Abstract
Background and objective CYP3A4 (rs2242480), CYP3A5 (rs776746) and SCN1A (rs3812718 and rs2298771) gene polymorphisms were previously indicated to be associated with carbamazepine (CBZ) metabolism and resistance in epilepsy. However, previous studies regarding the effects of these polymorphisms still remain controversial. Therefore, we performed a meta-analysis to evaluate whether the four polymorphisms are associated with CBZ metabolism and resistance.
Methods The PubMed, EMBASE, Cochrane library, Chinese National Knowledge Infrastructure, Chinese Science and Technique Journals Database, China Biology Medicine disc and Wan fang Database were searched up to January 2020 for appropriate studies regarding the association of rs2242480, rs776746, rs3812718 and rs2234922 polymorphisms with metabolism and resistance to CBZ. The meta-analysis was conducted by Review Manager 5.3 software.
Results Eighteen studies involving 2574 related epilepsy patients were included. Significant associations between CYP3A4 rs2242480, CYP3A5 rs776746 and SCN1A rs3812718 polymorphisms and plasma concentrations of CBZ were observed. Additionally, SCN1A rs3812718 polymorphism was significantly associated with CBZ resistance. However, no association was observed between SCN1A rs2298771 polymorphism and metabolism and resistance to CBZ.
Conclusion The CYP3A4 rs2242480, CYP3A5 rs776746 and SCN1A rs3812718 polymorphisms may play important roles in metabolism and resistance to CBZ, while SCN1A rs2298771 polymorphism is not associated with CBZ in epilepsy. These findings would improve the individualized therapy of epileptic patients in clinics.
1. INTRODUCTION
Epilepsy, one of the most common chronic neurological disorder, affects more than 70 million people worldwide[1]. In addition, long-term recurrent seizures was associated with increased morbidity and mortality, which can give rise to a large social and economic burden and seriously hurt patients’ physical and mental health[2]. For most people with epilepsy, anti-epileptic drug (AED) monotherapy is a preferred treatment modality, with the aim of reducing risks for drug–drug interactions, fewer adverse events, and better patient compliance[3, 4].
Carbamazepine (CBZ) therapy is one of the most economical treatment usually prescribed, exhibiting beneficial efficacy in control of simple partial, complex partial, and generalized tonic-clonic seizures and mitigating long-term mortality[5]. However, from the pharmacological point of view, CBZ is a powerful liver enzyme inducer, with the characteristic of inducing self-metabolism and hence resulting in the individualized differences of clinical efficacy[6]. Moreover, the metabolism of CBZ is a complex oxidative and epoxidase pathway. First, CBZ is mainly catalyzed by cytochrome P450 (CYP) 3A4 and 3A5 to produce active metabolite carbamazepine-10,11-epoxide (CBZE), which possesses a potent anticonvulsant effect[7]. Subsequently, CBZE is hydrolyzed via microsomal epoxide hydrolase, which is encoded by the EPHX1 gene, to an inactive metabolite carbamazepine-10,11-trans dihydrodiol (CBZD)[7-9]. It was previously reported that genetic variation of drug-metabolizing enzymes could generate the inter-individual differences in the pharmacokinetics and pharmacodynamics of epileptic patients[10-12]. In our recent meta-analysis, it was further proved that EPHX1 polymorphisms (rs1051740 and rs2234922) were significantly associated with concentrations of both CBZ and CBZE and metabolism ratio[13]. However, previous studies regarding the effects of polymorphisms of CYP3A4/3A5 on the pharmacokinetics and pharmacodynamics of CBZ still remain controversial. For example, it was shown that CYP3A4*1G (g.20230G>A, rs2242480) variant carriers had lower plasma concentration of CBZ than CYP3A4*1/*1 carriers[14, 15]. On the contrary, several other studies found no significant associations between the variant CYP3A4 and plasma concentration of CBZ[16, 17]. Additionally, similar inconsistent results were also found for polymorphism of CYP3A5 (g.6986A>G, rs776746). One study reported that the CYP3A5 rs776746 polymorphism had no associations with plasma concentration of CBZ[18], but several other studies revealed that CYP3A5 rs776746 nonexpressers had higher plasma concentration of CBZ than expressers[19, 20].
In addition, CBZ is a sodium channel blocker and mainly inhibit voltage-sensitive sodium channel activity[21]. Its mechanism of action was binding to a receptor site in the pore of neuronal sodium channels, decreasing sodium influx, reducing the number and duration of action potentials within a burst and thereby reducing the excitability of the central neurons[22, 23]. The voltage-gated sodium channel α subunit type 1 (SCN1A) is a heterologous complex, which comprises a large hollow glycosylation α subunit encoded by SCN1A gene and two small affiliated β subunits encoded by SCN2A gene[24, 25]. Previous studies indicated that variations in sensitivity of pharmacological targets might arise from single nucleotide polymorphisms (SNPs)[26-28]. However, these reports did not clarify the correlations between the SCN1A rs3812718 (IVS5 G>A) and rs2298771 (c.3184A>G) polymorphisms and resistance to CBZ in epilepsy. For example, it was shown that the rs3812718 and rs2298771polymorphisms were significantly associated with CBZ resistance in English and Chinese cohorts of patients[29, 30]. On the contrary, one study found that the variant SCN1A had no significant influence on CBZ resistance in Japanese epilepsy patients[31].
Therefore, it is necessary to comprehensively evaluate the associations of the abovementioned polymorphisms in CYP3A4, CYP3A5 and SCN1A genes with CBZ metabolism and resistance to achieve a more rational and individualized use of CBZ in epileptic treatment. In this regard, a systematic meta-analysis based on all currently eligible studies was hence performed in this study hoping to provide novel insights into the roles of genetic variations on the pharmacokinetics and pharmacodynamics of CBZ.
2. MATERIALS AND METHODS
2.1. Search strategy
The recommendations of the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement were followed in this study[32]. A comprehensive literature search of studies in all languages up to January 2020 across PubMed, EMBASE, Cochrane library, Chinese National Knowledge Infrastructure (CNKI), Chinese Science and Technique Journals Database (VIP), China Biology Medicine disc (CBM) and Wan fang Data Information Site was conducted.
Relevant studies were identified using the terms: “carbamazepine”, “CYP 450” or “Cytochrome P-450 Enzyme System”, “SCN1A protein” or “NAV1.1 Voltage-Gated Sodium Channel”, “polymorphism” or “genotype” and “epilepsy”. The search strategies for PubMed were provided in supplementary materials. In addition, we carried out a manual retrieve of references of all acquired articles to identify additional eligible studies. The literature retrieval was performed by two independent authors and the discrepancy was resolved by consensus.
2.2. Inclusion and exclusion criteria
Studies fulfilling the following criteria were considered as eligible for inclusion in this meta-analysis: (a) original studies that investigated the associations between the polymorphisms of SCN1A, CYP3A4 or CYP3A5 and plasma concentration or resistance of CBZ in epilepsy, (b) each patient was administrated with CBZ monotherapy, (c) no liver or renal diseases were reported, and (d) including at least three studies for each polymorphism to allow calculation of publication bias between studies. Exclusion criteria were the following: (a) insufficient information for data extraction, (b) reviews, case reports or articles only with an abstract, and (c) studies in vitro experiments.
2.3. Data extraction
Two reviewers independently extracted data included authors, publication date, country, sample size, allelic and genotype distribution, plasma concentration of CBZ, CBZE and CBZD, CBZE:CBZ, CBZD:CBZ and CBZD:CBZE ratios, drug resistant and responsive patients. According to the criteria International League Against Epilepsy (ILAE) 2010, drug resistant epilepsy was defined as failure of adequate trials of two tolerated and appropriately chosen and used AED schedules (whether as monotherapies or in combination) to achieve sustained seizure freedom; drug-responsive epilepsy was defined as epilepsy in which the patients receiving the current AED treatment regimen has been seizure free for a minimum of three times the longest pre-intervention interseizure interval or 12 months, whichever is longer[33]. The authors of the studies were contacted for additional data when necessary and applicable. Disagreement between the reviewers was resolved by discussion or consensus with the third investigator.
2.4. Assessment of methodological quality
The Newcastle-Ottawa Scale (NOS), which was developed to assess the quality of non-randomized studies in the meta-analysis, was used to assess the methodological quality of included studies[34]. The NOS used a star rating system to judge quality based on eight items and categorized into three broad perspectives: selection, comparability and exposure. High-quality studies were identified with a NOS score of five or more, whereas those with less than a score of five were considered as low-quality studies. The quality assessment was performed by two investigators independently, and disagreements were resolved by discussion and consensus.
2.5. Statistical analysis
The Review Manager 5.3 software was utilized for statistical analysis. Comparisons of the dichotomous variables were performed using risk ratio (RR) with 95% confidence interval (CI). For continuous variables, the standard mean differences (SMD) or mean differences (MD) with 95% CI were used in the meta-analysis. The statistical significance was determined by Z test, and P-value < 0.05 was considered as statistically significant. Heterogeneity was tested using the Cochran’s Q test and measured inconsistency by I2[35]. Data with low heterogeneity (P > 0.10 and I2 < 50%) were analyzed by a fixed-effects model while a random-effect model was used for data with high heterogeneity (P ≤ 0.10 and I2 ≥ 50%). Sensitivity analyses were performed by excluding one study at a time to explore sources of heterogeneity and evaluate the stability of pooled results. Moreover, a funnel plot was conducted to assess the publication bias when applicable.
3. RESULTS
3.1. Study selection and qualitative assessment
Detailed flow diagram of the study selection process is presented in Fig. 1. The initial search strategy identified 819 studies in the electronic databases, from which 53 were excluded for duplication. Then, we excluded 710 studies after reading titles, abstracts and full-text articles of the remaining 766 studies, which included 459 irrelevant studies, 251 review articles or commentaries, 13 in vitro or animal studies, 25 studies with insufficient data. Eventually, 18 studies were included according to the present inclusion and exclusion criteria. Of the included studies, the polymorphisms of rs2242480, rs776746, rs3812718 and rs2298771 were reported in 4[14-17], 10[14-16, 18, 20, 36-40], 7[16, 17, 30, 41-44] and 6[16, 30, 41, 42, 45, 46] studies, respectively.
Flow diagram for process of included studies selection.
A total of 2574 patients were included in this meta-analysis. The characteristics from selected studies in CYP3A4, CYP3A5 and SCN1A polymorphisms and CBZ metabolism and resistance were summarized in Table 1. These studies were published between 2009 and 2019 from different countries. All participants in the studies were epilepsy patients and were treated with CBZ monotherapy at a stable maintenance dose. The scores ranging from 5 to 8 based on the NOS evaluation system, indicating a relatively high quality of included studies. Furthermore, the publication bias was not conducted due to the insufficient number of studies in the present study[47].
Characteristics of all studies included in this meta-analysis.
3.2. Meta-analysis of the associations between CYP3A4 rs2242480 polymorphism and CBZ metabolism
The results of the associations between CYP3A4 rs2242480, CYP3A5 rs776746, SCN1A rs3812718 and rs2298771 polymorphisms and CBZ metabolism for meta-analysis were showed in supplemental Table S.1. Meta-analysis of the CYP3A4 rs2242480 polymorphism showed significant associations with plasma concentration of CBZ in the heterozygote codominant model (AG vs. GG, SMD = −0.27, 95% CI = −0.43 to −0.11, P = 0.001) (Fig. 2c), dominant model (AA+AG vs. GG, SMD = −0.23, 95% CI = −0.39 to −0.08, P = 0.003) (Fig. 2d) and overdominant model (AG vs. AA+GG, SMD = −0.28, 95% CI = −0.44 to −0.12, P = 0.0006) (Fig. 2f), indicating that this SNP might decrease the plasma CBZ concentration in epilepsy. Moreover, one study showed that in homozygote codominant model and recessive model the ratio of CBZE to CBZ was significantly lower in carriers of the AA genotype than carriers of the GG genotype (AA vs. GG, MD = −0.04, 95% CI = −0.08 to −0.00, P = 0.04) (supplemental Fig. S.1b) and AG+GG genotype (AA vs. AG+GG, MD = −0.04, 95% CI = −0.08 to −0.00, P =0.03) (supplemental Fig. S.1e)[16]. Regarding the plasma concentrations of CBZE and CBZD, no association was found in all the genetic models (supplemental Fig. S.2 and S.3).
Forest plot for association between CYP3A4 rs2242480 polymorphism and plasma CBZ concentration in allelic model (a), homozygote codominant model (b), heterozygote codominant model (c), dominant model (d), recessive model (e) and overdominant model (f).
3.3. Meta-analysis of the associations between CYP3A5 rs776746 polymorphism and CBZ metabolism
For rs776746 polymorphism, the GG genotype and GG+GA genotype was found to be significantly associated with plasma concentration of CBZ in homozygote codominant model (GG vs. AA, SMD = 0.68, 95% CI = 0.37 to 1.00, P < 0.0001) (Fig. 3b), dominant model (GG+GA vs. AA, SMD = 0.38, 95% CI = 0.05 to 0.72, P = 0.03) (Fig. 3d) and recessive model (GG vs. GA+AA, SMD = 0.57, 95% CI = 0.21 to 0.92, P = 0.002) (Fig. 3e). In addition, the sensitivity analysis was also conducted by excluding each study successively, which abrogated the heterogeneity but did not influence the present results (Fig. 3g and Fig 3h). As for the plasma concentrations of CBZE, CBZD and the ratio of CBZD to CBZ, our meta-analysis did not observed any associations between rs776746 polymorphism and the metabolism of CBZ in all the genetic models (supplemental Fig. S.4-S.6). However, when sensitivity analysis for recessive model of the plasma concentrations of CBZE was performed, the association became significant with the heterogeneity relatively reduced (SMD = −0.49, 95% CI = −0.82 to −0.17, P = 0.003) (supplemental Fig. S.4g).
Forest plot for association between CYP3A5 rs776746 polymorphism and plasma CBZ concentration in allelic model (a), homozygote codominant model (b), heterozygote codominant model (c), dominant model (d), recessive model (e) and overdominant model (f). Sensitivity analysis for effect of CYP3A5 rs776746 polymorphism on plasma CBZ concentration in recessive model (g) and overdominant model (h).
3.4. Meta-analysis of the associations between SCN1A rs3812718 and rs2298771 polymorphisms and CBZ metabolism
For rs3812718, the allelic model (A allele vs. G allele, SMD = −0.58, 95% CI = −0.73 to −0.44, P < 0.00001) (Fig. 4a) and the overdominant model (AG vs. AA+GG, SMD = −0.19, 95% CI = −0.30 to −0.08, P = 0.001) (Fig. 4f) showed a significant difference between rs3812718 polymorphism and plasma concentration of CBZ. The sensitivity analysis decreased the heterogeneity but did not influence the present results (supplemental Fig. S.7). In addition, a significant difference was found in the ratio of CBZE to CBZ in heterozygote codominant model (AG vs. GG, OR = 0.32, 95% CI = 0.05 to 0.59, P = 0.02) (supplemental Fig. S.8c) and dominant model (AA+AG vs. GG, OR = 0.29, 95% CI = 0.03 to 0.55, P = 0.03) (supplemental Fig. S.8d). Furthermore, one study reported that AA genotype increased the ratio of CBZD to CBZ in homozygote codominant model (AA vs. GG, MD = 0.20, 95% CI = 0.11 to 0.29, P < 0.0001) (supplemental Fig. S.9a), dominant model (AA+AG vs. GG, MD = 0.07, 95% CI = 0.01 to 0.13, P = 0.02) (supplemental Fig. S.9c) and recessive model (AA vs. AG+GG, OR = 0.18, 95% CI = 0.09 to 0.27, P < 0.0001) (supplemental Fig. S.9d)[41]. As for the plasma concentrations of CBZE and CBZD and the ratio of CBZD to CBZE, no statistically significant association was found between SCN1A rs3812718 polymorphism and CBZ metabolism (supplemental Fig. S.10-S.12). For rs2298771, no statistically significant association was found between this polymorphism and CBZ metabolism in all the six genetic models (supplemental Fig. S.13-S.19). Furthermore, sensitivity analysis decreased the heterogeneity but did not alter the initial result (supplemental Fig. S.13g and S.15).
Forest plot for association between SCN1A rs3812718 polymorphism and plasma CBZ concentration in allelic model (a), homozygote codominant model (b), heterozygote codominant model (c), dominant model (d), recessive model (e) and overdominant model (f).
3.5. Meta-analysis of the associations between CYP3A4 rs2242480, CYP3A5 rs776746, SCN1A rs3812718 and rs2298771 polymorphisms and CBZ resistance
The association between rs2242480, rs776746, rs3812718 and rs2298771 polymorphisms and CBZ resistance was pooled in supplemental Table S.2. For SCN1A rs3812718, the results showed that the A allele and mutant-type homozygote AA genotype increased CBZ resistance in allelic model (A allele vs. G allele, RR = 1.18, 95% CI = 1.05 to 1.33, P = 0.007) (Fig 5a), homozygote codominant model (AA vs. GG, RR = 1.24, 95% CI = 1.08 to 1.43, P = 0.003) (Fig. 5b) and recessive model (AA vs. AG+GG, RR = 1.47, 95% CI = 1.22 to 1.77, P < 0.0001) (Fig. 5e). However, we did not find any association regarding CYP3A4 rs2242480, CYP3A5 rs776746 and SCN1A rs2298771 polymorphisms (supplemental Fig. S.20–S.22), indicating that these polymorphisms might not the causal factors associated with resistance of CBZ in epilepsy. Furthermore, the sensitivity analysis also effectively abrogated the heterogeneity but did not influence the present results (supplemental Fig. S22g).
Forest plot for association between SCN1A rs3812718 polymorphism and CBZ resistance in allelic model (a), homozygote codominant model (b), heterozygote codominant model (c), dominant model (d), recessive model (e) and overdominant model (f).
4. DISCUSSION
With the development of pharmacogenomics, it has been shown that the variability in individual response to antiepileptic drugs depends not only on non-genetic factors (i.e., age, environment and concomitant medications), but also on genetic factors[48]. In a previous study, it was found that genetic factors accounted for 20 to 95 percent of the factors of individual differences in drug efficacy[49]. Hence, to provide a comprehensive profile of the associations of certain genetic polymorphisms with CBZ pharmacokinetics and pharmacodynamics would contribute to a more understanding of pharmacological characters of CBZ. To our knowledge, this is the first meta-analysis to address the associations between CYP3A4, CYP3A5 and SCN1A polymorphisms and metabolism and efficacy of CBZ and found significant relationships between rs2298771, rs2242480 and rs776746 polymorphisms and plasma concentration of CBZ, which might further explain individual differences of CBZ and provide effective references for personalized CBZ therapy in epileptic patients.
The SCN1A gene encodes the alpha subunit Nav1.1 of the voltage-gated sodium channel and plays a crucial role in the pharmacological effects of CBZ[50, 51]. This gene is located on chromosome 2q24.3, consists of 26 exons and spans 91480bp of DNA[52, 53]. Alternative splicing of Nav genes has been shown to generate channels with unique functional and pharmacologic properties[54]. Previous pharmacogenomics studies demonstrated that rs3812718 genetic variant was located within an intron splice donor site and altered the proportion of SCN1A transcripts incorporating the canonical (exon 5A) or alternative (exon 5N) exon 5[55, 56]. Previous initial reports suggested that exon 5N was expressed primarily during the neonatal period, whereas the exon 5A was the adult exon[56-58]. Further studies revealed that the G allele of rs3812718 allowed both the 5N and 5A forms of this exon to be alternatively expressed, while the A allele reduced expression of the 5N form relative to the 5A form[55, 59, 60]. In this meta-analysis, we found that the rs3812718 SNP was significantly associated with the plasma concentration of CBZ, CBZE:CBZ ratio, CBZD:CBZ ratio and CBZ resistance, which provided further evidence that this SNP influenced the relative expression of neonatal and adult transcripts of SCN1A. The rs3812718 polymorphism may serve as a biomarker for individualized CBZ therapy. However, the exact underlying mechanisms of how this SNP affects the potential associations remain unclear and further functional investigation is needed to clarify this issue.
The nonsynonymous mutation rs2298771 within exon 16 of the SCN1A gene is characterized by substitution of alanine for threonine at the amino acid position 1067 (Thr1067Ala) and is an A to G SNP [61]. In the present meta-analysis, we failed to detect any statistically significant association of the genetic variant with CBZ metabolism and resistance, which was contrary to certain previous findings[24, 38, 62]. Several studies found that this polymorphism might affect structure-function relationship of the channel leading to misfiring of brain neurons and affecting drug efficacy and patient response[62, 63]. The inconsistencies between the present meta-analysis data and these fundamental studies may be due to the effects of various other confounded factors, including ethnicity, age, environment of the patients and physicians’ bias as well[12, 64]. Regretfully, we did not perform further subgroup analyses of these possible underlying factors because of data insufficiency. Additionally, the limited sample size might also have underestimated subtle effects of the genetic variant. Therefore, further well-designed investigations are required to verify these findings.
The cytochrome P450 gene is one of the most extensively studied genes with pharmacogenomics relevance due to its involvement in the metabolism of conventional drugs, most of which have narrow therapeutic indexes[65, 66]. Meanwhile, CYP3A4 and CYP3A5 were considered to play a major role during the hepatic oxidation process of CBZ[7, 67]. Among the variants within CYP3A4 and CYP3A5 genes, the rs2242480 polymorphism, also known as CYP3A4*1G, is located in intron 10 of CYP3A4 while the rs776746 polymorphism, also known as CYP3A5*3, is located in intron 3 of CYP3A5[68-70]. Previous studies reported that these 2 genetic variations were respectively associated with the CYP3A4 or CYP3A5 enzyme activities in epileptic patients. As a functional locus, the rs2242480 polymorphism was demonstrated to enhance the expression and enzyme activity of CYP3A4 in human liver and blood brain barrier and hence to affect the metabolism and resistance of CBZ[71, 72]. With regard to the rs776746, it was found to play a major role in the expression of the CYP3A5 enzyme via a splicing defect of mRNA and producing a truncated and nonfunctional protein[20, 73]. In this mate-analysis, the rs2242480 polymorphism was observed to be associated with decreased serum concentration of CBZ and decreased CBZE:CBZ ratio, while the rs776746 polymorphism was observed to be associated with increased serum concentration of CBZ and decreased serum concentration of CBZE. The findings were consistent with the previous studies and further suggested that these 2 SNPs respectively affected the metabolism of CBZ[16, 55].
Some limitations of the present meta-analysis deserve consideration. Firstly, included studies recruited relatively few participants and thus further confirmatory evidence from large sample trials is required. Secondly, publication bias might not be avoided for only including published articles while missing certain related unpublished studies with negative results. Thirdly, CBZ metabolism and pharmacoresistance might be influenced by several confounding factors such as age, sex, ethnicity and interactions between SNPs. But original data deficiency limited our further assessment of these factors. Finally, there is fairly significant heterogeneity in partial outcomes of the study, which might reduce the validity of the results.
In conclusion, our meta-analysis indicated that the SCN1A rs3812718, CYP3A4 rs2242480 and CYP3A5 rs776746 polymorphisms were related to the metabolism of CBZ, while SCN1A rs2298771 had no obvious effects on CBZ metabolism for epilepsy. We also found that the SCN1A rs3812718 polymorphism was associated with an increased resistance of CBZ. These findings confirmed significant effects of SCN1A, CYP3A4 and CYP3A5 gene polymorphisms on pharmacokinetics and pharmacodynamics of CBZ and would improve the individualized therapy of epileptic patients in clinics.
Data Availability
The data used to support the findings of this study are included within the article.
ETHICS APPROVAL AND CONSENT TO PARTICIPATE
All results and analyses were based on previous ethically-approved studies, thus no further ethical approval and patient consent are required.
HUMAN AND ANIMAL RIGHTS
This article does not contain any studies with human or animal subjects performed by any of the authors.
CONFLICTS OF INTEREST
The authors declare that they have no conflict of interest.
FUNDING
This work was supported by Grants from the National Science Foundation of China (No. 81460560 and 81960664) and the Applied Basic Research Program of Yunnan Province of China (No.2017FB134).
SUPPLEMENTARY MATERIAL
Supplementary material is available on the publisher’s website along with the published article.
ACKNOWLEDGEMENTS
Declared none.