Alteration in this gene is associated with epilepsy, as it may affect transcription, translation efficiency, as well as mRNA stability.20 GABRG2 and GABRA1mutations can cause impairment in GABA receptor function and biogenesis, in addition to the association with receptor mobilization. For this, GABRG2 and GABRA1 are highly implicated in the etiology of epilepsy.21 CACNG5 gene is encoding type II transmembrane AMPA receptor regulatory proteins (TARP). TARPs have a role in trafficking regulations and channel gating of the α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPA) receptors, which contribute to neuronal developmental and may play a role in the etiology of multiple neurological disorders.22

Thus, epilepsy is a crucial issue in clinical research, and there are promising results in its diagnosis and treatment. In the study, we aiming to assess the possible association between the targeted genetic variants of SLC1A1, SLC6A1, FAM131B, GPLD1, F2, GABRG2, GABRA1, and CACNG5 genes in epilepsy susceptibility and AEDs response in Jordanian PWE.

Patients and Methods

Ethical Approval

This study was approved by the Ethics Committee of Jordan University of Science and Technology (16/111/2017), Queen Rania Al Abdullah Hospital (QRAH), and the Jordanian Royal Medical Services (JRMS). Informed written consent was provided by patients or their parents/guardians before enrolment in this study.

Participants and Treatment Approach

A total of 296 pediatric patients with epilepsy were recruited from the Pediatric Neurology Clinic at QRAH. Control subjects (299) were recruited from the Blood Bank at the JRMS. Participants’ data and selection criteria have been reported previously in more detail.10 The patients were classified according to the guidelines of the International League Against Epilepsy (ILAE, 2010).23,24 The patients were aged less than 15 years, having two attacks of seizures within more than 24 hours apart, and receiving AEDs for a minimum of three months. The exclusion criteria rule out patients that have no sufficient medical records, unreliable seizure frequency, have liver disease, incompliant patients, or not visiting the clinics regularly for the follow-up. In addition, patients with abnormal psychometric development and neurological examination were also excluded.

According to the treatment protocol of QRAH, treatment of patients with generalized epilepsy (GE) starts at a dose of 10mg/kg valproic acid (VPA) (G.L. Pharma GmbH, Lannach, Austria). For patients with focal epilepsy (FE), patients were initially received 5 mg/kg of carbamazepine (CBZ) (Novartis Pharmaceuticals UK Ltd., Surrey, England). After the initiation of the treatment, the seizure was monitored for the first three to four weeks in order to evaluate the effectiveness of drug doses. During the follow-up visits, the therapeutic doses were increased gradually to reached 20 and 10 mg/kg of VPA and CBZ, respectively, to minimize seizure recurrence.

Regarding their response to AEDs, patients are classified into either drug-responsive or drug-resistant during the evaluation interval. On the one hand, if patients are seizure-free for at least three times the longest inter-seizure interval, or 12 months before starting a new intervention, regardless of which is longer, therefore, patient classified as responsive (124 subjects).25 On the other hand, the resistant group (171 subjects) failed trials of two adequately chosen AED to achieve sustained seizure freedom, whether as monotherapies or in combination.25

DNA Extraction and Genotyping

Nine SNPs (rs10815018, rs10510403, rs4236482, rs1126617, rs2076317, rs1799963, rs209337, rs2279020, and rs740805) were selected from SNP databases of the Applied Biosystems (http://www.appliedbiosystems.com), Ensembl (http://www.ensembl.org/index.html), and the National Center for Biotechnology Information (NCBI) (http://www.ncbi.nlm.nih.gov/SNP/). These SNPs are either previously studied in other ethnic groups or located in genes known to be associated with epilepsy phenotypes or affect treatment response. The peripheral blood of the participants was collected in EDTA tubes, and the genomicDNA was extracted using Wizard Genomic DNA purification kit according to the supplier’s instructions (Promega Corporation, Madison, WI, USA). DNA samples were genotyped in association with the Australian Genome Research Facility (AGRF) using the MassARRAY (iPLEX GOLD) system (Sequenom, San Diego, CA, USA).

Statistical Analysis

The obtained data analysis has carried out using the Statistical Package for Social Sciences (SPSS) software version 22.0 (IBM Corporation, New York, USA) in addition to SNPStats Web Tool (https://www.snpstats.net/start.htm). Allele and genotype frequencies between patients and controls, as well as genotype frequencies deviation from Hardy–Weinberg equilibrium, were estimated by Chi-square (χ2) test. A P-value of less than 0.05 was considered a significant value.

Results

In this study, a total of 296 Jordanian pediatric patients with epilepsy were enrolled (162, 54.8% males; 134, 45.2% females; mean±SD age: 7.1±4.1), in addition to 299 healthy controls of matched sex and ethnicity (152, 50.8% males; 147, 49.2% females; meanage±SD: 5.94±3.7) with no significant differences in terms of gender and age. More than half of the patients were drug-resistant (171, 58.1%), and 41.9% (124) were drug-responsive. The studied SNPs exhibited no association with response status to AEDs except for the FAM131B rs4236482, GABRA1 rs2279020, and CACNG5 rs740805 (Table 1). For the rs4236482, G allele (ie, in homozygous or heterozygous genotype) has emerged as a risk factor in drug-resistant patients (P= 0.018, 0.016, and 0.021), which is also seen in rs2279020 (P= 0.04, and 0.013). The AA genotype of rs740805polymorphism has a higher risk of developing drug resistance in patients (P= 0.017, and 0.035).

Table 1 The Distributions of SLC1A1, SLC6A1, FAM131B, GPLD1, F2, GABRG2, GABRA1, and CACNG5 SNPs in Resistant and Responsive Patients

Genetic and allelic distribution of the polymorphisms between healthy controls and PWErevealed genetic association only in SLC1A1 rs10815018 and FAM131B rs4236482 SNPs (Table 2). It found that rs10815018AA genotype (P= 0.025) and rs4236482 G allele (P= 0.038, and 0.014) are risk factors for increasing epilepsy susceptibility. Furthermore, epileptic patients are classified into those with generalized epilepsy (172) and focal epilepsy (124). The analysis showed that rs1126617 SNP of GPLD1, and rs10815018 of the SLC1A1 gene are associated with the susceptibility to have GE (Table 3). As the latter SNP AA genotype is found earlier to be associated with epilepsy, it is further found to be associated with its generalized onset (P= 0.016, 0.004). In the case of focal epileptic patients, none of the studied SNPs were associated, except for the FAM131B rs4236482, where the homozygous AA genotype increases the risk of its development (Table 4; P= 0.23, 0.008).

Table 2 The Distributions of SLC1A1, SLC6A1, FAM131B, GPLD1, F2, GABRG2, GABRA1, and CACNG5 SNPs in PWE and Healthy Controls

Table 3 The Distributions of SLC1A1, SLC6A1, FAM131B, GPLD1, F2, GABRG2, GABRA1, and CACNG5 SNPs in Patients with Generalized Epilepsy (GE) and Healthy Controls

Table 4 The Distributions of SLC1A1, SLC6A1, FAM131B, GPLD1, F2, GABRG2, GABRA1, and CACNG5 SNPs in Patients with Focal Epilepsy (FE) and Healthy Controls

The three haplotypes of the GPLD1 gene failed to show any linkage with epilepsy, its broad types, or treatment response among the patients (Tables S1S4).

Discussion

Several genetic and pharmacogenetic studies attempted to investigate the correlation of different gene polymorphisms with respect to the susceptibility to develop epilepsy, and response to AEDs treatment.15,2631 However, as most studies investigate previously examined candidate genes, we tend to include some genes associated with various neural pathways, different neurological disorders, and response to antipsychotic drugs.15,3235

Although there are several different pharmacokinetic factors regulating drug metabolism at different stages (absorption, distribution, metabolism, and clearance), yet genetic polymorphisms are likely to alter the final phenotype.36,37 To date, the gap between drug-resistant epilepsy prevalence and the explored genetic variants still considerable. Around 20% of pediatric patients with epilepsy are pharmacoresistant, as they exhibit resistance to multiple AEDs.38 Epilepsy treatment response is characterized by the remission of seizures and responders to drug treatment, it is defined by the ILAE as “individuals being seizure-free for at least 12 months after starting AED therapy”.39 In our study, the examined SNPs rs10815018, rs10510403, rs1126617, rs2076317, rs1799963, and rs209337 did not show any linkage with treatment response. Based on the genotype and allele analysis, the G allele was of significantly higher distribution in drug-resistant patients withrs4236482 and rs2279020 intronic variants (278, 81% and 150, 55%, respectively) compare to the responsive group (175, 71% and 89, 37%, respectively). To the best of our knowledge, rs4236482 did not show previously to be associated with the efficacy of AEDs treatment. As a result, FAM131Bvariantsare expected to interfere with any of the drug modulation steps, an issue that needs substantial investigation. The GABRA1receptor variants, such as rs2279020 were examined as a potential factor affecting treatment efficacy.2628,40 Our results support the association of rs2279020 with AEDsresistance, which contradicts the result in Chinese Han PWE,40 but similar to Indian patients.27 Moreover, as suggested that voltage-dependent calcium channels (VDCCs) genes are one of the major effectors in pharmacogenetics of epilepsy,4143 A allele of CACNG5 rs740805 appear to associated with the drug resistance phenotype with a higher rate in resistant patients (323, 95% vs 225, 91% in the responsive group). This finding suggested the idea that other VDCCs genes may play a role in epilepsy treatment outcomes.

In this study, we failed to show an association between the SLC6A, F2, and GABRA1 polymorphisms and vulnerability to develop epilepsy in general, its broad phenotypes, or treatment efficacy in our population. However, SLC1A1rs10815018 is associated with epilepsy and its generalized seizures. This result is expected since this glutamate transporter variants have been reported in one of the reflux epilepsies; the hot water epilepsy,44 and other several neurodevelopmental disorders, such as schizophrenia,45 obsessive-compulsive,46,47 and Rett syndrome (RTT).14 Furthermore, SLC1A1 was observed in patients with either focal or generalized epilepsy from four different regions (Brussels, Belgium, Dublin, and Ireland).48 In addition, FAM131B rs4236482 is significantly associated in patients with epilepsy, and particularly, in patients with focal onset seizures. On the other hand, Kampen et al reported the association of the FAM131B gene in a sixteen‐year‐old boy with generalized tonic‐clonic seizures.49 Considering its association with drug resistance in the study population, this represents the first step toward an intensive characterization of the FAM131Bvariations. Moreover, GPLD1 rs1126617 is also associated with the generalized onset epilepsy. Of the epilepsy type classification studied by Shazadi et al, four out of 16 SNPs were in strong linkage disequilibrium with each other, including rs2076317 and rs1126617 variants of the GPLD1 gene.50 These findings shed light on the relation between SLC1A, GPLD, and FAM131B genes with epilepsy seizures classes and highlight the importance of patient classification with respect to their treatment response.

Conclusions

Overall, epilepsy is a common chronic neurological disorder affecting millions of people worldwide causing considerable morbidity and mortality.51 This disease is affected by several factors, and genetic factors are believed to play a major role in epileptogenesis. This study presents for the first time some variants that affect disease development and its treatment as well in the Jordanian epileptic patients. Further pharmacogenetic studies are needed to identify other genetic factors that affect genetic susceptibility and treatment responsiveness outcomes to improve the efficacy and safety of epilepsy treatment. The findings need to be confirmed by the recruitment of a larger sample size and including further analysis exploring SNP-SNP, gene-gene, and haplotype–haplotype interaction, in addition to functionality studies to characterize the appearance of particular phenotypes.

Funding

This work was supported by a grant from the Deanship of Research at Jordan University of Science and Technology (RN: 407/2017).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Löscher W, Potschka H, Sisodiya SM, et al. Drug resistance in epilepsy: clinical impact, potential mechanisms, and new innovative treatment options. Pharmacol Rev. 2020;72:606–638. doi:10.1124/pr.120.019539

2. World Health Organization. Epilepsy: aetiology, epidemiology and prognosis. Fact sheet no. 165; 2001. Available from: http://www.who.int/news-room/fact-sheets/detail/epilepsy. Accessed July 20, 2020.

3. Kjeldsen MJ, Kyvik KO, Friis ML, et al. Genetic and environmental factors in febrile seizures: a Danish population-based twin study. Epilepsy Res. 2002;51:167–177. doi:10.1016/S0920-1211(02)00121-3

4. Hedrich U, Maljevic S, Lerche H. Mechanisms of genetic epilepsies. e-Neuroforum. 2013;4:23–30.

5. Cavalleri GL, McCormack M, Alhusaini S, et al. Pharmacogenomics and epilepsy: the road ahead. Pharmacogenomics. 2011;12:1429–1447. doi:10.2217/pgs.11.85

6. Al-Eitan L, Haddad Y. Emergence of pharmacogenomics in academic medicine and public health in Jordan: history, present state and prospects. Curr Pharmacogenomics Person Med. 2014;12:167–175. doi:10.2174/1875692113666150115221210

7. AL-Eitan L, Tarkhan A. Practical challenges and translational issues in pharmacogenomics and personalized medicine from 2010 onwards. Curr Pharmacogenomics Person Med. 2016;14:7–17. doi:10.2174/1875692115666161215103842

8. Al-Eitan L, Al-Dalalah I, Mustafa M, et al. Genetic polymorphisms of CYP3A5, CHRM2, and ZNF498 and their association with epilepsy susceptibility: a pharmacogenetic and case–control study. Pharmgenomics Pers Med. 2019;12:225.

9. AL-Eitan L, Al-Dalalah I, Aljamal H. Effects of GRM4, SCN2A and SCN3B polymorphisms on antiepileptic drugs responsiveness and epilepsy susceptibility. Saudi Pharm J. 2019;27:731–737. doi:10.1016/j.jsps.2019.04.009

10. Al-Eitan L, Al-Dalalah I, Elshammari A, et al. The impact of potassium channel gene polymorphisms on antiepileptic drug responsiveness in arab patients with epilepsy. J Pers Med. 2018;8:37. doi:10.3390/jpm8040037

11. AL-Eitan LN, Al-Dalalah IM, Mustafa MM, et al. Effects of MTHFR and ABCC2 gene polymorphisms on antiepileptic drug responsiveness in Jordanian epileptic patients. Pharmgenomics Pers Med. 2019;12:87–95.

12. D’Adamo MC, Catacuzzeno L, Giovanni G, et al. K+ channelepsy: progress in the neurobiology of potassium channels and epilepsy. Front Cell Neurosci. 2013;7:134.

13. Kanai Y, Hediger M. The glutamate/neutral amino acid transporter family SLC1: molecular, physiological and pharmacological aspects. Pflugers Arch. 2004;447:469–479. doi:10.1007/s00424-003-1146-4

14. Lucariello M, Vidal E, Vidal S, et al. Whole exome sequencing of Rett syndrome-like patients reveals the mutational diversity of the clinical phenotype. Hum Genet. 2016;135:1343–1354.

15. Carvill G, McMahon J, Schneider A, et al. Mutations in the GABA transporter SLC6A1 cause epilepsy with myoclonic-atonic seizures. Am J Hum. 2015;96:808–815. doi:10.1016/j.ajhg.2015.02.016

16. Busse T, Roth J, Wilmoth D, et al. Copy number alterations determined by single nucleotide polymorphism array testing in the clinical laboratory are indicative of gene fusions in pediatric cancer patients. Genes Chromosomes Cancer. 2017;56:730–749. doi:10.1002/gcc.22477

17. Roth J, Santi M, Rorke-Adams L, et al. Diagnostic application of high resolution single nucleotide polymorphism array analysis for children with brain tumors. Cancer Genet. 2014;207:111–123. doi:10.1016/j.cancergen.2014.03.002

18. Rinde L, Morelli V, Smabrekke B, et al. Effect of prothrombotic genotypes on the risk of venous thromboembolism in patients with and without ischemic stroke. The Tromso Study. Thromb Haemost. 2019;17:749–758. doi:10.1111/jth.14410

19. Maggio N, Shavit E, Chapman J, Segal M. Thrombin induces long-term potentiation of reactivity to afferent stimulation and facilitates epileptic seizures in rat hippocampal slices: toward understanding the functional consequences of cerebrovascular insults. J Neurosci. 2008;28:732–736.

20. Abou El Ella S, Tawfik M, Abo El Fotoh W, et al. The genetic variant “C588T” of GABARG2 is linked to childhood idiopathic generalized epilepsy and resistance to antiepileptic drugs. Seizure. 2018;60:39–43. doi:10.1016/j.seizure.2018.06.004

21. Hernandez C, XiangWei W, Hu N, et al. Altered inhibitory synapses in de novo GABRA5 and GABRA1 mutations associated with early onset epileptic encephalopathies. Brain. 2019;5:5485825.

22. Cull-Candy S, Kelly L, Farrant M. Regulation of Ca2+-permeable AMPA receptors: synaptic plasticity and beyond. Curr Opin Neurobiol. 2006;16:288–297. doi:10.1016/j.conb.2006.05.012

23. Fisher R, Cross J, French J, et al. Operational classification of seizure types by the international league against epilepsy: position paper of the ilae commission for classification and terminology. Epilepsia. 2017;58:522–530. doi:10.1111/epi.13670

24. Scheffer IE, Berkovic S, Capovilla G, et al. ILAE classification of the epilepsies: position paper of the ILAE commission for classification and terminology. Epilepsia. 2017;58:512–521. doi:10.1111/epi.13709

25. Kwan P, Arzimanoglou A, Berg A, et al. Definition of drug resistant epilepsy: consensus proposal by the ad hoc Task Force of the ILAE Commission on Therapeutic Strategies. Epilepsia. 2010;51:069–1077.

26. Feng W, Mei S, Zhu L, et al. Effects of UGT1A6 and GABRA1 on standardized valproic acid plasma concentrations and treatment effect in children with epilepsy in China. Ther Drug Monit. 2016;38:738–743.

27. Kumari R, Lakhan R, Kalita J, et al. Association of alpha subunit of GABAA receptor subtype gene polymorphisms with epilepsy susceptibility and drug resistance in north Indian population. Seizure. 2010;19:237–241.

28. Balan S, Sathyan S, Radha SK, et al. GABRG2, rs211037 is associated with epilepsy susceptibility, but not with antiepileptic drug resistance and febrile seizures. Pharmacogenomics. 2013;23:605–610.

29. Wang J, Lin ZJ, Liu L, et al. Epilepsy-associated genes. Seizure. 2017;44:11–20. doi:10.1016/j.seizure.2016.11.030

30. The Epilepsy Genetic Association Database (epiGAD), updated in 19 feb 2020. Available from: http://www.epilepsygenes.org/egad_list.html. Accessed September 15, 2020.

31. Fricke-Galindo I, Ortega-Vázquez A, Monroy-Jaramillo N, et al. Allele and genotype frequencies of genes relevant to anti-epileptic drug therapy in Mexican-Mestizo healthy volunteers. Pharmacogenomics. 2016;17:1913–1930. doi:10.2217/pgs-2016-0078

32. Curtis D, Vine AE, McQuillin A, et al. Case-case genome-wide association analysis shows markers differentially associated with schizophrenia and bipolar disorder and implicates calcium channel genes. PSYCHIAT GENET. 2011;21:1–4. doi:10.1097/YPG.0b013e3283413382

33. Thoeringer CK, Ripke S, Unschuld PG, et al. The GABA transporter 1 (SLC6A1): a novel candidate gene for anxiety disorders. J Neural Transm. 2009;116:649–657. doi:10.1007/s00702-008-0075-y

34. Wilmsdorff MV, Blaich C, Zink M, et al. Gene expression of glutamate transporters SLC1A1, SLC1A3 and SLC1A6 in the cerebellar subregions of elderly schizophrenia patients and effects of antipsychotic treatment. World J Biol Psychiatry. 2013;14:pp.490–499. doi:10.3109/15622975.2011.645877

35. Imbrici P, Conte Camerino D, Tricarico D. Major channels involved in neuropsychiatric disorders and therapeutic perspectives. Front Genet. 2013;4:76. doi:10.3389/fgene.2013.00076

36. Kasperavičiūtė D, Sisodiya SM. Epilepsy pharmacogenetics. Pharmacogenomics. 2009;10:817–836. doi:10.2217/pgs.09.34

37. Szoeke CE, Newton M, Wood JM, et al. Update on pharmacogenetics in epilepsy: a brief review. Lancet Neurol. 2006;5:189–196. doi:10.1016/S1474-4422(06)70352-0

38. Wirrell EC. Predicting pharmacoresistance in pediatric epilepsy. Epilepsia. 2013;54:19–22. doi:10.1111/epi.12179

39. Kwan P, Brodie M. Definition of refractory epilepsy: defining the indefinable? Lancet Neurol. 2010;9:27–29. doi:10.1016/S1474-4422(09)70304-7

40. Zhou L, Cao Y, Long H, et al. ABCB1, ABCC2, SCN1A, SCN2A, GABRA1 gene polymorphisms and drug resistant epilepsy in the Chinese Han population. Pharmazie. 2015;70:416–420.

41. Glauser TA, Holland K, O’Brien VP, et al. Pharmacogenetics of antiepileptic drug efficacy in childhood absence epilepsy. Ann Neurol. 2017;81:444–453. doi:10.1002/ana.24886

42. Lv N, Qu J, Long H, et al. Association study between polymorphisms in the CACNA1A, CACNA1C, and CACNA1H genes and drug-resistant epilepsy in the Chinese Han population. Seizure. 2015;30:64–69. doi:10.1016/j.seizure.2015.05.013

43. Wolking S, Schulz H, Nies AT, et al. Pharmacoresponse in genetic generalized epilepsy: a genome-wide association study. Pharmacogenomics. 2020;21:325–335. doi:10.2217/pgs-2019-0179

44. Karan KR, Satishchandra P, Sinha S, et al. Rare SLC1A1 variants in hot water epilepsy. Hum Genet. 2017;136:693–703. doi:10.1007/s00439-017-1778-7

45. Afshari P, Myles-Worsley M, Cohen OS, et al. Characterization of a novel mutation in SLC1A1 associated with schizophrenia. Mol Psychiatry. 2015;1:125–144.

46. Walitza S, Wendland JR, Gruenblatt E, et al. Genetics of early-onset obsessive-compulsive disorder. Eur Child Adolesc Psychiatry. 2010;19:227–235. doi:10.1007/s00787-010-0087-7

47. Samuels J, Wang Y, Riddle MA, et al. Comprehensive family-based association study of the glutamate transporter gene SLC1A1 in obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet. 2011;156:472–477. doi:10.1002/ajmg.b.31184

48. Heinzen E, Radtke R, Urban T, et al. Rare deletions at 16p13. 11 predispose to a diverse spectrum of sporadic epilepsy syndromes. Am J Hum Genet. 2010;86:707–718. doi:10.1016/j.ajhg.2010.03.018

49. van Kampen F, Doornbos M, van Rijn M, et al. A low‐grade astrocytoma in a sixteen‐year‐old boy with a 7q11. 22 deletion. Clin Case Rep. 2018;6::274. doi:10.1002/ccr3.1312

50. Shazadi K. Genetic Predictors for Epilepsy Development, Treatment Response and Dosing [Dissertation]. University of Liverpool; 2013.

51. Banerjee PN, Filippi D, Hauser WA. The descriptive epidemiology of epilepsy—a review. Epilepsy Res. 2009;85(1):pp.31–45. doi:10.1016/j.eplepsyres.2009.03.003

Creative Commons License © 2020 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.