We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Pharmacogenetics and the treatment of epilepsy: what do we know?

    Nada Božina

    *Author for correspondence:

    E-mail Address: nbozina@kbc-zagreb.hr

    Department of Laboratory Diagnostics, Division of Pharmacogenomics & Therapy Individualiation, University Hospital Centre Zagreb, 10000 Zagreb, Croatia

    Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia

    ,
    Ivana Šušak Sporiš

    Department of Neurology, University Hospital Dubrava, 10000 Zagreb, Croatia

    Faculty of Dental Medicine & Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia

    ,
    Tamara Božina

    Department of Medical Chemistry, Biochemistry & Clinical Chemistry, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia

    ,
    Iva Klarica-Domjanović

    Croatian Agency for Medicinal Products & Medical Devices, 10000 Zagreb, Croatia

    ,
    Ante Tvrdeić

    Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia

    &
    Davor Sporiš

    Department of Neurology, University Hospital Dubrava, 10000 Zagreb, Croatia

    Faculty of Dental Medicine & Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia

    Published Online:https://doi.org/10.2217/pgs-2019-0085

    Seizure control with antiepileptic drugs (AEDs) as well as susceptibility to adverse drug reactions varies among individuals with epilepsy. This interindividual variability is partly determined by genetic factors. However, genetic testing to predict the efficacy and toxicity of AEDs is limited and genetic variability is, as yet, largely unexplainable. Accordingly, genetic testing can only be advised in a very limited number of cases in clinical routine. Currently, by applying different methodologies, many trials have been undertaken to evaluate cost benefits of preventive pharmacogenetic analysis for patients. There is significant progress in sequencing technologies, and focus is on next-generation sequencing-based methods, like exome and genome sequencing. In this review, an overview of the current scientific knowledge considering the pharmacogenetics of AEDs is given.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Kwan P, Brodie MJ. Early identification of refractory epilepsy. N. Engl. J. Med. 342(5), 314–319 (2000).
    • 2. Canevini MP, De Sarro G, Galimberti CA et al. Relationship between adverse effects of antiepileptic drugs, number of coprescribed drug, and drug load in a large cohort of consecutive patients with drug-refractory epilepsy. Epilepsia 51(5), 797–804 (2010).
    • 3. Franco V, Perucca E. CYP2C9 polymorphisms and phenytoin metabolism: implications for adverse effects. Expert Opin. Drug Metab. Toxicol. 11(8), 1269–1279 (2015).
    • 4. Loscher W, Klotz U, Zimprich F, Schmidt D. The clinical impact of pharmacogenetics on the treatment of epilepsy. Epilepsia 50(1), 1–23 (2009).
    • 5. Thomas RH, Berkovic SF. The hidden genetics of epilepsy clinically important new paradigm. Nat. Rev. Neurol. 10(5), 283–292 (2014).
    • 6. Stingl JC, Bartels H, Viviani R, Lehmanna ML, Brockmöllere J. Relevance of UDP-glucuronosyl-transferase polymorphisms for drug dosing: a quantitative systematic review. Pharmacol. Ther. 141(1), 92–116 (2014).
    • 7. Caudle KE, Rettie AE, Whirl-Carrillo M et al. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C9 and HLA-B genotypes and phenytoin dosing. Clin. Pharmacol. Ther. 96(5), 542–548 (2014). •• Recommendations for the use of phenytoin based on CYP2C9 and/or HLA-B genotype.
    • 8. Chung WH, Chang WC, Lee YS et al. Genetic variants associated with phenytoin-related severe cutaneous adverse reactions. JAMA 312(5), 525–534 (2014). • Report on the association between CYP2C9*3 polymorphism and phenytoin-related severe cutaneous adverse drug reactions.
    • 9. Yukawa E, Mamiya K. Effect of CYP2C19 genetic polymorphism on pharmacokinetics of phenytoin and phenobarbital in Japanese epileptic patients using non-linear mixed effects model approach. J. Clin. Pharm. Ther. 31(3), 275–282 (2006).
    • 10. Goto S, Seo T, Murata T et al. Population estimation of the effects of cytochrome P450 2C9 and 2C19 polymorphisms on phenobarbital clearance in Japanese. Ther. Drug Monit. 29(1), 118–121 (2007).
    • 11. Balestrini S, Sisodiya SM. Pharmacogenomics in epilepsy. Neurosci. Lett. 667, 27–39 (2018).
    • 12. Okada Y, Seo T, Ishitsu T et al. Population estimation regarding the effects of cytochrome P450 2C19 and 3A5 polymorphisms on zonisamide clearance. Ther. Drug Moni. 30(4), 540–543 (2008).
    • 13. Dean L. Brivaracetam therapy and CYP2C19 genotype. In: Medical Genetics Summaries. Pratt VMcLeod HRubinstein WKattman BMalheiro A (Eds). National Center for Biotechnology Information (US), Bethesda, MD, USA, 59–66 (2018).
    • 14. Kosaki K, Tamura K, Sato R, Samejima H, Tanigawara Y, Takahashi T. A major influence of CYP2C19 genotype on the steady-state concentration of N-desmethylclobazam. Brain Dev. 26, 530–534 (2004).
    • 15. Rosemary J, Adithan C. The pharmacogenetics of CYP2C9 and CYP2C19: ethnic variation and clinical significance. Curr. Clin. Pharmacol. 2(1), 93–109 (2007).
    • 16. Ganoci L, Božina T, Mirošević Skvrce N, Lovrić M, Mas P, Božina N. Genetic polymorphisms of cytochrome P450 enzymes: CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5 in the Croatian population. Drug Metab. Pers. Ther. 32, 11–21 (2017).
    • 17. Dean L. Lacosamide therapy and CYP2C19 genotype. In: Medical Genetics Summaries. Pratt VMcLeod HRubinstein WKattman BMalheiro A (Eds). National Center for Biotechnology Information (US), Bethesda, MD, USA, 215–218 (2012–2018).
    • 18. Zhou SF. Drugs behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. Curr. Drug Meta. 9(4), 310–322 (2008).
    • 19. Klein K, Thomas M, Winter S, Nussler AK, Niemi M, Schwab M. PPARA: a novel genetic determinant of CYP3A4 in vitro and in vivo. Clin. Pharmacol. Ther. 91, 1044–1052 (2012).
    • 20. Amirimani B, Ning B, Deitz AC, Weber BL, Kadlubar FF, Rebbeck TR. Increased transcriptional activity of the CYP3A4*1B promoter variant. Environ. Mol. Mutagen. 42(4), 299–305 (2003).
    • 21. García-Martín E. CYP3A4 variant alleles in white individuals with low CYP3A4 enzyme activity. Clin. Pharmacol. Ther. 71(3), 196–204 (2002).
    • 22. Okubo M, Murayama N, Shimizu M, Shimada T, Guengerich FP, Yamazaki H. CYP3A4 intron 6 C>T polymorphism (CYP3A4*22) is associated with reduced CYP3A4 protein level and function in human liver microsomes. J. Toxicol. Sci. 38(3), 349–354 (2013).
    • 23. Lilja J. Duration of effect of grapefruit juice on the pharmacokinetics of the CYP3A4 substrate simvastatin. Clin. Pharmacol. Ther. 68(4), 384–390 (2000).
    • 24. Klein K, Zanger UM. Pharmacogenomics of cytochrome P450 3A4: recent progress toward the “missing heritability” problem. Front. Genet. 4, 12 (2013).
    • 25. Puranik YG, Birnbaum AK, Marino SE et al. Association of carbamazepine major metabolism and transport pathway gene polymorphisms and pharmacokinetics in patients with epilepsy. Pharmacogenomics 14(1), 35–45 (2013).
    • 26. Nakajima Y, Saito Y, Shiseki K et al. Hyplotype structures of EPHX1 and their effects on the metabolism of carbamazepine-10,11-epoxide in Japanese epileptic patients. Eur. J. Clin. Pharmacol. 61(1), 25–34 (2005).
    • 27. Yun W, Zhang F, Hu C et al. Effects of EPHX1, SCN1A and CYP3A4 genetic polymorphisms on plasma carbamazepine concentrations and pharmacoresistance in Chinese patients with epilepsy. Epilepsy Res. 107(3), 231–237 (2013).
    • 28. Zhu X, Yun W, Sun X, Qiu F, Zhao L, Guo Y. Effects of major transporter and metabolizing enzyme gene polymorphisms on carbamazepine metabolism in Chinese patients with epilepsy. Pharmacogenomics 15(15), 1867–1879 (2014).
    • 29. Ghodke-Puranik Y, Thorn CF, Lamba JK et al. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet. Genomics 23(4), 236–241 (2013).
    • 30. Wang P, Lin XQ, Cai WK et al. Effect of UGT2B7 genotypes on plasma concentration of valproic acid: a meta-analysis. Eur. J. Clin. Pharmacol. 74(4), 433–442 (2018).
    • 31. Ho PC, Abbott FS, Zanger UM, Chang TK. Influence of CYP2C9 genotypes on the formation of a hepatotoxic metabolite of valproic acid in human liver microsomes. Pharmacogenomics J. 3(6), 335–342 (2003).
    • 32. Gulcebi MI, Ozkaynakci A, Goren MZ, Aker RG, Ozkara C, Onat FY. The relationship between UGT1A4 polymorphism and serum concentration of lamotrigine in patients with epilepsy. Epilepsy Res. 95(1-2), 1–8 (2011). • Report on the association of the UGT142T>G polymorphism and lower serum concentrations of lamotrigine.
    • 33. Ma CL, Wu XY, Jiao Z, Hong Z, Wu ZY, Zhong MK. SCN1A, ABCC2 and UGT2B7 gene polymorphisms in association with individualized oxcarbazepine therapy. Pharmacogenomics 16(4), 347–360 (2015).
    • 34. Bruhn O, Cascorbi I. Polymorphisms of the drug transporters ABCB1, ABCG2, ABCC2 and ABCC3 and their impact on drug bioavailability and clinical relevance. Expert Opin. Drug Metab. Toxicol. 10, 1337–1354 (2014).
    • 35. Dallas S, Miller DS, Bendayan R. Multidrug resistance-associated proteins: expression and function in the central nervous system. Pharmacol. Rev. 58(2), 140–161 (2006).
    • 36. Potschka H, Brodie MJ. Pharmacoresistance. Handb. Clin. Neurol. 108, 741–757 (2012).
    • 37. Siddiqui A, Kerb R, Weale ME et al. Association of multidrug resistance in epilepsy with a polymorphism in the drug-transporter gene ABCB1. N. Engl. J. Med. 348(15), 1442–1448 (2003).
    • 38. Manna I, Gambardella A, Labate A et al. Polymorphism of the multidrug resistance 1 gene MDR1/ABCB1 C3435T and response to antiepileptic drug treatment in temporal lobe epilepsy. Seizure 24, 124–126 (2015).
    • 39. Sills GJ, Mohanraj R, Butler E et al. Lack of association between the C3435T polymorphism in the human multidrug resistance (MDR1) gene and response to antiepileptic drug treatment. Epilepsia 46(5), 643–647 (2005).
    • 40. Haerian BS, Lim KS, Tan CT, Raymond AA, Mohamed Z. Association of ABCB1 gene polymorphisms and their haplotypes with response to antiepileptic drugs: a systematic review and meta-analysis. Pharmacogenomics 12(5), 713–725 (2011).
    • 41. Orlandi A, Paolino MC, Striano P, Parisi P. Clinical reappraisal of the influence of drug-transporter polymorphisms in epilepsy. Expert Opin. Drug Metab. Toxicol. 14(5), 505–512 (2018).
    • 42. Qian L, Fang S, Yan YL, Zeng SS, Xu ZJ, Gong ZC. The ABCC2 c.-24C>T polymorphism increases the risk of resistance to antiepileptic drugs: a meta-analysis. J. Clin. Neurosci. 37, 6–14 (2017).
    • 43. Wang Y, Tang L, Pan J et al. The recessive model of MPR2 G1249A polymorphism decrease the risk of drug-resistant in Asian epilepsy: a systematic review and meta-analysis. Epilepsy Res. 112, 56–63 (2015).
    • 44. Chen J, Su QB, Tao YQ et al. ABCC2 rs2273697 is associated with valproic acid concentrations in patients with epilepsy on valproic acid monotherapy. Pharmazie 73(5), 279–282 (2018).
    • 45. Klarica Domjanović I, Lovrić M, Trkulja V et al. Interaction between ABCG2 421C>A polymorphism and valproate in their effects on steady-state disposition of lamotrigine in adults with epilepsy. Br. J. Clin. Pharmacol. 84(9), 2106–2119 (2018). • This is first study indicating a potentially relevant interaction between ABCG2 421C>A polymorphism and valproate in their effects on lamotrigine disposition.
    • 46. Basic S, Hajnsek S, Bozina N et al. The influence of C3435T polymorphism of ABCB1 gene on penetration of phenobarbital across the blood–brain barrier in patients with generalized epilepsy. Seizure 17(6), 524–530 (2008).
    • 47. Tate SK, Depondt C, Sisodiya SM et al. Genetic predictors of the maximum doses patients receive during clinical use of the anti-epileptic drugs carbamazepine and phenytoin. Proc. Natl Acad. Sci. USA 102(15), 5507–5512 (2005).
    • 48. Zhou BT, Zhou QH, Yin JY et al. Effects of SCN1A and GABA receptor genetic polymorphisms on carbamazepine tolerability and efficacy in Chinese patients with partial seizures: 2-year longitudinal clinical follow-up. CNS Neurosci. Ther. 18(7), 566–572 (2012).
    • 49. Shi L, Zhu M, Li H et al. SCN1A and SCN2A polymorphisms are associated with response to valproic acid in Chinese epilepsy patients. Eur. J. Clin. Pharmacol. 75(5), 655–663 (2019).
    • 50. Haerian BS, Baum L, Kwan P, Tan HJ, Raymond AA, Mohamed Z. SCN1A, SCN2A and SCN3A gene polymorphisms and responsiveness to antiepileptic drugs: a multicenter cohort study and meta-analysis. Pharmacogenomics 14(10), 1153–1166 (2013).
    • 51. Markovic I, Pejanovic-Skobic N, Bozina N, Susak-Sporis I, Sporis D, Basic S. The lack of influence of IVS5-91 G>A polymorphism of the SCN1A gene on efficacy of lamotrigine in patients with focal epilepsy. Neurol. Res. 1–6 (2019). doi:10.1080/01616412.2019.1635321 (Epub ahead of print).
    • 52. Franco V, Perucca E. The pharmacogenomics of epilepsy. Expert Rev. Neurother. 15(10), 1161–1170 (2015).
    • 53. Qu J, Zhang Y, Yang ZQ et al. Gene-wide tagging study of the association between KCNT1 polymorphisms and the susceptibility and efficacy of genetic generalized epilepsy in Chinese population. CNS Neurosci. Ther. 20(2), 140–146 (2014).
    • 54. Lynch JM, Tate SK, Kinirons P et al. No major role of common SV2A variation for predisposition or levetiracetam response in epilepsy. Epilepsy Res. 83(1), 44–51 (2009).
    • 55. Leen WG, Klepper J, Verbeek MM et al. Glucose transporter-1-deficiency syndrome: the expanding clinical and genetic spectrum of a treatable disorder. Brain 133(3), 655–670 (2010).
    • 56. Mills PB, Struys E, Jakobs C et al. Mutations in individuals with pyridoxine-dependent seizures. Nat. Med. 12(3), 307–309 (2006).
    • 57. Perucca P, Perucca E. Identifying mutations in epilepsy genes. Impact on treatment selection. Epilepsy Res. 152, 18–30 (2019).
    • 58. Gunthorpe MJ, Large CH, Sankar R. The mechanism of action of retigabine (ezogabine), a first-in-class K+ channel opener for the treatment of epilepsy. Epilepsia 53(3), 412–424 (2012).
    • 59. Pierson TM, Yuan H, Marsh ED et al. GRIN2A mutation and early-onset epileptic encephalopathy: personalized therapy with memantine. Ann. Clin. Transl. Neurol. 1(3), 190–198 (2014).
    • 60. Fricke-Galindo I, Llerena A, López-López M. An update on HLA alleles associated with adverse drug reactions. Drug Metab. Pers. Ther. 32(2), 73–87 (2017).
    • 61. Mullan KA, Anderson A, Illing PT, Kwan P, Purcell AW, Mifsud NA. HLA associated antiepileptic drug-induced cutaneous adverse reactions. HLA 93(6), 417–435 (2019).
    • 62. Chen CB, Abe R, Pan RY et al. An updated review of the molecular mechanisms in drug hypersensitivity. J. Immunol. Res. 2018, 6431694 (2018).
    • 63. Chung WH, Hung SI, Hong HS et al. A marker for Stevens–Johnson syndrome. Nature 428(6982), 486 (2004). •• First report on the strong association between a genetic marker, HLA–B*1502, and Stevens–Johnson syndrome induced by carbamazepine.
    • 64. Liu Y, Yu Y, Nie X, Zhao L, Wang X. Association between HLA-B*15:02 and oxcarbazepine-induced cutaneous adverse reaction: a meta-analysis. Pharmacogenomics 19(6), 547–552 (2018).
    • 65. Ozeki T, Mushiroda T, Yowang A et al. Genome-wide association study identifies HLA-A*3101 allele as a genetic risk factor for carbamazepine-induced cutaneous adverse drug reactions in Japanese population. Hum. Mol. Genet. 20, 1034–1041 (2011).
    • 66. McCormack M, Alfirevic A, Bourgeois S et al. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N. Engl. J. Med. 364(12), 1134–1143 (2011). • Report on the contribution of the of HLA-A*31:01 to the pathogenesis of carbamazepine-induced hypersensitivity reactions.
    • 67. Plumpton CO, Yip VL, Alfirevic A, Marson AG, Pirmohamed M, Hughes DA. Cost-effectiveness of screening for HLA-A*31:01 prior to initiation of carbamazepine in epilepsy. Epilepsia 56, 556–563 (2015).
    • 68. Saneto RP, Lee IC, Koenig MK et al. POLG DNA testing as an emerging standard of care before instituting valproic acid therapy for pediatric seizure disorders. Seizure 19(3), 140–146 (2010). •• Report on the mutations in POLG and the risk of sodium valproate-induced liver toxicity.
    • 69. Ingelman-Sundberg M, Cascorbi I. Pharmacogenomic or epigenomic biomarkers in drug treatment: two sides of the same medal? Clin. Pharmacol. Ther. 99, 478–480 (2016).
    • 70. Navarrete-Modesto V, Orozco-Suárez S, Feria-Romero IA, Rocha L. The molecular hallmarks of epigenetic effects mediated by antiepileptic drugs. Epilepsy Res. 149, 53–65 (2019).
    • 71. Kozyra M, Ingelman-Sundberg M, Lauschke V. Rare genetic variants in cellular transporters, metabolic enzymes and nuclear receptors can be important determinants of interindividual differences in drug response. Genet. Med. 19, 20–29 (2017). •• Significance of rare genetic variants for the overall pharmacogenomics variability.
    • 72. US Food and Drug Association. Table of pharmacogenomic biomarkers in drug labeling. http://www.fda.gov.