Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Exome sequencing allows detection of relevant pharmacogenetic variants in epileptic patients

Abstract

Beyond the identification of causal genetic variants in the diagnosis of Mendelian disorders, exome sequencing can detect numerous variants with potential relevance for clinical care. Clinical interventions can thus be conducted to improve future health outcomes for patients and their at-risk relatives, such as predicting late-onset genetic disorders accessible to prevention, treatment or identifying differential drug efficacy and safety. To evaluate the interest of such pharmacogenetic information, we designed an “in house” pipeline to determine the status of 122 PharmGKB (Pharmacogenomics Knowledgebase) variant-drug combinations in 31 genes. This pipeline was applied to a cohort of 90 epileptic patients who had previously an exome sequencing (ES) analysis, to determine the frequency of pharmacogenetic variants. We performed a retrospective analysis of drug plasma concentrations and treatment efficacy in patients bearing at least one relevant PharmGKB variant. For PharmGKB level 1A variants, CYP2C9 status for phenytoin prescription was the only relevant information. Nineteen patients were treated with phenytoin, among phenytoin-treated patients, none were poor metabolizers and four were intermediate metabolizers. While being treated with a standard protocol (10–23 mg/kg/30 min loading dose followed by 5 mg/kg/8 h maintenance dose), all identified intermediate metabolizers had toxic plasma concentrations (20 mg/L). In epileptic patients, pangenomic sequencing can provide information about common pharmacogenetic variants likely to be useful to guide therapeutic drug monitoring, and in the case of phenytoin, to prevent clinical toxicity caused by high plasma levels.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Description of the study design.
Fig. 2: Kinetics of phenytoin concentration in the 4 patients. Primary vertical axis representing the phenytoin concentration and secondary vertical axis representing the administered dose.

Similar content being viewed by others

Data availability

For data sharing requests, please contact SV or YD at simon.verdez@chu-dijon.fr or yannis.duffourd@u-bourgogne.fr respectively.

Code availability

Code is available at the following link: http://gitlab.gad-bioinfo.org/gad-public/pharmAnnot/tree/public

References

  1. Hesselink D. Genetic polymorphisms of the CYP3A4, CYP3A5, and MDR-1 genes and pharmacokinetics of the calcineurin inhibitors cyclosporine and tacrolimus. Clin Pharmacol Therapeutics. 2003;74:245–54.

    Article  CAS  Google Scholar 

  2. Relling MV, Evans WE. Pharmacogenomics in the clinic. Nature 2015;526:343–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sim SC, Kacevska M, Ingelman-Sundberg M. Pharmacogenomics of drug-metabolizing enzymes: a recent update on clinical implications and endogenous effects. Pharmacogenomics J. 2013;13:1–11.

    Article  CAS  PubMed  Google Scholar 

  4. Relling MV, Klein TE, Gammal RS, Whirl-Carrillo M, Hoffman JM, Caudle KE. The clinical pharmacogenetics implementation consortium: 10 years later. Clin Pharmacol Ther. 2020;107:171–5.

    Article  PubMed  Google Scholar 

  5. Bank PCD, Caudle KE, Swen JJ, Gammal RS, Whirl‐Carrillo M, Klein TE, et al. Comparison of the guidelines of the clinical pharmacogenetics implementation Consortium and the Dutch pharmacogenetics Working group. Clin Pharm Ther. 2018;103:599–618.

    Article  CAS  Google Scholar 

  6. Visscher H, Ross CJD, Rassekh SR, Barhdadi A, Dubé MP, Al-Saloos H, et al. Canadian Pharmacogenomics Network for Drug Safety Consortium Pharmacogenomic prediction of anthracycline-induced cardiotoxicity in children. J Clin Oncol. 2012;30:1422–8.

    Article  PubMed  Google Scholar 

  7. Picard N, Boyer JC, Etienne‐Grimaldi MC, Barin‐Le GC, Thomas F, Loriot MA. Pharmacogenetics‐based personalized therapy: levels of evidence and recommendations from the French Network of Pharmacogenetics (RNPGx). Therapie. 2017;72:185–92.

    Article  PubMed  Google Scholar 

  8. Whirl-Carrillo M, McDonagh EM, Hebert JM, Gong L, Sangkuhl K, Thorn CF, et al. Pharmacogenomics Knowledge for Personalized Medicine. Clin Pharm Ther. 2012;92:414–7.

    Article  CAS  Google Scholar 

  9. Thijs RD, Surges R, O’Brien TJ, Sander JW. Epilepsy in adults. Lancet. 2019;393:689–701.

    Article  PubMed  Google Scholar 

  10. Orsini A, Zara F, Striano P. Recent advances in epilepsy genetics. Neurosci Lett. 2018;667:4–9.

    Article  CAS  PubMed  Google Scholar 

  11. Symonds JD, Zuberi SM, Johnson MR. Advances in epilepsy gene discovery and implications for epilepsy diagnosis and treatment. Curr Opin Neurol. 2017;30:193–9.

    Article  PubMed  Google Scholar 

  12. Bruel A-L, Levy J, Elenga N, Defo A, Favre A, Lucron H, et al. INTU -related oral-facial-digital syndrome type VI: A confirmatory report. Clin Genet. 2018;93:1205–9.

    Article  CAS  PubMed  Google Scholar 

  13. Thevenon J, Duffourd Y, Masurel-Paulet A, Lefebvre M, Feillet F, El Chehadeh-Djebbar S, et al. Diagnostic odyssey in severe neurodevelopmental disorders: toward clinical whole-exome sequencing as a first-line diagnostic test. Clin Genet. 2016;89:700–7.

    Article  CAS  PubMed  Google Scholar 

  14. Bruel A-L, Nambot S, Quéré V, Vitobello A, Thevenon J, et al. Increased diagnostic and new genes identification outcome using research reanalysis of singleton exome sequencing. Eur J Hum Genet. 2019;27:1519–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cousin MA, Matey ET, Blackburn PR, Boczek NJ, McAllister TM, Kruisselbrink TM, et al. Pharmacogenomic findings from clinical whole exome sequencing of diagnostic odyssey patients. Mol Genet Genom Med. 2017;5:269–79.

    Article  Google Scholar 

  16. Blom S. trigeminal neuralgia: its treatment with a new anticonvulsant drug (G-32883). Lancet. 1962;279:839–40.

    Article  Google Scholar 

  17. Arif H, Buchsbaum R, Weintraub D, Koyfman S, Salas-Humara C, Bazil CW, et al. Comparison and predictors of rash associated with 15 antiepileptic drugs. Neurology. 2007;68:1701–9.

    Article  CAS  PubMed  Google Scholar 

  18. Karnes JH, Rettie AE, Somogyi AA, Huddart R, Fohner AE, Formea CM, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C9 and HLA‐B Genotypes and Phenytoin Dosing: 2020 Update. Clin. Pharmacol. Ther. 2020;96:542–8.

    Google Scholar 

  19. Locharernkul C, Loplumlert J, Limotai C, Korkij W, Desudchit T, Tongkobpetch S, et al. Carbamazepine and phenytoin induced Stevens-Johnson syndrome is associated with HLA-B*1502 allele in Thai population. Epilepsia. 2008;49:2087–91.

    Article  PubMed  Google Scholar 

  20. Chung WH, Hung SI, Hong HS, Hsih MS, Yang LC, Ho HC, et al. A marker for Stevens–Johnson syndrome. Nature. 2004;428:486.

    Article  CAS  PubMed  Google Scholar 

  21. Anzenbacher P, Anzenbacherová E. Cytochromes P450 and metabolism of xenobiotics: CMLS. Cell Mol Life Sci. 2001;58:737–47.

    Article  CAS  PubMed  Google Scholar 

  22. Kidd RS, Curry TB, Gallagher S, Edeki T, Blaisdell J, Goldstein JA. Identification of a null allele of CYP2C9 in an African–American exhibiting toxicity to phenytoin. Pharmacogenetics Genomics. 2001;11:803–8.

    Article  CAS  Google Scholar 

  23. Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J. 2005;5:6–13.

    Article  CAS  PubMed  Google Scholar 

  24. Karnes JH, Rettie AE, Somogyi AA, Huddart R, Fohner AE, Formea CM, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C9 and HLA-B Genotypes and Phenytoin Dosing: 2020 Update. Clin Pharmacol Therap. 2020;109:302–9.

    Article  Google Scholar 

  25. Thauvin-Robinet C, Thevenon J, Nambot S, Delanne J, Kuentz P, Bruel AL, et al. Secondary actionable findings identified by exome sequencing: expected impact on the organisation of care from the study of 700 consecutive tests. Eur J Hum Genet. 2019;27:1197–214.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Ruden DM, Cingolani P, Patel VM, Coon M, Nguyen T, Land SJ, et al. Using Drosophila melanogaster as a Model for Genotoxic Chemical Mutational Studies with a New Program, SnpSift. Front Gene. 2012;3:35.

    Google Scholar 

  27. Ka S, Lee S, Hong J, Cho Y, Sung J, Kim HN, et al. HLAscan: genotyping of the HLA region using next-generation sequencing data. BMC Bioinforma. 2017;18:1–11.

    Article  Google Scholar 

  28. Giancarlo GM, Venkatakrishnan K, Granda BW, von Moltke LL, Greenblatt DJ. Relative contributions of CYP2C9 and 2C19 to phenytoin 4-hydroxylation in vitro: inhibition by sulfaphenazole, omeprazole, and ticlopidine. Eur J Clin Pharm. 2001;57:31–36.

    Article  CAS  Google Scholar 

  29. Bajpai M, Roskos LK, Shen DD, Levy RH. Roles of cytochrome P4502C9 and cytochrome P4502C19 in the stereoselective metabolism of phenytoin to its major metabolite. Drug Metab Dispos. 1996;24:1401–3.

    CAS  PubMed  Google Scholar 

  30. van der Weide J, Steijns LS, van Weelden MJ, de Haan K. The effect of genetic polymorphism of cytochrome P450 CYP2C9 on phenytoin dose requirement. Pharmacogenetics. 2001;11:87–91.

    Google Scholar 

  31. Kudo T, Endo Y, Taguchi R, Yatsu M, Ito K. Metronidazole reduces the expression of cytochrome P450 enzymes in HepaRG cells and cryopreserved human hepatocytes. Xenobiotica. 2015;45:413–9.

    Article  CAS  PubMed  Google Scholar 

  32. Fohner AE, Ranatunga DK, Thai KK, Lawson BL, Rischx N, Oni-Orisan A, et al. Assessing the clinical impact of CYP2C9 pharmacogenetic variation on phenytoin prescribing practice and patient response in an integrated health system. Pharmacogenet Genomics. 2019;29:192–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dorado P, López-Torres E, Peñas-Lledó EM, Martínez-Antón J, Llerena A. Neurological toxicity after phenytoin infusion in a pediatric patient with epilepsy: influence of CYP2C9, CYP2C19 and ABCB1 genetic polymorphisms. Pharmacogenomics J. 2013;13:359–61.

    Article  CAS  PubMed  Google Scholar 

  34. Cresteil T, Beaune P, Kremers P, Celier C, Guengerich FP, Leroux J-P. Immunoquantification of epoxide hydrolase and cytochrome P-450 isozymes in fetal and adult human liver microsomes. Eur J Biochem. 1985;151:345–50.

    Article  CAS  PubMed  Google Scholar 

  35. Koukouritaki SB, Manro JR, Marsh SA, Stevens JC, Rettie AE, McCarver DG. et al. Developmental Expression of Human Hepatic CYP2C9 and CYP2C19. J Pharm Exp Ther. 2004;308:965–74.

    Article  CAS  Google Scholar 

  36. McCormack M, Alfirevic A, Bourgeois S, et al. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N. Engl J Med. 2011;364:1134–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yip VL, Marson AG, Jorgensen AL, Pirmohamed M, Alfirevic A. HLA genotype and carbamazepine-induced cutaneous adverse drug reactions: a systematic review. Clin Pharm Ther. 2012;92:757–65.

    Article  CAS  Google Scholar 

  38. Zhou Y, Krebs K, Milani L, Lauschke VM. Global Frequencies of Clinically Important HLA Alleles and Their Implications For the Cost‐Effectiveness of Preemptive Pharmacogenetic Testing. Clin Pharmacol Ther. 2021;109:160–74.

    Article  PubMed  Google Scholar 

  39. Xue Y, Ankala A, Wilcox WR, Hegde MR. Solving the molecular diagnostic testing conundrum for Mendelian disorders in the era of next-generation sequencing: single-gene, gene panel, or exome/genome sequencing. Genet Med. 2015;17:444–51.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the University of Burgundy Centre de Calcul (CCuB) for providing technical support and management of the informatics platform. This work was supported by grants from Dijon University Hospital, the ISITE-BFC (PIA ANR) and the European Union through the FEDER programs. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. The authors thank Suzanne Rankin from the Dijon University Hospital, Nicole Dorssers from simon fraser university and Cynthia Reichling for proofreading the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

SV, PG, LF, NP and YD designed the study. SV, QT performed the clinical analysis. AV, FTMT, CP, interpreted exome data. FTMT, AV and CP performed the molecular laboratory work. SV, YD and ET performed the bioinformatics analysis. All the authors contributed to the writing and review of the paper.

Corresponding authors

Correspondence to Simon Verdez or Yannis Duffourd.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Verdez, S., Thomas, Q., Garret, P. et al. Exome sequencing allows detection of relevant pharmacogenetic variants in epileptic patients. Pharmacogenomics J 22, 258–263 (2022). https://doi.org/10.1038/s41397-022-00280-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41397-022-00280-w

Search

Quick links