Open Access

Genetic variations associated with pharmacoresistant epilepsy (Review)

  • Authors:
    • Noemí Cárdenas‑Rodríguez
    • Liliana Carmona‑Aparicio
    • Diana L. Pérez‑Lozano
    • Daniel Ortega‑Cuellar
    • Saúl Gómez‑Manzo
    • Iván Ignacio‑Mejía
  • View Affiliations

  • Published online on: February 24, 2020     https://doi.org/10.3892/mmr.2020.10999
  • Pages: 1685-1701
  • Copyright: © Cárdenas‑Rodríguez et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epilepsy is a common, serious neurological disorder worldwide. Although this disease can be successfully treated in most cases, not all patients respond favorably to medical treatments, which can lead to pharmacoresistant epilepsy. Drug‑resistant epilepsy can be caused by a number of mechanisms that may involve environmental and genetic factors, as well as disease‑ and drug‑related factors. In recent years, numerous studies have demonstrated that genetic variation is involved in the drug resistance of epilepsy, especially genetic variations found in drug resistance‑related genes, including the voltage‑dependent sodium and potassium channels genes, and the metabolizer of endogenous and xenobiotic substances genes. The present review aimed to highlight the genetic variants that are involved in the regulation of drug resistance in epilepsy; a comprehensive understanding of the role of genetic variation in drug resistance will help us develop improved strategies to regulate drug resistance efficiently and determine the pathophysiological processes that underlie this common human neurological disease.

Introduction

Epilepsy is one of the most common neurological diseases worldwide and is considered a major public health problem (1,2). The International League Against Epilepsy (ILAE) has established that the term ‘epilepsy’ refers to a disease of the brain that meets any of the following conditions: i) At least two non-induced seizures, or reflexes, that occur ≥24 h apart; ii) one non-induced seizure, or reflex, and a risk of further seizures similar to the general recurrence risk (≥60%) following two non-induced seizures that occur over the next 10 years; or iii) the diagnosis of an epilepsy syndrome (3,4). Epilepsy is considered to be resolved when an individual with epilepsy has remained seizure-free for 10 years and without antiepileptic drug treatment for ≥5 years (4).

For the most accurate study of epilepsy, the ILAE (5,6) has organized and classified seizures and several epilepsy types as focal, generalized and of unknown onset, based on certain characteristics, including seizure type, electroencephalography (EEG) features, imaging studies, age-related features and triggering factors, such as comorbidities and prognosis (6,7); this classification involved the work of epileptologists, neurophysiologists and epilepsy researchers (6).

A previous study demonstrated that epileptic seizures are associated with several mechanisms that involve the glutamate excitotoxicity process, microglial activation, mitochondrial dysfunction, degenerative processes, and the presence of reactive oxygen species and oxidative stress (8). In addition, it has previously been reported that certain regulatory processes are involved at the transcriptional level; for example, the nuclear transcription factor erythroid-derived 2-like 2 was revealed to serve a role in epileptic seizures (9,10). A recent study conducted by our group used microarray analysis in children with epilepsy to demonstrate that those with epilepsy overexpressed genes that were related to the transcriptional factor cAMP-response element binding protein (CREB) compared with normal children, in addition to significantly altered expression levels of genes involved in energy metabolism, redox balance and the immune response (11). The differential gene expression, particularly genes related to CREB, observed in children before and after the administration of valproic acid indicated that the activity of antiepileptic drugs (AED) is dependent on target genes. These data suggested a role for genetics in epilepsy development and highlighted the importance of studying the genetic mechanisms associated with drug resistance. This would provide an improved understanding of the impact of pharmacological treatment on epilepsy and in the patient's daily activities, of which both are influenced by: i) The patient's response to treatment; ii) the relationship between the number and type of seizures, and the modified transportation of proteins or their receptors due to the drug's activity and the presence of genetic variations; iii) the possible influence of the phenotypic characteristics of the patient in response to the treatment; iv) the impact of the presence of the genetic variants in the functionality of the transporting proteins and AED target proteins; v) the interference of the potential alterations in the target protein on the mechanism of action of the AED due to the presence of the variant; vi) the influence of the inflammatory and immunological response; vii) the predisposition to some of the different aspects of epilepsy, including refractoriness or decreased sensitivity to the AED effect; and viii) the result following the combination of several of these or other mechanisms (1215). In accordance with the last point, the objective of the present review was to focus on describing the findings of genetic alterations involved in pharmacoresistant epilepsy.

Pharmacoresistant epilepsy

The term pharmacoresistant epilepsy refers to a type of epilepsy that does not respond to at least two AEDs, which were chosen and used in monotherapy or combination therapy (bi- or polytherapy) and fail to fully control seizures for an adequate period (16). In 2011, the ILAE proposed to standardize the definition of pharmacoresistant epilepsy as the presence of seizures in a period of 6 months, even under proper therapeutic regimens (either monotherapy or in combination) (16). Two studies performed in 2000 and 2012 reported that people with epilepsy (PWE) responded differently to AED treatment, since 47–49.5% of the patients required one AED to control the seizures, 13–13.3% required a second AED, and 3.7–4% needed a third AED, which was administered either alone or in combination (17,18). To summarize, Kwan and Brodie (17) observed that in a prospective study of 525 PWE (age, 9–93 years), 333 of them (63%) remained seizure-free during AED administration and seizures that did occur were more persistent in patients with symptomatic and cryptogenic epilepsy (40%) compared with those with idiopathic epilepsy (26%). Moreover, among 470 previously untreated patients, 222 of them (47%) became seizure-free during treatment with their first AED, 67 patients (14%) became seizure-free during treatment with a second or third AED and 12 patients (3%) were controlled with two AEDs administered together (17). Brodie et al (18) subsequently discovered that patients have differential responses to AEDs; out of 1,098 PWE (ages, 9–93 years) studied, 749 of them (68%) were seizure-free with AED monotherapy, but in 272 patients (25%), freedom from seizures was never attained. Moreover, <2% of patients became seizure-free with the use of up to six or seven AEDs (18). In addition to the above findings, epidemiological studies conducted among children and adults have discovered that 20–40% of PWE present with pharmacoresistant epilepsy (1922), which negatively impacts their quality of life because it is also associated with increases in psychiatric comorbidities and the risk of premature death and social discrimination (23).

It is important to highlight that pharmacoresistant epilepsy may also cause serious socioeconomic problems. For instance, Argumosa and Herranz (24) evaluated the economic cost of controlled and uncontrolled epilepsy in Spain (participants were <14 years old) and reported that the mean annual cost of controlled epilepsy was $2,002.36 USD, whereas the cost of uncontrolled epilepsy was $5,348.50 USD; thus, uncontrolled epilepsy was 2.7 times more expensive compared with controlled epilepsy. Given the elevated costs of treatment, alternative therapeutic approaches, such as the ketogenic diet (25), high doses of steroids (26) and brain surgery (27) have all been implemented The ketogenic diet has proved beneficial in PWE in which pharmacological and/or surgical treatment is not effective; this diet is centered around a very high-fat and low-carbohydrate intake, reducing carbohydrates to <10% of used energy (90% of the total caloric intake comes from fat, 6% from protein and 4% from carbohydrates). These proportions trigger a systemic shift from glucose metabolism towards the metabolism of fatty acids, which yields ketone bodies that serve as the energy source to replace glucose in the brain (28). In patients with pharmacoresistant epilepsy, a ketogenic diet has been observed to improve the quality of life and decreases seizure frequency in ~30% of patients (25). In a significant number of patients with pharmacoresistant epilepsy, curative epilepsy surgery cannot be offered since there are multiple epileptogenic zones; for these patients, neurostimulation techniques, such as vagus nerve stimulation, deep brain stimulation and responsive neurostimulation, are viable treatment options that should be considered in every patient with this type of epilepsy that is unsuitable for surgery (27,29). These techniques provide palliative care, resulting in a 10–80% reduction in seizure occurrence (29). Furthermore, if all the aforementioned treatment approaches fail to control the seizures, cannabidiol (Epidiolex®) can be prescribed; this is a pharmaceutical product approved by the U.S. Food and Drug Administration that consists of 99% cannabidiol derived from cannabis (30,31). A previous study reported that out of 43 Mexican children with pharmacoresistant epilepsy, a decrease in the number of epileptic seizures were observed in 81.3% of patients and 20.9% patients displayed a reduction in the number of AEDs with the use of cannabidiol. Significant adverse effects related to appetite or sleep were only observed in 42% of patients following the use of cannabidiol (31).

Genetic variations associated with pharmacoresistant epilepsy

Previous studies have suggested that pharmacokinetic and pharmacodynamic mechanisms form the physiopathological basis of pharmacoresistant epilepsy (Table I) (22,32,33). Advances in genomic technologies have facilitated the genome-wide discovery of common and rare variants and have increased our understanding of genetics in epilepsy (34); however, the mechanisms underlying pharmacological resistance have not been fully elucidated (35). Some of the most important genes associated with the physiopathology of epilepsy were associated with the neuronal acetylcholine receptor, neuronal potassium channels (KCNs), voltage-dependent sodium channels (SCNs), calcium channels and γ-aminobutyric acid (GABA) receptors (32,36,37). Table II summarizes some of the genetic variants that occur in the main genes linked to epilepsy from recent studies.

Table I.

Mechanisms associated with pharmacoresistant epilepsy.

Table I.

Mechanisms associated with pharmacoresistant epilepsy.

MechanismDescription
PharmacokineticsImpairment of the AED to achieve optimal concentration levels at the action site, where it is mainly influenced by the liposolubility, absorption, metabolism and elimination of the drug.
PharmacodynamicsAll the factors that alter the action of the AED in their action sites (synapses, ion channels and receptors). There are at least three hypotheses that try to explain the pharmacodynamics: i) Alterations involving transporters (i.e. P-glycoprotein and the multiple drug resistance gene); ii) modifications of pharmacological targets due to genetic alterations related to the disease (i.e. genetic alterations in receptors or ion channels, structural alterations, autoimmunity and pharmacological interactions); iii) the intrinsic gravity model of epilepsy, in which a continuous severity of the disease is proposed and will be determined due to the medication response (i.e. etiology of epilepsy, type of seizures, electroencephalography or imaging studies, environmental influences or failure of AED).

[i] Adapted from Refs. 22,83,84. AED, antiepileptic drug.

Table II.

Variations in the main genes associated with the physiopathology of epilepsy.

Table II.

Variations in the main genes associated with the physiopathology of epilepsy.

Author, yearGeneObservations(Refs.)
Trivisano et al, 2015Cholinergic receptor nicotinic α2 subunitA mutation has been described in benign familial infantile seizures (c.1126 C>T).(85)
Wang et al, 2014; Villa et al, 2019Cholinergic receptor nicotinic α4 subunitThe novel mutation c.823 A>T has been identified in non-familial nocturnal frontal lobe epilepsy. The c.754T>C mutation is involved in autosomal dominant nocturnal frontal lobe epilepsy.(86,87)
Goldberg-Stern et al, 2009; Chen et al, 2018; Allen et al, 2014KCNQ2The novel mutation c.63-66delGGTG was associated with the diagnosis and prognosis of BFNS. Two mutations, c.811C>T and c.875T>C, were with early infantile encephalopathies.(8890)
Allen et al, 2014; Piro et al, 2019; Miceli et al, 2015KCNQ3The novel mutation c.914A>T was related to BFNS and was associated with a specific electroclinical pattern and favorable neurodevelopmental outcomes. The mutation c.989G>T was associated with intellectual disability in BFNS and the mutation c.989G>A was also associated with this type of epilepsy.(9092)
Lehman et al, 2017KCNQ5Three mutations, c.1343G>T, c.434T>G and c.1021C>A, were associated with epileptic encephalopathy and caused intellectual disability.(93)
Krepischi et al, 2010SCN1AThe deletion in 2q24, del(2)(q24.2q24.3), was associated with Dravet syndrome.(94)
Liang et al, 2017SCN2AThe novel mutation c.1270G>A was associated with early-onset epileptic encephalopathy and Rett-like features.(95)
Davidsson et al, 2008Gene cluster (SCN1A-SCN2A-SCN3A-SCN7A-SCN9A)The deletion in 2q24, del(2)(q24.3q31.1), was associated with severe epilepsy of infancy (Dravet syndrome) and was correlated with dysmorphic features and brain abnormalities.(96)
Zaman et al, 2018SCN3AMutations have been associated with early infantile epileptic encephalopathy.(97)
Yang et al, 2019; Butler et al, 2018; Orenstein et al, 2018; Zhang et al, 2017; Hernandez et al, 2017; Farnaes et al, 2017; Iqbal et al, 2018; Bhat et al, 2018; Le et al, 2017; Ishii et al, 2017 GABAAThe presence of various variants was associated with multiple seizure types, including focal seizures, generalized tonic-clonic seizures, myoclonic seizures, epileptic spasm and Dravet's syndrome, Ohtahara syndrome and West syndrome. De novo variants in GABRA2 (c.875C>A and c.1003A>C), GABRA5 (c.880G>C) and GABRB3 (c.5G>A, c.509T>G, c.914C>T and c.863C>A) were associated with severe early onset epilepsy. The de novo mutation c.789G>A in GABRA1 was associated with West syndrome. Mutational analysis of GABRG2 found an association between the presence of a single polymorphic site in exon 3 (AAC>AAT) and the absence epilepsy and generalized tonic clonic seizures. Variants in GABRG2 (588C>T) and GABRD (659G>A) were associated with juvenile myoclonic epilepsy and Lennox-Gastaut syndrome The mutation 965C>A and the polymorphism 15A>G in GABRA1 were associated with Lennox-Gastaut syndrome. The de novo mutation c.695G>A in GABRB3 was associated with Dravet syndrome The de novo mutation c.859A>C in GABRB2 was associated with early myoclonic encephalopathy.   (98107)
Butler et al, 2018; Yoo et al, 2017 GABABThe de novo variant c.902C>T was associated with intractable seizures and developmental delay. Numerous variants in GABBR2 were associated with Rett syndrome.(99,108)
Epi4K Consortium, 2016; Liu et al, 2018; Hayashida et al, 2018; Byers et al, 2016; Epperson et al, 2018; Du et al, 2017CACNA1AVarious mutations (c.2137G>A, c.5422G>C, c.4118C>T and c.301G>C) were associated with epileptic encephalopathies. Two novel mutations, c.2128 G>A and c.410A>G, were associated with Rett syndrome and absence epilepsy.(109114)
Heron et al, 2007CACNA1HOne hundred variants in exons 3–8 and 12–35 were detected in patients with various epileptic phenotypes, including childhood and juvenile absence, juvenile myoclonic and myoclonic astatic epilepsies, febrile seizures and temporal lobe epilepsy.(115)

[i] BFNS, benign familial neonatal seizures; CACNA, calcium voltage-gated channel subunit α; GABAA, γ-aminobutyric acid receptor; GABR, GABA receptor; KCNQ, potassium voltage-gated channel subfamily Q member; SCNA, sodium voltage-gated channel α subunit.

In the particular case of pharmacoresistant epilepsy, the most frequently studied polymorphisms are those associated with multidrug resistance genes (MDR): ATP-binding cassette subfamily B member 1 (ABCB1 or MDR1) and ATP-binding cassette subfamily C member 2 (ABCC2 or MRP2); SCN α subunits 1, 2 and 3 (SCN1, SCN2 and SCN3); and metabolizers of endogenous and xenobiotic substances, cytochromes P450 families 2 and 3 (CYP2 and CYP3). Additional details related to the findings of these studies are found in Table III.

Table III.

Genes associated with pharmacoresistant epilepsy.

Table III.

Genes associated with pharmacoresistant epilepsy.

Author, yearGeneMain observations(Refs.)
Yoshida et al, 2018BAF chromatin remodeling complex subunit BCL11AJapanese patients with epileptic encephalopathy.(116)
Kurian et al, 2018PCDH19Mutations were determined in pediatric patients with pharmacoresistant early childhood epilepsy.(117)
Ko et al, 2018SCN1A, CDKL5, KCNQ2, SCN2A and SCN8AMutations were determined in Asian patients with epileptic syndromes.(118)
Wang et al, 2018; Feng et al, 2018; Margari et al, 2018SCN1A and CYP3A4Polymorphisms were associated in Chinese children and adults with drug-resistant generalized epilepsy treated with valproic acid monotherapy. Also, polymorphisms in the SCN1A gene were associated with pharmacoresistance in Italian pediatric patients with epilepsy.(58,119,120)
Ajmi et al, 2018ABCB1Polymorphisms increased the risk of developing drug resistance in Tunisian epileptic patients.(121)
Abou El Ella et al, 2018GABR γ2 subunitPolymorphisms were associated with pharmacoresistance in Egyptian children with idiopathic generalized epilepsy.(122)
Skalski et al, 2017MDR1Polymorphisms were not associated with pharmacoresistance in Polish patients with refractory epilepsy.(123)
López-Garcia et al, 2017CYP2D6, CYP2C9, CYP2C19 and CYP3A4Polymorphisms were associated with refractory epilepsy in Mexican pediatric patients.(124)
Lv et al, 2017CACNA1AMutations were identified in Chinese patients with refractory progressive myoclonic epilepsy.(125)
Kozera-Kępniak et al, 2017Nuclear receptor subfamily 1 group I member 2Polymorphisms were associated with pharmacoresistance in Polish epileptic patients.(126)
Wang et al, 2017SCN8AMutations were identified in Chinese family with epilepsy.(127)
Parrini et al, 2017KCNA, KCNB, GABR and PNPOMutations were identified in Italian children diagnosed with several types of pharmacoresistant epilepsy.(128)
Kimizu et al, 2017SLC35A1A mutation was identified in a female Japanese pediatric patient with hepatic encephalopathy.(129)
Shen et al, 2017GABRG2Mutations were identified among Caucasian patients with epileptic encephalopathy.(130)
Zhang et al, 2017; Miao et al, 2017CDKL5, KCNQ2, KCNT1, KCNB1 SCN2A, SCN8A and SLC2A1Mutations were identified in Chinese patients diagnosed with pharmacoresistant epilepsy.(131,132)
Perucca et al, 2017SCN1AA variant was identified in an Australian patient with pharmacoresistant temporal lobe epilepsy.(133)
Guo et al, 2016Advanced glycosylation end-product specific receptorVariants were discovered in Chinese patients and associated with pharmacoresistant temporal lobe epilepsy.(134)
Stasiołek et al, 2016MDR1Variants were identified in Polish children diagnosed with refractory epilepsy.(135)
Abo El Fotoh et al, 2016SCN1A and CYP3A5Polymorphisms were associated with pharmacoresistance in pediatric patients with refractory idiopathic and symptomatic epilepsy.(136)
Xue and Lu, 2016ABCB1 and ABCC2Variants were identified in Chinese patients with refractory symptomatic epilepsy and associated with pharmacoresistance.(137)
Moen et al, 2016Potassium channel tetramerization domain containing 7, Sonic hedgehog signaling molecule, Smoothened, frizzled class receptor, Wnt16 and Wnt2A mutation was discovered in an Arab family with two children with pharmacoresistant progressive myoclonic epilepsy.(138)
Hildebrand et al, 2016Genes related to the Shh wayMutations were found in Caucasian patients with gelastic epilepsy.(139)
Hardies et al, 2016Synaptojanin 1Authors analyzed patients from three Caucasian families who had been treated for refractory seizures and progressive neurological diseases. Variants that abolished enzymatic activity were identified.(140)
Lionel et al, 2016Mediator complex subunit 23A variant was identified in a pediatric patient with refractory epilepsy.(141)
Balestrini et al, 2016TBC1 domain family member 24Mutations were analyzed in patients with pharmacoresistant epilepsy from 30 independent families. Pathogenic mutations were identified in the first conserved motif, some of which were associated with epileptic syndromes.(142)
Fahrner et al, 2016; Vanstone et al, 2016Dynamin 1-likeA mutation was determined in two American pediatric patients with epileptic encephalopathy and a variant was determined in a Canadian female pediatric patient diagnosed with epileptic encephalopathy.(143,144)
Janssen et al, 2016DNA polymerase γ catalytic subunitMutations were found in Belgian patients with refractory epilepsy and status epilepticus.(145)
Li et al, 2016GRIN2DA mutation was found in children with epileptic encephalopathy (one patient was African American and had European descent and the other patient was from Tunisia).(146)
Segal et al, 2016; Palmer et al, 2016SCN1A, PCDH19, SLC6A1 and SLC9A6Mutations were found in American children with pharmacoresistant epilepsy along with significant clinical abnormalities. An SLC6A1 mutation was specifically discovered in an American female pediatric patient with Doose syndrome.(147,148)
Inui et al, 2016Eukaryotic translation elongation factor 1 α2A mutation was found in Japanese patients with epileptic encephalopathy.(149)
Horta et al, 2015Major histocompatibility complex, class II, DR β1Variants were found in patients with pharmacoresistant temporal lobe epilepsy.(150)
Damiano et al, 2015Lamin B2A mutation was found in 10 patients with epilepsy who were members of an Arab-Palestinian family with a history of pharmacoresistant autosomal progressive myoclonic epilepsy with early-onset ataxia.(151)
Bene et al, 2015; Berghuis et al, 2015SCN1A and SCN8AMutations were identified in a Hungarian pediatric patient with Dravet syndrome. A mutation in the SCN8A gene was discovered in a Norwegian patient with epileptic encephalopathy.(152,153)
Damaj et al, 2015CACNA1AA mutation was determined in Franco-Canadian families with members diagnosed with epileptic encephalopathy.(154)
Guo et al, 2015KCNJ10Variants were determined in Chinese patients with refractory genetic generalized epilepsy.(155)
Liu et al, 2015Kelch-like ECH associated protein 1, nuclear factor, erythroid 2-like 2Variants were determined in patients with pharmacoresistant temporal lobe epilepsy and associated with pharmacoresistance.(156)
Emich-Widera et al, 2014ABCB1; also known as MDR1A variant was determined in Polish pediatric patients with refractory partial epilepsy.(157)
Venkateswaran et al, 2014GRIN2AA mutation was found in a female Canadian pediatric patient with refractory epilepsy.(158)
Picard et al, 2014DEP domain containing 5, GATOR1 subcomplex subunitMutations were identified in Caucasian individuals diagnosed with dominant nocturnal temporal lobe epilepsy.(159)
Martin et al, 2014KCNQ2, SCN2A and KCNT1Mutations were identified in Caucasian patients with severe early-onset epilepsy.(160)
Seven et al, 2014; Escalante-Santiago et al, 2014MDR and MRP2Variants were identified among Turkish pediatric patients with refractory partial and generalized epilepsy. Variants in this genes were also identified in 22 Mexican children diagnosed with refractory partial complex epilepsy(161,162)
Seven et al, 2014CYP2C9, CYP2C1 and CYP2D6Polymorphisms were determined in Turkish pediatric patients with partial epilepsy or refractory generalized epilepsy and associated with pharmacoresistance.(163)
Ma et al, 2014SCN1A, SCN2A and ABCC2Variants were analyzed in Chinese patients with refractory partial or generalized epilepsy and associated with pharmacoresistance.(164)
He et al, 2013C-C motif chemokine ligand 2A variant was determined in Chinese children with refractory partial and generalized epilepsy and associated with pharmacoresistance.(165)
Serino et al, 2013KCNQ2A mutation was found in an Italian pediatric patient with epileptic encephalopathy.(166)
Emich-Widera et al, 2013CYP3A5 and MDR1A variant was found in Polish children diagnosed with refractory epilepsy.(167)
Veeramah et al, 2013CDK, chloride voltage-gated channel 1, SCN, KCNHMutations were identified in the families of American children diagnosed with epileptic encephalopathy.(168)
Fragaki et al, 2013Glutamate receptor, metabotropic 6A mutation was identified in the family of two French pediatric patients with refractory epilepsy.(169)
Subenthiran et al, 2013; Qu et al, 2012ABCB1 and ABCC2Variants were associated with pharmacoresistance in Malaysian (49.7%), Chinese (26.8%) and Indian (23.5%) adult patients with epilepsy with complex partial seizures treated with carbamazepine. Variants were also found in Chinese patients with partial idiopathic or refractory cryptogenic epilepsy and associated with pharmacoresistance.(170,171)
Dimova et al, 2012PCDH19A mutation was identified in one female pediatric patient with refractory epilepsy.(172)
Sayyah et al, 2011ABCB1Variants were identified in Iranian children and adults diagnosed with refractory idiopathic, cryptogenic and symptomatic epilepsy and associated with pharmacoresistance.(173)
Lakhan et al, 2011CYP2C9 and CYP2C19Variants were determined in Indian patients with refractory idiopathic and symptomatic epilepsy.(174)
Kumari et al, 2011SCN2, GABRVariants were identified in Indian patients diagnosed with refractory epilepsy and associated with pharmacoresistance.(175)
Kwan et al, 2011ABCC2, ABCC5 and ABCG2Polymorphisms were analyzed in Chinese patients with refractory epilepsy.(176)
Kim et al, 2011SLC6A11Variants were found in Korean patients with refractory idiopathic, symptomatic and cryptogenic epilepsy and associated with pharmacoresistance.(177)
Meng et al, 2011SLC6A11Variants were found in Chinese patients with drug-resistant focal and generalized epilepsy and treated with carbamazepine monotherapy.(178)
Alpman et al, 2010MDR1Variants were determined in Turkish pediatric patients with refractory generalized or partial epilepsy.(179)
Maleki et al, 2010ABCB1Variants were determined in Iranian pediatric and adults with refractory idiopathic and symptomatic epilepsy and associated with pharmacoresistance.(180)
Di Bonaventura et al, 2009Leucine rich glioma inactivated 1A mutation was determined in an Italian family with refractory autosomal dominant lateral temporal lobe epilepsy.(181)
Jang et al, 2009SCN1A, SCN1B and SCN2AVariants were determined in Korean patients with refractory symptomatic and idiopathic epilepsy.(182)
Vahab et al, 2009; Kwan et al, 2009ABCB1Variants were determined in Indian pediatric patients with refractory epilepsy, in Chinese patients with refractory idiopathic, symptomatic or cryptogenic epilepsy and were associated with pharmacoresistance.(183,184)
Kauffman et al, 2009SLC6A4A variant was determined in adult Argentine patients with drug-resistant mesial temporal lobe epilepsy and associated with pharmacoresistance.(185)
Lakhan et al, 2009SCN1A and SCN2AA variant was determined in Indian patients with drug-resistant epilepsy and associated with pharmacoresistance.(186)
Bahi-Buisson et al, 2008CDKL5Mutations were identified in female Caucasian patients with Rett syndrome with refractory epilepsy.(187)
Kwan et al, 2008SCN1A, SCN2A and SCN3AVariants were identified in Chinese patients with refractory symptomatic, idiopathic and cryptogenic epilepsy and associated with pharmacoresistance.(188)
Elia et al, 2008CDKL5Mutations were analyzed in Italian children with profound mental retardation and seizures (myoclonic or tonic spasms) that were refractory to treatment. A total of three de novo missense mutations were identified in the gene.(189)
Abe et al, 2008SCN1AA variant was identified in Japanese drug-responsive epileptic patients and associated with pharmacoresistance.(190)
Shahwan et al, 2007; Kwan et al, 2007; Hung et al, 2007ABCB1Variants were determined in Irish and in Chinese adult patients with refractory epilepsy. A logistic model revealed that the interaction of the polymorphisms was associated with drug-resistant epilepsy after adjusting for etiology and type of epilepsy in Chinese patients.(191193)
Leschziner et al, 2007ralA binding protein 1Variants were genotyped in English patients with drug-resistant epilepsy associated with pharmacoresistance. A total of six SNPs were genotyped displaying an association between rs329017 and the risk of pharmacoresistance(194)
Seo et al, 2006; Hung et al, 2005ABCB1Variants were genotyped in patients with epilepsy and associated with pharmacoresistance. Haplotype analysis indicated that drug-resistant patients tended to display CGC/CGC, CGC/TGC, CGC/TTT and TGC/CGT combinations.(195,196)
Mills et al, 2005PNPOMutations in Asian families with epileptic encephalopathy were determined.(197)
Buono et al, 2004KCNJ10A variant was detected in Caucasian patients diagnosed with refractory mesial temporal lobe epilepsy and associated with pharmacoresistance.(198)
Siddiqui et al, 2003ABCB1A variant was identified in English patients with refractory epilepsy and associated with pharmacoresistance.(199)
Gambardella et al, 2003GABAB receptor 1A variant was determined in Italian patients with temporal lobe epilepsy and associated with pharmacoresistance.(200)

[i] ABCB1, ATP binding-cassette subfamily B member 1; ABCC2, ATP binding-cassette subfamily C member 2; ABCC5, ATP binding-cassette subfamily C member 5; ABCG2, ATP binding-cassette subfamily G member 2; CACNA1A, calcium voltage-gated channel subunit α 1A; CYP2C1, cytochrome P450 family 2 subfamily C member 1; CYP2C19, cytochrome P450 family 2 subfamily C member 19; CYP2C9, cytochrome P450 family 2 subfamily C member 9; CYP2D6, cytochrome P450 family 2 subfamily D member 6; CYP3A5, cytochrome P450 family 3 subfamily A member 5; CYP3A4, cytochrome P450 family 3 subfamily A member 4; GABA, γ-aminobutyric acid receptor; GABR, γ-aminobutyric acid type A receptor; GRIN2, glutamate ionotropic receptor NMDA-type subunit 2; KCNA, potassium voltage-gated channel subfamily A; KCNB, potassium voltage-gated channel subfamily B; KCNH, potassium voltage-gated channel subfamily H; KCNJ10, potassium inwardly rectifying channel subfamily J member 10; KCNT1, potassium sodium-activated channel subfamily T member 1; MDR1, ABC transporter B family member 1; PNPO, pyridoxamine 5′-phosphate oxidase; SCN2A, sodium voltage-gated channel α subunit 2; SCN3A, sodium voltage-gated channel α subunit 3; SCN8A, sodium voltage-gated channel α subunit 8; SCN1B, sodium voltage-gated channel β subunit 1; SLC35A1, solute carrier family 35 member A2; SLC6A1, solute carrier family 6 member 1; SLC6A4, solute carrier family 6 member 4; SLC6A11, solute carrier family 6 member 11; SLC9A6, solute carrier family 9 member 6.

The role of genetic variants in the diagnosis and treatment of pharmacoresistant epilepsy

Currently, a patient's medical history and EEG results are used to diagnose the type of seizure, but they must be interpreted with caution so that diagnostic errors are not made; this suggests the use of complementary studies (38). Emerging genomic technologies, high-throughput screening and chip technologies have accelerated our understanding of the genetic makeup of epilepsy (34); for instance, the identification of mutations or polymorphisms in specific genes, such as those that encode ion channels, aforementioned, that are mainly expressed in brain neurons, specific neurotransmitter receptors and molecules that have functions in intercellular communication (39). For example, a study by Wang et al (40) investigated the genetic etiology of epilepsy in a cohort of 120 children with unexplained epilepsy using whole-exome sequencing (WES); it was found that the pathogenic variant c.1174G>A in the KCN subfamily D member 3 (KCND3) gene may be responsible for a broader phenotypic spectrum than was previously thought, including infantile epileptic encephalopathy. In addition, this study discovered that the glutamate receptor, ionotropic glutamate ionotropic receptor NMDA-type subunit 1 (GRIN1) and hyperpolarization-activated cyclic nucleotide-gated potassium channel 1 (HCN1), were candidate gene variants (c.2530C>T and c.1138A>T for GRIN1 and HCN1, respectively) for the Dravet and Dravet-like phenotypes (40). In intractable epilepsy and other mental disabilities, WES identified de novo variants in the Bernardinelli-Seip congenital lipodystrophy 2 (BSCL2) gene in two patients (41), of which one of the variants (c.985C>T) has been observed in other populations of epilepsy and developmental regression, regressive autism spectrum disorder, motor stereotypies, lower limb hypertonia and frontal lobe syndrome (41). As it was discovered that BSCL2 serves a role in neuronal function, it was suggested to be a potential candidate gene for epileptogenesis (41).

In pyridoxine-dependent epilepsy (PDE), despite seizure control, ≥75% of patients experience intellectual disability and developmental delay, which emphasizes the importance of early diagnosis. Genetic tests are increasingly being used as first-level tests for epileptic encephalopathies, which aim to provide a general description of the mutations in the aldehyde dehydrogenase 7 family member A1 that causes PDE (42). Epilepsy with myoclonic-atonic seizures (EMAS) accounts for 1–2% of all childhood onset epilepsies (43). EMAS has been demonstrated to have an underlying genetic component and several genes have been associated with this disease, such as SCN1A, SCN2A, CHD2, STX1B, SLC2A1, SLC6A1, POLG1, NRXN1, PIGN, CSNK2A1, GABRG2 and GABRB3; however, the genetic basis for this disorder remains unknown and the diagnostic potential of genetic tests remains low. This could be explained by the lack of several of the genes that may be associated with EMAS in the most commonly ordered epilepsy panels, although some have recently been added (4347). Furthermore, a study conducted by Ortega-Moreno et al (48) analyzed a multigenic panel of 87 PWE and developmental delay, including classified and unclassified epileptic encephalopathies, epileptic spasms, severe myoclonic epilepsy of infancy, Lennox-Gastaut and Landau-Kleffner syndromes; they found mutations in various genes, such as potassium voltage-gated channel subfamily Q member 2 (KCNQ2), syntaxin binding protein 1, UDP-N-acetylglucosaminyltransferase subunit, cyclin dependent kinase-like 5 (CDKL5), protocadherin 19, SCN1A, CHD2, SLC2A1, synaptic Ras GTPase activating protein 1, aristaless related homeobox, DNA polymerase gamma, catalytic subunit and GRIN1. Although a high proportion of these patients had unclassified epilepsies, the results supported the use of the multigene epilepsy panel because it offered rapid testing with a good diagnostic efficiency (48).

It is hypothesized that genetic variants may also contribute to the efficacy of drug treatments for epilepsy; for example, adverse or toxic reactions, teratogenic risk in pregnancy, as well as long-term outcomes have been observed among PWE (4963). Consistent with the findings in numerous other disorders with complex genetic backgrounds, the associated genetic variants that have been successfully verified are limited. Nevertheless, it is likely that new techniques and improved research approaches will increase this number in the near future (49). In recent studies, the association between genetic polymorphisms, treatment responses in epilepsy and AEDs reactions (toxic, adverse or those related with its efficacy) have been investigated; it was reported that polymorphisms in the human leukocyte antigen (HLA) gene were associated with severe cutaneous adverse AED reactions (50), and polymorphisms in a number of other genes, including ABCB1, ABCC2, GABRA6, GABRG2, CYP2C9, CYP3A4, UDP-glucuronosyltransferase (UGT)1A1, UGT1A4, UGT1A6, UGT2B7, SCN2A and SCN1A, have been associated with the concentration, response and efficacy of some of the most commonly used AEDs in clinical practice, including carbamazepine, oxcarbazepine, phenytoin, lamotrigine and valproic acid (5163). Esmaeilzadeh et al (50) reported an association between HLA polymorphisms and severe cutaneous adverse reactions (SCARs) induced by drugs; in this study, 61 patients with diverse SCARs were recruited, and it was found that the hypersensitivity to different AEDs, including phenytoin, carbamazepine, valproic acid, topiramate and lamotrigine, was associated with HLA-A gene polymorphisms. Berghuis et al (64) recruited 1,328 adult PWE who had received oxcarbazepine (n=1,031) and carbamazepine (n=297) and performed a genome-wide association study to demonstrate the association between genetic factors and sodium levels and AED metabolism. The authors did not observe significant associations between sodium levels and other clinical variables, but in relation to carbamazepine metabolism, they observed a significant association with the intronic rs2234922 polymorphism in the epoxide hydrolase 1 gene. Furthermore, the same authors reported in 2017 that carbamazepine and oxcarbazepine induced hyponatremia in those with epilepsy (65); in this study, 1,782 adult patients who carbamazepine (n=1,424) and oxcarbazepine (n=358) were recruited; using an electronic database designed for pharmacogenomics studies, it was found that sodium levels were significantly associated with serum levels of carbamazepine (P<0.001) and oxcarbazepine (P=0.001), whilst age, sex and the number of concomitantly used AEDs did not influence this association. Serum levels of carbamazepine [Odds Ratio (OR)=1.2; 95% CI=1.12–1.28; P<0.001] and oxcarbazepine (OR=1.06; 95% CI=1.02–1.1; P=0.001) were significantly associated with hyponatremia. The co-treatment and the sequential use of carbamazepine and oxcarbazepine were also related to severe hyponatremia (65). McCormack et al (66) recruited patients with maculopapular exanthema (MPE) associated with AED use (all aromatic AEDs: n=259 European and n=116 Han Chinese patients; carbamazepine: n=95 European and n=85 Han Chinese patients; lamotrigine: n=118 European and n=16 Han Chinese patients; phenytoin: n=52 European and n=22 Han Chinese patients) and 1,321 controls and performed a genome-wide association to analyze the association between AED use and MPE. It was found that within the European population, variations in HLA-A*31:01 were significantly associated with carbamazepine-induced MPE (OR=5.5; 95% CI=3–10; P=1.47×10−10). Regarding phenytoin use, a significant association was identified between the rs78239784 polymorphism and an intronic variant of the complement factor H-related 4 gene in the European population (OR=8.8; 95% CI=4–19; P=2.94×10−10) (66). Bai et al (67) observed that VPA induced obesity in PWE, as following the recruitment of 225 Chinese Han patients with epilepsy receiving VPA treatment, 19.6% were found to be obese. The authors also found genotypic associations of rs1194197 in the CD36 gene and rs10865710 in the peroxisome proliferator-activated receptor γ gene following weight gain. In another study conducted by Li et al (68), associations between the rs1137101 polymorphism in the leptin receptor (P<0.001), the rs1800497 polymorphism in the ankyrin repeat kinase domain-containing 1 (P=0.017) and the rs10789038 polymorphism in AMP protein kinase (P=0.02) with valproic acid-induced weight gain were observed in 212 PWE (68). Wang et al (69) discovered that some polymorphisms were associated with the adverse effects of valproic acid in Chinese PWE by direct sequencing; following the recruitment of 254 Chinese PWE that received valproic acid monotherapy, a correlation was identified between CYP2C9 and acyl-coenzyme A synthetase 2A (ACSM2A) gene polymorphisms with serum alanine aminotransferase and aspartate aminotransferase levels (P<0.03) indicating that these gene polymorphisms can be used to identify liver dysfunction (69).

The therapeutic effect of valproic acid among children with focal epilepsy (89 children) was also studied, and the results identified 66 single nucleotide polymorphisms (SNPs) that were involved in the metabolism and transport of valproic acid target receptors (54); however, among the children with focal seizures, the selected genetic polymorphisms were not significantly associated with the response to valproic acid. Nonetheless, three variants of GABRA6 (rs9313892, rs4921195 and rs424740) demonstrated the potential to be associated with the response to valproic acid (54). In addition, although polymorphisms in the SCN1A gene are thought to influence the efficacy of carbamazepine and phenytoin, Manna et al (70) found that the rs3812718 variant in SCN1A was not associated with the response to carbamazepine in patients with focal epilepsy. It has been reported that disorders related to changes in the KCNQ2 gene included both benign seizure disorders and early onset epileptic encephalopathies (EOEE), especially the latter, which includes patients who present refractory seizures following standard AED treatment and development delay (7173). Kuersten et al (74) conducted a systematic review (52 studies including data from 217 patients), in which they analyzed AEDs in KCNQ2-related epilepsies; it was discovered that seizures associated with KCNs could be controlled upon treatment with carbamazepine, lamotrigine, oxcarbazepine, phenytoin, valproic acid, levetiracetam, topiramate, phenobarbital, piracetam, vigabatrin, clonazepam, diazepam and midazolam in patients with benign infantile or neonatal seizures (n=133 patients, including 74 who were seizure-free with monotherapy, 4 who were unsuccessfully treated with monotherapy, 11 who were seizure-free with polytherapy, 4 patients with no response to polytherapy or any treatmet and 40 showed seizure cessation spontaneously without AEDs). Moreover, the results also demonstrated that moderate control of seizures was achieved with the use of carbamazepine, oxcarbazepine, lamotrigine, lacosamide, phenytoin, phenobarbital, valproic acid, topiramate, levetiracetam, retigabine, zonisamide, sultiame, ethosuximide, acetazolamide, clonazepam, diazepam, clobazam, nitrazepam and midazolam in patients with EOEE (n=84, including 48 who were seizure-free with monotherapy, 12 that did not respond to monotherapy, 20 who were seizure-free following polytherapy and 4 patients exhibited seizure reduction without AED). Phenobarbital was the most common prescribed monotherapy in the majority of patients with benign seizures and EOEE (n=65 in benign seizures and n=35 in patients with EOEE); however, the use of sodium channel blockers, such as carbamazepine, lamotrigine, oxcarbazepine and phenytoin, led to seizure cessation in the majority of patients with benign seizures and EOEE (n=21 benign seizures and n=45 with EOEE). With regards to the genetics, 25.6 and 67.9% of patients with benign seizures and EOEE, respectively, were reported to have de novo mutations, including missense, frameshift, splice site, deletion and truncation mutations. However, sparse systemic data are available on the response of treatment in KCNQ2-related epilepsy in larger cohorts (74), which limit our ability to comment on the efficacy of personalized medicine approaches to treat the large number of newly discovered genetic channelopathies.

The effects of SNPs in three KCN genes, including KCNA1 (rs112561866, rs2227910 and rs7974459), KCNA2 (rs3887820) and KCNV2 (rs7029012, rs10967705 and rs10967728), and their association with the susceptibility to epilepsy and their ability to respond to AEDs (carbamazepine for partial epilepsy and valproic acid for generalized epilepsy) was analyzed in a pharmacogenetic cohort of 595 patients (75). The results suggested that KCNA1, KCNA2 and KCNV2 did not influence the susceptibility of the disease or the capacity to respond to drugs (75). Mutations in the SCN2A gene have also been associated with neonatal seizures and a wide number of epileptic syndromes (76,77). Recently, an association between rs17183814 in SCN2A and the function of oxcarbazepine was demonstrated in a cohort of 218 patients; the results indicated that the presence of the SNP was associated with higher oxcarbazepine maintenance doses in patients with lower body weights and lower oxcarbazepine maintenance doses in patients who were overweight (51). In a cohort of 201 patients treated with valproic acid, an association was reported between the presence of the SNP rs230416 in SCN2A and the response to the drug (78). Similarly, another study reported an association between the SNPs in SCN2A (rs2304016) and SCN3B (rs3851100) and AED responsiveness in a cohort of 595 patients who were treated with valproic acid for generalized epilepsy and carbamazepine for partial epilepsy; the results demonstrated that none of these SNPs were significantly associated with a response to the AED (79).

Previous studies have indicated that to improve the efficacy and safety of epilepsy treatments, it is necessary to conduct studies to identify the following: i) The possible influence of some of the phenotypic characteristics of patients in response to the treatments; ii) the relationship between the number and type of seizures; iii) the differences in drug transporting protein activity or receptor activity caused by the genetic variations; iii) the impact of the presence of a genetic variant on the functionality of transporting proteins and AED target proteins; iv) the effects of potential alterations in target proteins, owing to a genetic variant, in the mechanisms of action of an AED; and v) factors that may contribute to susceptibility and treatment outcomes.

Clinical implications of genetic variants in pharmacoresistant epilepsy

Personalized medicine is treatment of patients with therapy aimed at targeting their specific pathophysiology; however, currently, this has limited applications in clinical practice. Advancements in genetic epilepsy models and deep phenotyping techniques have the potential to revolutionize translational research, and will bring precision medicine to the forefront of clinical practice (80). In a needs assessment aimed at identifying the clinical challenges faced by physicians in diagnosing and treating children with epilepsy in Germany, Spain and the United States, it was reported that the main challenges were the application of guidelines in clinical practice, the identification of epilepsy and epileptic events, the integration of genetic tests into clinical practice, the integration of non-pharmacological treatments, the transition from pediatric to adult care and the participation and commitment of caregivers (80). These findings may support neuropediatricians who wish to specialize in epileptology to address these identified challenges based through precision medicine treatments (80). As we continue to gain an improved understanding of the true complexity underlying the physiopathology of genetic epilepsy and the identification of factors that are involved in phenotypic variations, it will be easier to address and understand genotype-phenotype correlations (81).

Previous studies on genetic epilepsy syndromes have provided insight into the mechanisms of epileptogenesis, and have suggested roles for a number of genes with different functions, including ion channel proteins and those associated with the synaptic vesicle cycle and energy metabolism (82). In addition, advanced genomic technologies, high-throughput sequencing and molecular diagnostics are increasingly improving our understanding of the genetic architecture in epilepsy, and molecular confirmation may influence the treatment prescribed for some monogenic epilepsies. Moreover, it is of crucial importance that genetic methods that are able to analyze all known genes at a reasonable cost be developed to discover novel therapeutic options and to implement individualized precision medical treatment regimens (82).

Conclusions and future perspectives

Genetic variations in the most common genes that encode channels, transporters, drug-metabolizing enzymes and receptors have been discussed in this Review with regards to their association with drug-resistant epilepsy, including: Sodium voltage-gated channels SCN1A, SCN2A, SCN3A, SCN8A and SCN1B); potassium voltage-gated channels (KCNA1, KCNA2, KCNB1, KCND7, KCNH5, KCNJ10, KCNQ2 and KCNT1); calcium voltage-gated channel subunit α1 H; ATP binding-cassette transporters (ABCB1, ABCC2, ABCC5 and ABCG2); mitochondrial transporter family members (SLC2A1, SLC6A1, SLC6A4, SLC6A11, SLC9A6, SLC25A22 and SLC35A2); drug-metabolizing enzymes (CYP2C1, CYP2C9, CYP2C19, CYP2D6, CYPP3A4 and CYP3A5); CDKL5; and GABA receptors (GABRA1 and GABBR1). These data may prove useful for future studies of drug resistance in epilepsy and may contribute to the generation of new diagnostic methods. In addition, these methods could subsequently support the development of improved treatment regimens, including novel pharmacological targets and pharmacological therapeutics. Overall, these findings may also improve the application of more personalized therapies, which would lead to the reduction in treatment and medical care costs, and increase the quality of life for patients and caregivers.

Acknowledgements

The technical assistance of Dr Perla Michelle Martínez of the Department of Public Health Sciences, Austin, University of Texas (TX, USA).

Funding

This work was supported by the E022 Program of the National Institute of Pediatrics (protocol no. 041/2018) and from CONACyT (Mexico) Doctoral Program of Medical, Dental and Health Sciences, Clinical Experimental Health Research, The National Autonomous University of Mexico (grant no. 706968) and the SNI-CONACYT Fellows (NC-R, LC-A, DO-C and SG-M).

Availability of data and materials

Not applicable.

Authors' contributions

LCA, DLPL, SGM and IIM wrote and revised the manuscript; DOC critically revised and corrected the manuscript; and NCR conceived the idea for the review, collected and interpreted the studies included, reviewed the manuscript and contributed significantly to the writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

PWE

people with epilepsy

EMAS

epilepsy with myoclonic-atonic seizures

EOEE

early onset epileptic encephalopathies

References

1 

Camfield P and Camfield C: Incidence, prevalence and aetiology of seizures and epilepsy in children. Epileptic Disord. 17:117–123. 2015.PubMed/NCBI

2 

Carod-Artal FJ and Vázquez-Cabrera CB: An anthropological study about epilepsy in native tribes from Central and South America. Epilepsia. 48:886–893. 2007. View Article : Google Scholar : PubMed/NCBI

3 

López González FJ, Rodríguez Osorio X, Gil-Nagel Rein A, Carreño Martínez M, Serratosa Fernández J, Villanueva Haba V, Donaire Pedraza AJ and Mercadé Cerdá JM: Drug-resistant epilepsy: Definition and treatment alternatives. Neurologia. 30:439–446. 2015.(In English, Spanish). View Article : Google Scholar : PubMed/NCBI

4 

Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, Engel J, Forsgren L, French JA, Glynn M, et al: ILAE official report: A practical clinical definition of epilepsy. Epilepsia. 55:475–482. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Golyala A and Kwan P: Drug development for refractory epilepsy: The past 25 years and beyond. Seizure. 44:147–156. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Falco-Walter JJ, Scheffer IE and Fisher RS: The new definition and classification of seizures and epilepsy. Epilepsy Res. 139:73–79. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Scheffer IE, Berkovic S, Capovilla G, Connolly MB, French J, Guilhoto L, Hirsch E, Jain S, Mathern GW, Moshé SL, et al: ILAE classification of the epilepsies: Position paper of the ILAE commission for classification and terminology. Epilepsia. 58:512–521. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Waldbaum S and Patel M: Mitochondria, oxidative stress, and temporal lobe epilepsy. Epilepsy Res. 88:23–45. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Carmona-Aparicio L, Pérez-Cruz C, Zavala-Tecuapetla C, Granados-Rojas L, Rivera-Espinosa L, Montesinos-Correa H, Hernández-Damián J, Pedraza-Chaverri J, Sampieri A III, Coballase-Urrutia E, et al: Overview of Nrf2 as therapeutic target in epilepsy. Int J Mol Sci. 16:18348–18367. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Cardenas-Rodriguez N, Coballase-Urrutia E, Pérez-Cruz C, Montesinos-Correa H, Rivera-Espinosa L, Sampieri A III and Carmona-Aparicio L: Relevance of the glutathione system in temporal lobe epilepsy: Evidence in human and experimental models. Oxid Med Cell Longev. 2014:7592932014. View Article : Google Scholar : PubMed/NCBI

11 

Floriano-Sánchez E, Brindis F, Ortega-Cuellar D, Ignacio-Mejia I, Moreno-Arriola E, Romero-Morelos P, Ceballos-Vasquez E, Cordova-Espinoza MG, Arregoitia-Sarabia CK, Sandoval- Pacheco R, et al: Differential gene expression profile induced by valproic acid (VPA) in pediatric epileptic patients. Genes (Basel). 9(pii): E3282018. View Article : Google Scholar : PubMed/NCBI

12 

Köhling R: Voltage-gated sodium channels in epilepsy. Epilepsia. 43:1278–1295. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Tan NC, Mulley JC and Berkovic SF: Genetic association studies in epilepsy: ‘The truth is out there’. Epilepsia. 45:1429–1442. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Semah F and Ryvlin P: Can we predict refractory epilepsy at the time of diagnosis? Epileptic Disord. 7 (Suppl 1):S10–S13. 2005.PubMed/NCBI

15 

Fishman MC and Porter JA: Pharmaceuticals: A new grammar for drug discovery. Nature. 437:491–493. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Kwan P, Arzimanoglou A, Berg AT, Brodie MJ, Allen Hauser W, Mathern G, Moshé SL, Perucca E, Wiebe S and French J: Definition of drug resistant epilepsy: Consensus proposal by the ad hoc task force of the ILAE commission on therapeutic strategies. Epilepsia. 51:1069–1077. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Kwan P and Brodie MJ: Early identification of refractory epilepsy. N Engl J Med. 342:314–319. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Brodie MJ, Barry SJ, Bamagous GA, Norrie JD and Kwan P: Patterns of treatment response in newly diagnosed epilepsy. Neurology. 78:1548–1554. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Picot MC, Baldy-Moulinier M, Daurès JP, Dujols P and Crespel A: The prevalence of epilepsy and pharmacoresistant epilepsy in adults: A population-based study in a Western European country. Epilepsia. 49:1230–1238. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Ngugi AK, Bottomley C, Kleinschmidt I, Sander JW and Newton CR: Estimation of the burden of active and life-time epilepsy: A meta-analytic approach. Epilepsia. 51:883–890. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Zavala-Tecuapetla C, Sampieri A III, Calderón-Guzmán D and Carmona-Aparicio L: Principales mecanismos de acción farmacológica en las crisis epilépticas díficiles de estabilizar. Acta Pediátr México. 32:125–127. 2011.

22 

Espinosa-Jovel CA and Sobrino-Mejia FE: Drug resistant epilepsy. Clinical and neurobiological concepts. Rev Neurol. 61:159–166. 2015.(In Spanish). PubMed/NCBI

23 

Luoni C, Bisulli F, Canevini MP, De Sarro G, Fattore C, Galimberti CA, Gatti G, La Neve A, Muscas G, Specchio LM, et al: Determinants of health-related quality of life in pharmacoresistant epilepsy: Results from a large multicenter study of consecutively enrolled patients using validated quantitative assessments. Epilepsia. 52:2181–2191. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Argumosa A and Herranz J: La repercusión económica de las enfermedades crónicas: El coste de la epilepsia infantil en el año 2000. Bol Pediatr. 41:23–29. 2001.

25 

Martin K, Jackson CF, Levy RG and Cooper PN: Ketogenic diet and other dietary treatments for epilepsy. Cochrane Database Syst Rev. 2:CD0019032016.PubMed/NCBI

26 

Chudomelova L, Scantlebury MH, Raffo E, Coppola A, Betancourth D and Galanopoulou AS: Modeling new therapies for infantile spasms. Epilepsia. 51 (Suppl 3):S27–S33. 2010. View Article : Google Scholar

27 

Baumgartner C, Koren JP, Britto-Arias M, Zoche L and Pirker S: Presurgical epilepsy evaluation and epilepsy surgery. F1000Res. 8(pii): F1000 Faculty Rev-1818. 2019.PubMed/NCBI

28 

Ułamek-Kozioł M, Czuczwar SJ, Januszewski S and Pluta R: Ketogenic diet and epilepsy. Nutrients. 11(pii): E25102019. View Article : Google Scholar : PubMed/NCBI

29 

Boon P, De Cock E, Mertens A and Trinka E: Neurostimulation for drug-resistant epilepsy: A systematic review of clinical evidence for efficacy, safety, contraindications and predictors for response. Curr Opin Neurol. 31:198–210. 2018. View Article : Google Scholar : PubMed/NCBI

30 

García-Contreras F, Constantino-Casas P, Castro-Ríos A, Nevárez-Sida A, Estrada Correa Gdel C, Carlos Rivera F, Guzmán-Caniupan J, Torres-Arreola Ldel P, Contreras- Hernández I, Mould-Quevedo J and Garduño-Espinosa J: Direct medical costs for partial refractory epilepsy in Mexico. Arch Med Res. 37:376–383. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Aguirre-Velázquez CG: Report from a survey of parents regarding the use of cannabidiol (Medicinal cannabis) in Mexican children with refractory epilepsy. Neurol Res Int. 2017:29857292017. View Article : Google Scholar : PubMed/NCBI

32 

Younus I and Reddy DS: A resurging boom in new drugs for epilepsy and brain disorders. Expert Rev Clin Pharmacol. 11:27–45. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Tang F, Hartz AMS and Bauer B: Drug-resistant epilepsy: Multiple hypotheses, few answers. Front Neurol. 8:3012017. View Article : Google Scholar : PubMed/NCBI

34 

Myers CT and Mefford HC: Advancing epilepsy genetics in the genomic era. Genome Med. 7:912015. View Article : Google Scholar : PubMed/NCBI

35 

Depondt C: The potential of pharmacogenetics in the treatment of epilepsy. Eur J Paediatr Neurol. 10:57–65. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Mulley JC and Dibbens LM: Genetic variations and associated pathophysiology in the management of epilepsy. Appl Clin Genet. 4:113–125. 2011.PubMed/NCBI

37 

Noebels J: Pathway-driven discovery of epilepsy genes. Nat Neurosci. 18:344–350. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Devinsky O: Patients with refractory seizures. N Engl J Med. 340:1565–1570. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Pal DK and Greenberg DA: Evaluating genetic heterogeneity in complex disorders. Hum Hered. 53:216–226. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Wang J, Wen Y, Zhang Q, Yu S, Chen Y, Wu X, Zhang Y and Bao X: Gene mutational analysis in a cohort of Chinese children with unexplained epilepsy: Identification of a new KCND3 phenotype and novel genes causing Dravet syndrome. Seizure. 66:26–30. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Fernández-Marmiesse A, Sánchez-Iglesias S, Darling A, O'Callaghan MM, Tonda R, Jou C and Araújo-Vilar D: A de novo heterozygous missense BSCL2 variant in 2 siblings with intractable developmental and epileptic encephalopathy. Seizure. 71:161–165. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Coughlin CR II, Swanson MA, Spector E, Meeks NJL, Kronquist KE, Aslamy M, Wempe MF, van Karnebeek CDM, Gospe SM Jr, Aziz VG, et al: The genotypic spectrum of ALDH7A1 mutations resulting in pyridoxine dependent epilepsy: A common epileptic encephalopathy. J Inherit Metab Dis. 42:353–361. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Angione K, Eschbach K, Smith G, Joshi C and Demarest S: Genetic testing in a cohort of patients with potential epilepsy with myoclonic-atonic seizures. Epilepsy Res. 150:70–77. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Wolff M, Johannesen KM, Hedrich UBS, Masnada S, Rubboli G, Gardella E, Lesca G, Ville D, Milh M, Villard L, et al: Genetic and phenotypic heterogeneity suggest therapeutic implications in SCN2A-related disorders. Brain. 140:1316–1336. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Kang JQ and Macdonald RL: Molecular pathogenic basis for GABRG2 mutations associated with a spectrum of epilepsy syndromes, from generalized absence epilepsy to dravet syndrome. JAMA Neurol. 73:1009–1016. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Møller RS, Wuttke TV, Helbig I, Marini C, Johannesen KM, Brilstra EH, Vaher U, Borggraefe I, Talvik I, Talvik T, et al: Mutations in GABRB3: From febrile seizures to epileptic encephalopathies. Neurology. 88:483–492. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Johannesen KM, Gardella E, Linnankivi T, Courage C, de Saint Martin A, Lehesjoki AE, Mignot C, Afenjar A, Lesca G, Abi-Warde MT, et al: Defining the phenotypic spectrum of SLC6A1 mutations. Epilepsia. 59:389–402. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Ortega-Moreno L, Giráldez BG, Soto-Insuga V, Losada-Del Pozo R, Rodrigo-Moreno M, Alarcón-Morcillo C, Sánchez- Martín G, Díaz-Gómez E, Guerrero-López R, Serratosa JM, et al: Molecular diagnosis of patients with epilepsy and developmental delay using a customized panel of epilepsy genes. PLoS One. 12:e01889782017. View Article : Google Scholar : PubMed/NCBI

49 

Steinlein OK: Gene polymorphisms and their role in epilepsy treatment and prognosis. Naunyn Schmiedebergs Arch Pharmacol. 382:109–118. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Esmaeilzadeh H, Farjadian S, Alyasin S, Nemati H, Nabavizadeh H and Esmaeilzadeh E: Epidemiology of severe cutaneous adverse drug reaction and its HLA association among pediatrics. Iran J Pharm Res. 18:506–522. 2019.PubMed/NCBI

51 

Yang X, Yan Y, Fang S, Zeng S, Ma H, Qian L, Chen X, Wei J, Gong Z and Xu Z: Comparison of oxcarbazepine efficacy and MHD concentrations relative to age and BMI: Associations among ABCB1, ABCC2, UGT2B7, and SCN2A polymorphisms. Medicine (Baltimore). 98:e149082019. View Article : Google Scholar : PubMed/NCBI

52 

Huang J, Qian Z, Chen H, Huang Q, Huang L, Liu G and Tang X: Association of single nucleotide polymorphisms of SCN1A gene with therapeutic effect of carbamazepine among ethnic Zhuang Chinese patients with epilepsy. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 36:271–274. 2019.(In Chinese). PubMed/NCBI

53 

Shi L, Zhu M, Li H, Wen Z, Chen X, Luo J, Lin C and Zhang Z: SCN1A and SCN2A polymorphisms are associated with response to valproic acid in Chinese epilepsy patients. Eur J Clin Pharmacol. 75:655–663. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Feng W, Mei S, Han J, Zhu L, Yu Y, Gao B, Wu Y, Li J, Zhao Z and Fang F: Lack of association between valproic acid response and polymorphisms of its metabolism, transport, and receptor genes in children with focal seizures. Neurol Sci. 40:523–528. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Suzuki T, Mihara K, Nagai G, Kagawa S, Nakamura A, Nemoto K and Kondo T: Relationship between UGT1A4 and UGT2B7 polymorphisms and the steady-state plasma concentrations of lamotrigine in patients with treatment-resistant depressive disorder receiving lamotrigine as augmentation therapy. Ther Drug Monit. 41:86–90. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Li Z and Wang Y: Relationship between UGT1A4 142T>G polymorphism and serum concentration of lamotrigine in Chinese epileptic patients: A meta-analysis. Zhonghua Yi Xue Za Zhi. 98:3365–3370. 2018.(In Chinese). PubMed/NCBI

57 

Gailite L, Rots D, Pukite I, Cernevska G and Kreile M: Case report: Multiple UGT1A1 gene variants in a patient with Crigler-Najjar syndrome. BMC Pediatr. 18:3172018. View Article : Google Scholar : PubMed/NCBI

58 

Feng W, Mei S, Zhu L, Yu Y, Yang W, Gao B, Wu X, Zhao Z and Fang F: Effects of UGT2B7, SCN1A and CYP3A4 on the therapeutic response of sodium valproate treatment in children with generalized seizures. Seizure. 58:96–100. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Liu L, Zhao L, Wang Q, Qiu F, Wu X and Ma Y: Influence of valproic acid concentration and polymorphism of UGT1A4*3, UGT2B7-161C > T and UGT2B7*2 on serum concentration of lamotrigine in Chinese epileptic children. Eur J Clin Pharmacol. 71:1341–1347. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Sun YX, Zhuo WY, Lin H, Peng ZK, Wang HM, Huang HW, Luo YH and Tang FQ: The influence of UGT2B7 genotype on valproic acid pharmacokinetics in Chinese epilepsy patients. Epilepsy Res. 114:78–80. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Kousar S, Wafai ZA, Wani MA, Jan TR and Andrabi KI: Clinical relevance of genetic polymorphism in CYP2C9 gene to pharmacodynamics and pharmacokinetics of phenytoin in epileptic patients: Validatory pharmacogenomic approach to pharmacovigilance. Int J Clin Pharmacol Ther. 53:504–516. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Zhu X, Yun W, Sun X, Qiu F, Zhao L and Guo Y: Effects of major transporter and metabolizing enzyme gene polymorphisms on carbamazepine metabolism in Chinese patients with epilepsy. Pharmacogenomics. 15:1867–1879. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Reimers A, Sjursen W, Helde G and Brodtkorb E: Frequencies of UGT1A4*2 (P24T) and *3 (L48V) and their effects on serum concentrations of lamotrigine. Eur J Drug Metab Pharmacokinet. 41:149–155. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Berghuis B, Stapleton C, Sonsma ACM, Hulst J, de Haan GJ, Lindhout D, Demurtas R, EpiPGX Consortium, Krause R, Depondt C, et al: A genome-wide association study of sodium levels and drug metabolism in an epilepsy cohort treated with carbamazepine and oxcarbazepine. Epilepsia Open. 4:102–109. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Berghuis B, van der Palen J, de Haan GJ, Lindhout D, Koeleman BPC and Sander JW; EpiPGX Consortium, : Carbamazepine- and oxcarbazepine-induced hyponatremia in people with epilepsy. Epilepsia. 58:1227–1233. 2017. View Article : Google Scholar : PubMed/NCBI

66 

McCormack M, Gui H, Ingason A, Speed D, Wright GEB, Zhang EJ, Secolin R, Yasuda C, Kwok M, Wolking S, et al: Genetic variation in CFH predicts phenytoin-induced maculopapular exanthema in European-descent patients. Neurology. 90:e332–e341. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Bai X, Xu C, Wen D, Chen Y, Li H, Wang X, Zhou L, Huang M and Jin J: Polymorphisms of peroxisome proliferator-activated receptor γ (PPARγ) and cluster of differentiation 36 (CD36) associated with valproate-induced obesity in epileptic patients. Psychopharmacology (Berl). 235:2665–2673. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Li H, Wang X, Zhou Y, Ni G, Su Q, Chen Z, Chen Z, Li J, Chen X, Hou X, et al: Association of LEPR and ANKK1 gene polymorphisms with weight gain in epilepsy patients receiving valproic acid. Int J Neuropsychopharmacol. 18:pyv0212015. View Article : Google Scholar : PubMed/NCBI

69 

Wang C, Wang P, Yang LP, Pan J, Yang X and Ma HY: Association of CYP2C9, CYP2A6, ACSM2A, and CPT1A gene polymorphisms with adverse effects of valproic acid in Chinese patients with epilepsy. Epilepsy Res. 132:64–69. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Manna I, Gambardella A, Bianchi A, Striano P, Tozzi R, Aguglia U, Beccaria F, Benna P, Campostrini R, Canevini MP, et al: A functional polymorphism in the SCN1A gene does not influence antiepileptic drug responsiveness in Italian patients with focal epilepsy. Epilepsia. 52:e40–e44. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Kothur K, Holman K, Farnsworth E, Ho G, Lorentzos M, Troedson C, Gupta S, Webster R, Procopis PG, Menezes MP, et al: Diagnostic yield of targeted massively parallel sequencing in children with epileptic encephalopathy. Seizure. 59:132–140. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Fang ZX, Zhang M, Xie LL, Jiang L, Hong SQ, Li XJ, Hu Y and Chen J: KCNQ2 related early-onset epileptic encephalopathies in Chinese children. J Neurol. 266:2224–2232. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Goto A, Ishii A, Shibata M, Ihara Y, Cooper EC and Hirose S: Characteristics of KCNQ2 variants causing either benign neonatal epilepsy or developmental and epileptic encephalopathy. Epilepsia. 60:1870–1880. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Kuersten M, Tacke M, Gerstl L, Hoelz H, Stülpnagel CV and Borggraefe I: Antiepileptic therapy approaches in KCNQ2 related epilepsy: A systematic review. Eur J Med Genet. 63:1036282020. View Article : Google Scholar : PubMed/NCBI

75 

Al-Eitan LN, Al-Dalalah IM, Elshammari AK, Khreisat WH and Almasri AY: The impact of potassium channel gene polymorphisms on antiepileptic drug responsiveness in Arab patients with epilepsy. J Pers Med. 8(pii): E372018. View Article : Google Scholar : PubMed/NCBI

76 

Howell KB, McMahon JM, Carvill GL, Tambunan D, Mackay MT, Rodriguez-Casero V, Webster R, Clark D, Freeman JL, Calvert S, et al: SCN2A encephalopathy: A major cause of epilepsy of infancy with migrating focal seizures. Neurology. 85:958–966. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Turkdogan D, Thomas G and Demirel B: Ketogenic diet as a successful early treatment modality for SCN2A mutation. Brain Dev. 41:389–391. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Li X, Zhang J, Wu X, Yan H, Zhang Y, He RH, Tang YJ, He YJ, Tan D, Mao XY, et al: Polymorphisms of ABAT, SCN2A and ALDH5A1 may affect valproic acid responses in the treatment of epilepsy in Chinese. Pharmacogenomics. 17:2007–2014. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Al-Eitan LN, Al-Dalalah IM and Aljamal HA: Effects of GRM4, SCN2A and SCN3B polymorphisms on antiepileptic drugs responsiveness and epilepsy susceptibility. Saudi Pharm J. 27:731–737. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Demarest ST and Brooks-Kayal A: From molecules to medicines: The dawn of targeted therapies for genetic epilepsies. Nat Rev Neurol. 14:735–745. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Murray S, Labbé S, Kothare S, Málaga I, Kluger G, Ogden P, Lazure P and Arzimanoglou A: Identifying the educational needs of physicians in pediatric epilepsy in order to improve care: Results from a needs assessment in Germany, Spain, and the United States. Epileptic Disord. 20:239–256. 2018.PubMed/NCBI

82 

Striano P, Vari MS, Mazzocchetti C, Verrotti A and Zara F: Management of genetic epilepsies: From empirical treatment to precision medicine. Pharmacol Res. 107:426–429. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Sánchez Alvarez JC, Serrano Castro PJ and Serratosa Fernández JM: Clinical implications of mechanisms of resistance to antiepileptic drugs. Neurologist. 13 (Suppl 1):S38–S46. 2007. View Article : Google Scholar : PubMed/NCBI

84 

Cardenas-Rodriguez N, Huerta-Gertrudis B, Rivera-Espinosa L, Montesinos-Correa H, Bandala C, Carmona-Aparicio L and Coballase-Urrutia E: Role of oxidative stress in refractory epilepsy: Evidence in patients and experimental models. Int J Mol Sci. 14:1455–1476. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Trivisano M, Terracciano A, Milano T, Cappelletti S, Pietrafusa N, Bertini ES, Vigevano F and Specchio N: Mutation of CHRNA2 in a family with benign familial infantile seizures: Potential role of nicotinic acetylcholine receptor in various phenotypes of epilepsy. Epilepsia. 56:e53–e57. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Wang MY, Liu XZ, Wang J and Wu LW: A novel mutation of the nicotinic acetylcholine receptor gene CHRNA4 in a Chinese patient with non-familial nocturnal frontal lobe epilepsy. Epilepsy Res. 108:1927–1931. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Villa C, Colombo G, Meneghini S, Gotti C, Moretti M, Ferini-Strambi L, Chisci E, Giovannoni R, Becchetti A and Combi R: CHRNA2 and nocturnal frontal lobe epilepsy: Identification and characterization of a novel loss of function mutation. Front Mol Neurosci. 12:172019. View Article : Google Scholar : PubMed/NCBI

88 

Goldberg-Stern H, Kaufmann R, Kivity S, Afawi Z and Heron SE: Novel mutation in KCNQ2 causing benign familial neonatal seizures. Pediatr Neurol. 41:367–370. 2009. View Article : Google Scholar : PubMed/NCBI

89 

Chen DY, Chowdhury S, Farnaes L, Friedman JR, Honold J, Dimmock DP and Gold OBOTRIJJ: Rapid diagnosis of KCNQ2-associated early infantile epileptic encephalopathy improved outcome. Pediatr Neurol. 86:69–70. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Allen NM, Mannion M, Conroy J, Lynch SA, Shahwan A, Lynch B and King MD: The variable phenotypes of KCNQ-related epilepsy. Epilepsia. 55:e99–e105. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Piro E, Nardello R, Gennaro E, Fontana A, Taglialatela M, Donato Mangano G, Corsello G and Mangano S: A novel mutation in KCNQ3-related benign familial neonatal epilepsy: Electroclinical features and neurodevelopmental outcome. Epileptic Disord. 21:87–91. 2019.PubMed/NCBI

92 

Miceli F, Striano P, Soldovieri MV, Fontana A, Nardello R, Robbiano A, Bellini G, Elia M, Zara F, Taglialatela M and Mangano S: A novel KCNQ3 mutation in familial epilepsy with focal seizures and intellectual disability. Epilepsia. 56:e15–e20. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Lehman A, Thouta S, Mancini GMS, Naidu S, van Slegtenhorst M, McWalter K, Person R, Mwenifumbo J, Salvarinova R; CAUSES Study, ; et al: Loss-of-function and gain-of-function mutations in KCNQ5 cause intellectual disability or epileptic encephalopathy. Am J Hum Genet. 101:65–74. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Krepischi AC, Knijnenburg J, Bertola DR, Kim CA, Pearson PL, Bijlsma E, Szuhai K, Kok F, Vianna-Morgante AM and Rosenberg C: Two distinct regions in 2q24.2-q24.3 associated with idiopathic epilepsy. Epilepsia. 51:2457–2460. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Liang JS, Lin LJ, Yang MT, Wang JS and Lu JF: The therapeutic implication of a novel SCN2A mutation associated early-onset epileptic encephalopathy with Rett-like features. Brain Dev. 39:877–881. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Davidsson J, Collin A, Olsson ME, Lundgren J and Soller M: Deletion of the SCN gene cluster on 2q24.4 is associated with severe epilepsy: An array-based genotype-phenotype correlation and a comprehensive review of previously published cases. Epilepsy Res. 81:69–79. 2008. View Article : Google Scholar : PubMed/NCBI

97 

Zaman T, Helbig I, Bozovic IB, DeBrosse SD, Bergqvist AC, Wallis K, Medne L, Maver A, Peterlin B, Helbig KL, et al: Mutations in SCN3A cause early infantile epileptic encephalopathy. Ann Neurol. 83:703–717. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Yang Y, Zhang YH, Chen JY, Zhang J, Yang XL, Chen Y, Yang ZX and Wu XR: Clinical features of epilepsies associated with GABRB2 variants. Zhonghua Er Ke Za Zhi. 57:532–537. 2019.(In Chinese). PubMed/NCBI

99 

Butler KM, Moody OA, Schuler E, Coryell J, Alexander JJ, Jenkins A and Escayg A: De novo variants in GABRA2 and GABRA5 alter receptor function and contribute to early-onset epilepsy. Brain. 141:2392–2405. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Orenstein N, Goldberg-Stern H, Straussberg R, Bazak L, Hubshman MW, Kropach N, Gilad O, Scheuerman O, Dory Y, Kraus D, et al: A de novo GABRA2 missense mutation in severe early-onset epileptic encephalopathy with a choreiform movement disorder. Eur J Paediatr Neurol. 22:516–524. 2018. View Article : Google Scholar : PubMed/NCBI

101 

Zhang Y, Lian Y and Xie N: Early onset epileptic encephalopathy with a novel GABRB3 mutation treated effectively with clonazepam: A case report. Medicine (Baltimore). 96:e92732017. View Article : Google Scholar : PubMed/NCBI

102 

Hernandez CC, Zhang Y, Hu N, Shen D, Shen W, Liu X, Kong W, Jiang Y and Macdonald RL: GABA a receptor coupling junction and pore GABRB3 mutations are linked to early-onset epileptic encephalopathy. Sci Rep. 7:159032017. View Article : Google Scholar : PubMed/NCBI

103 

Farnaes L, Nahas SA, Chowdhury S, Nelson J, Batalov S, Dimmock DM and Kingsmore SF; RCIGM Investigators, : Rapid whole-genome sequencing identifies a novel GABRA1 variant associated with West syndrome. Cold Spring Harb Mol Case Stud. 3(pii): a0017762017. View Article : Google Scholar : PubMed/NCBI

104 

Iqbal MJ, Wasim M, Rashid U, Zeeshan N, Ali R, Nayyab S, Habib S, Manzoor B and Zahid N: Mutational screening of GABRG2 gene in Pakistani population of Punjab with generalized tonic clonic seizures and children with childhood absence epilepsy. J Chin Med Assoc. 81:665–669. 2018. View Article : Google Scholar : PubMed/NCBI

105 

Bhat MA, Guru SA, Mir R, Waza AA, Zuberi M, Sumi MP, Bodeliwala S, Puri V and Saxena A: Association of GABAA receptor gene with epilepsy syndromes. J Mol Neurosci. 65:141–153. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Le SV, Le PHT, Le TKV, Huynh TTK and Do TTH: A mutation in GABRB3 associated with Dravet syndrome. Am J Med Genet A. 173:2126–2131. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Ishii A, Kang JQ, Schornak CC, Hernandez CC, Shen W, Watkins JC, Macdonald RL and Hirose S: A de novo missense mutation of GABRB2 causes early myoclonic encephalopathy. J Med Genet. 54:202–211. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Yoo Y, Jung J, Lee YN, Lee Y, Cho H, Na E, Hong J, Kim E, Lee JS, Lee JS, et al: GABBR2 mutations determine phenotype in rett syndrome and epileptic encephalopathy. Ann Neurol. 82:466–478. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Epi4K Consortium: De novo mutations in SLC1A2 and CACNA1A are important causes of epileptic encephalopathies. Am J Hum Genet. 99:287–298. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Liu J, Tong L, Song S, Niu Y, Li J, Wu X, Zhang J, Zai CC, Luo F, Wu J, et al: Novel and de novo mutations in pediatric refractory epilepsy. Mol Brain. 11:482018. View Article : Google Scholar : PubMed/NCBI

111 

Hayashida T, Saito Y, Ishii A, Yamada H, Itakura A, Minato T, Fukuyama T, Maegaki Y and Hirose S: CACNA1A-related early-onset encephalopathy with myoclonic epilepsy: A case report. Brain Dev. 40:130–133. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Byers HM, Beatty CW, Hahn SH and Gospe SM Jr: Dramatic response after lamotrigine in a patient with epileptic encephalopathy and a de novo CACNA1A variant. Pediatr Neurol. 60:79–82. 2016. View Article : Google Scholar : PubMed/NCBI

113 

Epperson MV, Haws ME, Standridge SM and Gilbert DL: An atypical rett syndrome phenotype due to a novel missense mutation in CACNA1A. J Child Neurol. 33:286–289. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Du X, Chen Y, Zhao Y, Luo W, Cen Z and Hao W: Dramatic response to pyridoxine in a girl with absence epilepsy with ataxia caused by a de novo CACNA1A mutation. Seizure. 45:189–191. 2017. View Article : Google Scholar : PubMed/NCBI

115 

Heron SE, Khosravani H, Varela D, Bladen C, Williams TC, Newman MR, Scheffer IE, Berkovic SF, Mulley JC and Zamponi GW: Extended spectrum of idiopathic generalized epilepsies associated with CACNA1H functional variants. Ann Neurol. 62:560–568. 2007. View Article : Google Scholar : PubMed/NCBI

116 

Yoshida M, Nakashima M, Okanishi T, Kanai S, Fujimoto A, Itomi K, Morimoto M, Saitsu H, Kato M, Matsumoto N and Chiyonobu T: Identification of novel BCL11A variants in patients with epileptic encephalopathy: Expanding the phenotypic spectrum. Clin Genet. 93:368–373. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Kurian M, Korff CM, Ranza E, Bernasconi A, Lübbig A, Nangia S, Ramelli GP, Wohlrab G, Nordli DR and Bast T: Focal cortical malformations in children with early infantile epilepsy and PCDH19 mutations: Case report. Dev Med Child Neurol. 60:100–105. 2018. View Article : Google Scholar : PubMed/NCBI

118 

Ko A, Youn SE, Kim SH, Lee JS, Kim S, Choi JR, Kim HD, Lee ST and Kang HC: Targeted gene panel and genotype- phenotype correlation in children with developmental and epileptic encephalopathy. Epilepsy Res. 141:48–55. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Wang ZJ, Chen J, Chen HL, Zhang LY, Xu D and Jiang WT: Association between SCN1A polymorphism rs3812718 and valproic acid resistance in epilepsy children: A case-control study and meta-analysis. Biosci Rep. 38(pii): BSR201816542018. View Article : Google Scholar : PubMed/NCBI

120 

Margari L, Legrottaglie AR, Vincenti A, Coppola G, Operto FF, Buttiglione M, Cassano A, Bartolomeo N and Mariggio MA: Association between SCN1A gene polymorphisms and drug resistant epilepsy in pediatric patients. Seizure. 55:30–35. 2018. View Article : Google Scholar : PubMed/NCBI

121 

Ajmi M, Boujaafar S, Zouari N, Amor D, Nasr A, Rejeb NB, Amor SB, Omezzine A, Benammou S and Bouslama A: Association between ABCB1 polymorphisms and response to first-generation antiepileptic drugs in a Tunisian epileptic population. Int J Neurosci. 128:705–714. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Abou El Ella SS, Tawfik MA, Abo El Fotoh WMM and Soliman OAM: The genetic variant ‘C588T’ of GABARG2 is linked to childhood idiopathic generalized epilepsy and resistance to antiepileptic drugs. Seizure. 60:39–43. 2018. View Article : Google Scholar : PubMed/NCBI

123 

Skalski D, Wendorff J, Romanowicz H, Rysz A, Marchel A, Stasiołek M and Smolarz B: Associations between MDR1 C3435T polymorphism and drug-resistant epilepsy in the Polish population. Acta Neurol Belg. 117:153–158. 2017. View Article : Google Scholar : PubMed/NCBI

124 

López-Garcia MA, Feria-Romero IA, Serrano H, Rayo-Mares D, Fagiolino P, Vázquez M, Escamilla-Núñez C, Grijalva I, Escalante-Santiago D and Orozco-Suarez S: Influence of genetic variants of CYP2D6, CYP2C9, CYP2C19 and CYP3A4 on antiepileptic drug metabolism in pediatric patients with refractory epilepsy. Pharmacol Rep. 69:504–511. 2017. View Article : Google Scholar

125 

Lv Y, Wang Z, Liu C and Cui L: Identification of a novel CACNA1A mutation in a Chinese family with autosomal recessive progressive myoclonic epilepsy. Neuropsychiatr Dis Treat. 13:2631–2636. 2017. View Article : Google Scholar : PubMed/NCBI

126 

Kozera-Kępniak A, Jastrzębski K, Walenczak J, Klimek A and Głąbiński A: 66034T/C polymorphism of the human pregnane X receptor (hPXR) as potential risk factor for drug resistance in epilepsy-Preliminary study. Neurol Neurochir Pol. 51:19–23. 2017. View Article : Google Scholar : PubMed/NCBI

127 

Wang J, Gao H, Bao X, Zhang Q, Li J, Wei L, Wu X, Chen Y and Yu S: SCN8A mutations in Chinese patients with early onset epileptic encephalopathy and benign infantile seizures. BMC Med Genet. 18:1042017. View Article : Google Scholar : PubMed/NCBI

128 

Parrini E, Marini C, Mei D, Galuppi A, Cellini E, Pucatti D, Chiti L, Rutigliano D, Bianchini C, Virdo S, et al: Diagnostic targeted resequencing in 349 patients with drug-resistant pediatric epilepsies identifies causative mutations in 30 different genes. Hum Mutat. 38:216–225. 2017. View Article : Google Scholar : PubMed/NCBI

129 

Kimizu T, Takahashi Y, Oboshi T, Horino A, Koike T, Yoshitomi S, Mori T, Yamaguchi T, Ikeda H, Okamoto N, et al: A case of early onset epileptic encephalopathy with de novo mutation in SLC35A2: Clinical features and treatment for epilepsy. Brain Dev. 39:256–260. 2017. View Article : Google Scholar : PubMed/NCBI

130 

Shen D, Hernandez CC, Shen W, Hu N, Poduri A, Shiedley B, Rotenberg A, Datta AN, Leiz S, Patzer S, et al: De novo GABRG2 mutations associated with epileptic encephalopathies. Brain. 140:49–67. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Zhang Q, Li J, Zhao Y, Bao X, Wei L and Wang J: Gene mutation analysis of 175 Chinese patients with early-onset epileptic encephalopathy. Clin Genet. 91:717–724. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Miao P, Peng J, Chen C, Gai N and Yin F: A novel mutation in KCNB1 gene in a child with neuropsychiatric comorbidities with both intellectual disability and epilepsy and review of literature. Zhonghua Er Ke Za Zhi. 55:115–119. 2017.(In Chinese). PubMed/NCBI

133 

Perucca P, Scheffer IE, Harvey AS, James PA, Lunke S, Thorne N, Gaff C, Regan BM, Damiano JA, Hildebrand MS, et al: Real-world utility of whole exome sequencing with targeted gene analysis for focal epilepsy. Epilepsy Res. 131:1–8. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Guo M, Wang J, Qi H, Liu F, Yao L, Zhang S and Li K: Polymorphisms in the receptor for advanced glycation end products gene are associated with susceptibility to drug-resistant epilepsy. Neurosci Lett. 619:137–141. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Stasiołek M, Romanowicz H, Połatyńska K, Chamielec M, Skalski D, Makowska M and Smolarz B: Association between C3435T polymorphism of MDR1 gene and the incidence of drug-resistant epilepsy in the population of Polish children. Behav Brain Funct. 12:212016. View Article : Google Scholar : PubMed/NCBI

136 

Abo El Fotoh WM, Abd El Naby SA, Habib MS, ALrefai AA and Kasemy ZA: The potential implication of SCN1A and CYP3A5 genetic variants on antiepileptic drug resistance among Egyptian epileptic children. Seizure. 41:75–80. 2016. View Article : Google Scholar : PubMed/NCBI

137 

Xue T and Lu ZN: Association between the polymorphisms in the ATP-binding cassette genes ABCB1 and ABCC2 and the risk of drug-resistant epilepsy in a Chinese Han population. Genet Mol Res. 15:2016. View Article : Google Scholar :

138 

Moen MN, Fjaer R, Hamdani EH, Laerdahl JK, Menchini RJ, Vigeland MD, Sheng Y, Undlien DE, Hassel B, Salih MA, et al: Pathogenic variants in KCTD7 perturb neuronal K+ fluxes and glutamine transport. Brain. 139:3109–3120. 2016. View Article : Google Scholar : PubMed/NCBI

139 

Hildebrand MS, Griffin NG, Damiano JA, Cops EJ, Burgess R, Ozturk E, Jones NC, Leventer RJ, Freeman JL, Harvey AS, et al: Mutations of the sonic hedgehog pathway underlie hypothalamic hamartoma with gelastic epilepsy. Am J Hum Genet. 99:423–429. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Hardies K, Cai Y, Jardel C, Jansen AC, Cao M, May P, Djémié T, Hachon Le Camus C, Keymolen K, Deconinck T, et al: Loss of SYNJ1 dual phosphatase activity leads to early onset refractory seizures and progressive neurological decline. Brain. 139:2420–2430. 2016. View Article : Google Scholar : PubMed/NCBI

141 

Lionel AC, Monfared N, Scherer SW, Marshall CR and Mercimek-Mahmutoglu S: MED23-associated refractory epilepsy successfully treated with the ketogenic diet. Am J Med Genet A. 170:2421–2425. 2016. View Article : Google Scholar : PubMed/NCBI

142 

Balestrini S, Milh M, Castiglioni C, Lüthy K, Finelli MJ, Verstreken P, Cardon A, Stražišar BG, Holder JL Jr, Lesca G, et al: TBC1D24 genotype-phenotype correlation: Epilepsies and other neurologic features. Neurology. 87:77–85. 2016. View Article : Google Scholar : PubMed/NCBI

143 

Fahrner JA, Liu R, Perry MS, Klein J and Chan DC: A novel de novo dominant negative mutation in DNM1L impairs mitochondrial fission and presents as childhood epileptic encephalopathy. Am J Med Genet A. 170:2002–2011. 2016. View Article : Google Scholar : PubMed/NCBI

144 

Vanstone JR, Smith AM, McBride S, Naas T, Holcik M, Antoun G, Harper ME, Michaud J, Sell E, Chakraborty P, et al: DNM1L-related mitochondrial fission defect presenting as refractory epilepsy. Eur J Hum Genet. 24:1084–1088. 2016. View Article : Google Scholar : PubMed/NCBI

145 

Janssen W, Quaegebeur A, Van Goethem G, Ann L, Smets K, Vandenberghe R and Van Paesschen W: The spectrum of epilepsy caused by POLG mutations. Acta Neurol Belg. 116:17–25. 2016. View Article : Google Scholar : PubMed/NCBI

146 

Li D, Yuan H, Ortiz-Gonzalez XR, Marsh ED, Tian L, McCormick EM, Kosobucki GJ, Chen W, Schulien AJ, Chiavacci R, et al: GRIN2D recurrent de novo dominant mutation causes a severe epileptic encephalopathy treatable with NMDA receptor channel blockers. Am J Hum Genet. 99:802–816. 2016. View Article : Google Scholar : PubMed/NCBI

147 

Segal E, Pedro H, Valdez-Gonzalez K, Parisotto S, Gliksman F, Thompson S, Sabri J and Fertig E: Diagnostic yield of epilepsy panels in children with medication-refractory epilepsy. Pediatr Neurol. 64:66–71. 2016. View Article : Google Scholar : PubMed/NCBI

148 

Palmer S, Towne MC, Pearl PL, Pelletier RC, Genetti CA, Shi J, Beggs AH, Agrawal PB and Brownstein CA: SLC6A1 mutation and ketogenic diet in epilepsy with myoclonic-atonic seizures. Pediatr Neurol. 64:77–79. 2016. View Article : Google Scholar : PubMed/NCBI

149 

Inui T, Kobayashi S, Ashikari Y, Sato R, Endo W, Uematsu M, Oba H, Saitsu H, Matsumoto N, Kure S and Haginoya K: Two cases of early-onset myoclonic seizures with continuous parietal delta activity caused by EEF1A2 mutations. Brain Dev. 38:520–524. 2016. View Article : Google Scholar : PubMed/NCBI

150 

Horta WG, Paradela E, Figueiredo A, Meira ID, Pereira VC, Rego CC, Oliveira R, Andraus ME, de Lacerda GC, Moura P, et al: Genetic association study of the HLA class II alleles DRB1, DQA1, and DQB1 in patients with pharmacoresistant temporal lobe epilepsy associated with mesial hippocampal sclerosis. Seizure. 31:7–11. 2015. View Article : Google Scholar : PubMed/NCBI

151 

Damiano JA, Afawi Z, Bahlo M, Mauermann M, Misk A, Arsov T, Oliver KL, Dahl HH, Shearer AE, Smith RJ, et al: Mutation of the nuclear lamin gene LMNB2 in progressive myoclonus epilepsy with early ataxia. Hum Mol Genet. 24:4483–4490. 2015. View Article : Google Scholar : PubMed/NCBI

152 

Bene J, Hadzsiev K, Komlósi K, Kövesdi E, Mátyás P and Melegh B: De novo SCN1A gene deletion in therapy-resistant Dravet syndrome. Orv Hetil. 156:2009–2012. 2015.(In Hungarian). View Article : Google Scholar : PubMed/NCBI

153 

Berghuis B, de Kovel CG, van Iterson L, Lamberts RJ, Sander JW, Lindhout D and Koeleman BP: Complex SCN8A DNA-abnormalities in an individual with therapy resistant absence epilepsy. Epilepsy Res. 115:141–144. 2015. View Article : Google Scholar : PubMed/NCBI

154 

Damaj L, Lupien-Meilleur A, Lortie A, Riou É, Ospina LH, Gagnon L, Vanasse C and Rossignol E: CACNA1A haploinsufficiency causes cognitive impairment, autism and epileptic encephalopathy with mild cerebellar symptoms. Eur J Hum Genet. 23:1505–1512. 2015. View Article : Google Scholar : PubMed/NCBI

155 

Guo Y, Yan KP, Qu Q, Qu J, Chen ZG, Song T, Luo XY, Sun ZY, Bi CL and Liu JF: Common variants of KCNJ10 are associated with susceptibility and anti-epileptic drug resistance in Chinese genetic generalized epilepsies. PLoS One. 10:e01248962015. View Article : Google Scholar : PubMed/NCBI

156 

Liu Z, Yin X, Liu L, Tao H, Zhou H, Ma G, Cui L, Li Y, Zhang S, Xu Z, et al: Association of KEAP1 and NFE2L2 polymorphisms with temporal lobe epilepsy and drug resistant epilepsy. Gene. 571:231–236. 2015. View Article : Google Scholar : PubMed/NCBI

157 

Emich-Widera E, Likus W, Kazek B, Sieron AL and Urbanek K: Polymorphism of ABCB1/MDR1 C3435T in children and adolescents with partial epilepsy is due to different criteria for drug resistance-preliminary results. Med Sci Monit. 20:1654–1661. 2014. View Article : Google Scholar : PubMed/NCBI

158 

Venkateswaran S, Myers KA, Smith AC, Beaulieu CL, Schwartzentruber JA; FORGE Canada Consortium, ; Majewski J, Bulman D, Boycott KM and Dyment DA: Whole-exome sequencing in an individual with severe global developmental delay and intractable epilepsy identifies a novel, de novo GRIN2A mutation. Epilepsia. 55:e75–e79. 2014. View Article : Google Scholar : PubMed/NCBI

159 

Picard F, Makrythanasis P, Navarro V, Ishida S, de Bellescize J, Ville D, Weckhuysen S, Fosselle E, Suls A, De Jonghe P, et al: DEPDC5 mutations in families presenting as autosomal dominant nocturnal frontal lobe epilepsy. Neurology. 82:2101–2106. 2014. View Article : Google Scholar : PubMed/NCBI

160 

Martin HC, Kim GE, Pagnamenta AT, Murakami Y, Carvill GL, Meyer E, Copley RR, Rimmer A, Barcia G, Fleming MR, et al: Clinical whole-genome sequencing in severe early-onset epilepsy reveals new genes and improves molecular diagnosis. Hum Mol Genet. 23:3200–3211. 2014. View Article : Google Scholar : PubMed/NCBI

161 

Seven M, Batar B, Unal S, Yesil G, Yuksel A and Guven M: The drug-transporter gene MDR1 C3435T and G2677T/A polymorphisms and the risk of multidrug-resistant epilepsy in Turkish children. Mol Biol Rep. 41:331–336. 2014. View Article : Google Scholar : PubMed/NCBI

162 

Escalante-Santiago D, Feria-Romero IA, Ribas-Aparicio RM, Rayo-Mares D, Fagiolino P, Vázquez M, Escamilla-Núñez C, Grijalva-Otero I, López-Garcia MA and Orozco-Suárez S: MDR-1 and MRP2 gene polymorphisms in mexican epileptic pediatric patients with complex partial seizures. Front Neurol. 5:1842014. View Article : Google Scholar : PubMed/NCBI

163 

Seven M, Batar B, Unal S, Yesil G, Yuksel A and Guven M: The effect of genetic polymorphisms of cytochrome P450 CYP2C9, CYP2C19, and CYP2D6 on drug-resistant epilepsy in Turkish children. Mol Diagn Ther. 18:229–236. 2014. View Article : Google Scholar : PubMed/NCBI

164 

Ma CL, Wu XY, Zheng J, Wu ZY, Hong Z and Zhong MK: Association of SCN1A, SCN2A and ABCC2 gene polymorphisms with the response to antiepileptic drugs in Chinese Han patients with epilepsy. Pharmacogenomics. 15:1323–1336. 2014. View Article : Google Scholar : PubMed/NCBI

165 

He X, Li Y, Liu Z, Yue X, Zhao P, Hu J, Wu G, Mao B, Sun D, Zhang H, et al: The association between CCL2 polymorphisms and drug-resistant epilepsy in Chinese children. Epileptic Disord. 15:272–277. 2013. View Article : Google Scholar : PubMed/NCBI

166 

Serino D, Specchio N, Pontrelli G, Vigevano F and Fusco L: Video/EEG findings in a KCNQ2 epileptic encephalopathy: A case report and revision of literature data. Epileptic Disord. 15:158–165. 2013.PubMed/NCBI

167 

Emich-Widera E, Likus W, Kazek B, Niemiec P, Balcerzyk A, Sieroń AL and Zak I: CYP3A5*3 and C3435T MDR1 polymorphisms in prognostication of drug-resistant epilepsy in children and adolescents. Biomed Res Int. 2013:5268372013. View Article : Google Scholar : PubMed/NCBI

168 

Veeramah KR, Johnstone L, Karafet TM, Wolf D, Sprissler R, Salogiannis J, Barth-Maron A, Greenberg ME, Stuhlmann T, Weinert S, et al: Exome sequencing reveals new causal mutations in children with epileptic encephalopathies. Epilepsia. 54:1270–1281. 2013. View Article : Google Scholar : PubMed/NCBI

169 

Fragaki K, Ait-El-Mkadem S, Chaussenot A, Gire C, Mengual R, Bonesso L, Beneteau M, Ricci JE, Desquiret-Dumas V, Procaccio V, et al: Refractory epilepsy and mitochondrial dysfunction due to GM3 synthase deficiency. Eur J Hum Genet. 21:528–534. 2013. View Article : Google Scholar : PubMed/NCBI

170 

Subenthiran S, Abdullah NR, Joseph JP, Muniandy PK, Mok BT, Kee CC, Ismail Z and Mohamed Z: Linkage disequilibrium between polymorphisms of ABCB1 and ABCC2 to predict the treatment outcome of Malaysians with complex partial seizures on treatment with carbamazepine mono-therapy at the Kuala Lumpur Hospital. PLoS One. 8:e648272013. View Article : Google Scholar : PubMed/NCBI

171 

Qu J, Zhou BT, Yin JY, Xu XJ, Zhao YC, Lei GH, Tang Q, Zhou HH and Liu ZQ: ABCC2 polymorphisms and haplotype are associated with drug resistance in Chinese epileptic patients. CNS Neurosci Ther. 18:647–651. 2012. View Article : Google Scholar : PubMed/NCBI

172 

Dimova PS, Kirov A, Todorova A, Todorov T and Mitev V: A novel PCDH19 mutation inherited from an unaffected mother. Pediatr Neurol. 46:397–400. 2012. View Article : Google Scholar : PubMed/NCBI

173 

Sayyah M, Kamgarpour F, Maleki M, Karimipoor M, Gharagozli K and Shamshiri AR: Association analysis of intractable epilepsy with C3435T and G2677T/A ABCB1 gene polymorphisms in Iranian patients. Epileptic Disord. 13:155–165. 2011. View Article : Google Scholar : PubMed/NCBI

174 

Lakhan R, Kumari R, Singh K, Kalita J, Misra UK and Mittal B: Possible role of CYP2C9 & CYP2C19 single nucleotide polymorphisms in drug refractory epilepsy. Indian J Med Res. 134:295–301. 2011.PubMed/NCBI

175 

Kumari R, Lakhan R, Garg RK, Kalita J, Misra UK and Mittal B: Pharmacogenomic association study on the role of drug metabolizing, drug transporters and drug target gene polymorphisms in drug-resistant epilepsy in a north Indian population. Indian J Hum Genet. 17 (Suppl 1):S32–S40. 2011. View Article : Google Scholar : PubMed/NCBI

176 

Kwan P, Wong V, Ng PW, Lui CH, Sin NC, Wong KS and Baum L: Gene-wide tagging study of the association between ABCC2, ABCC5 and ABCG2 genetic polymorphisms and multidrug resistance in epilepsy. Pharmacogenomics. 12:319–325. 2011. View Article : Google Scholar : PubMed/NCBI

177 

Kim DU, Kim MK, Cho YW, Kim YS, Kim WJ, Lee MG, Kim SE, Nam TS, Cho KH, Kim YO and Lee MC: Association of a synonymous GAT3 polymorphism with antiepileptic drug pharmacoresistance. J Hum Genet. 56:640–646. 2011. View Article : Google Scholar : PubMed/NCBI

178 

Meng H, Guo G, Ren J, Zhou H, Ge Y and Guo Y: Effects of ABCB1 polymorphisms on plasma carbamazepine concentrations and pharmacoresistance in Chinese patients with epilepsy. Epilepsy Behav. 21:27–30. 2011. View Article : Google Scholar : PubMed/NCBI

179 

Alpman A, Ozkinay F, Tekgul H, Gokben S, Pehlivan S, Schalling M and Ozkinay C: Multidrug resistance 1 (MDR1) gene polymorphisms in childhood drug-resistant epilepsy. J Child Neurol. 25:1485–1490. 2010. View Article : Google Scholar : PubMed/NCBI

180 

Maleki M, Sayyah M, Kamgarpour F, Karimipoor M, Arab A, Rajabi A, Gharagozli K, Shamshiri AR and Shahsavand Ananloo E: Association between ABCB1-T1236C polymorphism and drug-resistant epilepsy in Iranian female patients. Iran Biomed J. 14:89–96. 2010.PubMed/NCBI

181 

Di Bonaventura C, Carni M, Diani E, Fattouch J, Vaudano EA, Egeo G, Pantano P, Maraviglia B, Bozzao L, Manfredi M, et al: Drug resistant ADLTE and recurrent partial status epilepticus with dysphasic features in a family with a novel LGI1mutation: Electroclinical, genetic, and EEG/fMRI findings. Epilepsia. 50:2481–2486. 2009. View Article : Google Scholar : PubMed/NCBI

182 

Jang SY, Kim MK, Lee KR, Park MS, Kim BC, Cho KH, Lee MC and Kim YS: Gene-to-gene interaction between sodium channel-related genes in determining the risk of antiepileptic drug resistance. J Korean Med Sci. 24:62–68. 2009. View Article : Google Scholar : PubMed/NCBI

183 

Vahab SA, Sen S, Ravindran N, Mony S, Mathew A, Vijayan N, Nayak G, Bhaskaranand N, Banerjee M and Satyamoorthy K: Analysis of genotype and haplotype effects of ABCB1 (MDR1) polymorphisms in the risk of medically refractory epilepsy in an Indian population. Drug Metab Pharmacokinet. 24:255–260. 2009. View Article : Google Scholar : PubMed/NCBI

184 

Kwan P, Wong V, Ng PW, Lui CH, Sin NC, Poon WS, Ng HK, Wong KS and Baum L: Gene-wide tagging study of association between ABCB1 polymorphisms and multidrug resistance in epilepsy in Han Chinese. Pharmacogenomics. 10:723–732. 2009. View Article : Google Scholar : PubMed/NCBI

185 

Kauffman MA, Consalvo D, Gonzalez-Morón D, Aguirre F, D'Alessio L and Kochen S: Serotonin transporter gene variation and refractory mesial temporal epilepsy with hippocampal sclerosis. Epilepsy Res. 85:231–234. 2009. View Article : Google Scholar : PubMed/NCBI

186 

Lakhan R, Kumari R, Misra UK, Kalita J, Pradhan S and Mittal B: Differential role of sodium channels SCN1A and SCN2A gene polymorphisms with epilepsy and multiple drug resistance in the north Indian population. Br J Clin Pharmacol. 68:214–220. 2009. View Article : Google Scholar : PubMed/NCBI

187 

Bahi-Buisson N, Nectoux J, Rosas-Vargas H, Milh M, Boddaert N, Girard B, Cances C, Ville D, Afenjar A, Rio M, et al: Key clinical features to identify girls with CDKL5 mutations. Brain. 131:2647–2661. 2008. View Article : Google Scholar : PubMed/NCBI

188 

Kwan P, Poon WS, Ng HK, Kang DE, Wong V, Ng PW, Lui CH, Sin NC, Wong KS and Baum L: Multidrug resistance in epilepsy and polymorphisms in the voltage-gated sodium channel genes SCN1A, SCN2A, and SCN3A: Correlation among phenotype, genotype, and mRNA expression. Pharmacogenet Genom. 18:989–998. 2008. View Article : Google Scholar

189 

Elia M, Falco M, Ferri R, Spalletta A, Bottitta M, Calabrese G, Carotenuto M, Musumeci SA, Lo Giudice M and Fichera M: CDKL5 mutations in boys with severe encephalopathy and early-onset intractable epilepsy. Neurology. 71:997–999. 2008. View Article : Google Scholar : PubMed/NCBI

190 

Abe T, Seo T, Ishitsu T, Nakagawa T, Hori M and Nakagawa K: Association between SCN1A polymorphism and carbamazepine-resistant epilepsy. Br J Clin Pharmacol. 66:304–307. 2008. View Article : Google Scholar : PubMed/NCBI

191 

Shahwan A, Murphy K, Doherty C, Cavalleri GL, Muckian C, Dicker P, McCarthy M, Kinirons P, Goldstein D and Delanty N: The controversial association of ABCB1 polymorphisms in refractory epilepsy: An analysis of multiple SNPs in an Irish population. Epilepsy Res. 73:192–198. 2007. View Article : Google Scholar : PubMed/NCBI

192 

Kwan P, Baum L, Wong V, Ng PW, Lui CH, Sin NC, Hui AC, Yu E and Wong LK: Association between ABCB1 C3435T polymorphism and drug-resistant epilepsy in Han Chinese. Epilepsy Behav. 11:112–117. 2007. View Article : Google Scholar : PubMed/NCBI

193 

Hung CC, Jen Tai J, Kao PJ, Lin MS and Liou HH: Association of polymorphisms in NR1I2 and ABCB1 genes with epilepsy treatment responses. Pharmacogenomics. 8:1151–1158. 2007. View Article : Google Scholar : PubMed/NCBI

194 

Leschziner GD, Jorgensen AL, Andrew T, Williamson PR, Marson AG, Coffey AJ, Middleditch C, Balding DJ, Rogers J, Bentley DR, et al: The association between polymorphisms in RLIP76 and drug response in epilepsy. Pharmacogenomics. 8:1715–1722. 2007. View Article : Google Scholar : PubMed/NCBI

195 

Seo T, Ishitsu T, Ueda N, Nakada N, Yurube K, Ueda K and Nakagawa K: ABCB1 polymorphisms influence the response to antiepileptic drugs in Japanese epilepsy patients. Pharmacogenomics. 7:551–561. 2006. View Article : Google Scholar : PubMed/NCBI

196 

Hung CC, Tai JJ, Lin CJ, Lee MJ and Liou HH: Complex haplotypic effects of the ABCB1 gene on epilepsy treatment response. Pharmacogenomics. 6:411–417. 2005. View Article : Google Scholar : PubMed/NCBI

197 

Mills PB, Surtees RA, Champion MP, Beesley CE, Dalton N, Scambler PJ, Heales SJ, Briddon A, Scheimberg I, Hoffmann GF, et al: Neonatal epileptic encephalopathy caused by mutations in the PNPO gene encoding pyridox(am)ine 5′-phosphate oxidase. Hum Mol Genet. 14:1077–1086. 2005. View Article : Google Scholar : PubMed/NCBI

198 

Buono RJ, Lohoff FW, Sander T, Sperling MR, O'Connor MJ, Dlugos DJ, Ryan SG, Golden GT, Zhao H, Scattergood TM, et al: Association between variation in the human KCNJ10 potassium ion channel gene and seizure susceptibility. Epilepsy Res. 58:175–183. 2004. View Article : Google Scholar : PubMed/NCBI

199 

Siddiqui A, Kerb R, Weale ME, Brinkmann U, Smith A, Goldstein DB, Wood NW and Sisodiya SM: Association of multidrug resistance in epilepsy with a polymorphism in the drug-transporter gene ABCB1. N Engl J Med. 348:1442–1448. 2003. View Article : Google Scholar : PubMed/NCBI

200 

Gambardella A, Manna I, Labate A, Chifari R, La Russa A, Serra P, Cittadella R, Bonavita S, Andreoli V, LePiane E, et al: GABA(B) receptor 1 polymorphism (G1465A) is associated with temporal lobe epilepsy. Neurology. 60:560–563. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2020
Volume 21 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cárdenas‑Rodríguez N, Carmona‑Aparicio L, Pérez‑Lozano DL, Ortega‑Cuellar D, Gómez‑Manzo S and Ignacio‑Mejía I: Genetic variations associated with pharmacoresistant epilepsy (Review). Mol Med Rep 21: 1685-1701, 2020
APA
Cárdenas‑Rodríguez, N., Carmona‑Aparicio, L., Pérez‑Lozano, D.L., Ortega‑Cuellar, D., Gómez‑Manzo, S., & Ignacio‑Mejía, I. (2020). Genetic variations associated with pharmacoresistant epilepsy (Review). Molecular Medicine Reports, 21, 1685-1701. https://doi.org/10.3892/mmr.2020.10999
MLA
Cárdenas‑Rodríguez, N., Carmona‑Aparicio, L., Pérez‑Lozano, D. L., Ortega‑Cuellar, D., Gómez‑Manzo, S., Ignacio‑Mejía, I."Genetic variations associated with pharmacoresistant epilepsy (Review)". Molecular Medicine Reports 21.4 (2020): 1685-1701.
Chicago
Cárdenas‑Rodríguez, N., Carmona‑Aparicio, L., Pérez‑Lozano, D. L., Ortega‑Cuellar, D., Gómez‑Manzo, S., Ignacio‑Mejía, I."Genetic variations associated with pharmacoresistant epilepsy (Review)". Molecular Medicine Reports 21, no. 4 (2020): 1685-1701. https://doi.org/10.3892/mmr.2020.10999