Skip to main content
Log in

Pharmacological Management of the Genetic Generalised Epilepsies in Adolescents and Adults

  • Review Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

Common genetic generalised epilepsy syndromes encountered by clinicians include childhood and juvenile absence epilepsies, juvenile myoclonic epilepsy and generalised tonic–clonic seizures on awakening. Treatment of these syndromes involves largely the use of broad-spectrum antiseizure drugs. Those effective for the generalised epilepsies include sodium valproate, phenobarbital, ethosuximide, clobazam, clonazepam, lamotrigine, levetiracetam, topiramate, zonisamide and, more recently, perampanel and brivaracetam. Results from the few rigorous studies comparing outcomes with drugs for genetic generalised epilepsies show valproate to be the most effective. The majority of patients with genetic generalised epilepsy syndromes will become seizure free on antiseizure monotherapy; those for whom control proves elusive may benefit from combination regimens. Early counselling regarding management may assist the patient to come to terms with their diagnosis and improve long-term outcomes. Treatment can be lifelong in some individuals, although others may remain seizure free without medication. Choice of antiseizure medication depends on the efficacy for specific seizure types, as well as tolerability. For patients prescribed comedication, drug interactions should be considered. In particular, for young women taking oral hormonal contraceptives, ≥ 200 mg/day of topiramate can decrease the circulating concentration of ethinylestradiol and ≥ 12 mg/day of perampanel can induce levonorgestrel metabolism. The use of valproate in women of childbearing potential is limited by associated teratogenic and neurodevelopmental effects in offspring. Given that valproate is often the antiseizure drug of choice for genetic generalised epilepsies, this creates a dilemma for patients and clinicians. Decision making can be aided by comprehensive assessment and discussion of treatment options. Psychiatric comorbidities are common in adolescents and adults with genetic generalised epilepsies. These worsen the prognosis, both in terms of seizure control and quality of life. Attendant lifestyle issues can impact significantly on the individual and society. Frontal lobe dysfunction, which can present in patients with juvenile myoclonic epilepsy, can adversely affect the long-term outlook, regardless of the nature of seizure control. Ongoing management requires consideration of psychosocial and behavioural factors that can complicate diagnosis and treatment. An assured supportive attitude by the neurologist can be an important contributor to a positive outcome. The mechanisms underlying genetic generalised epilepsies, including genetic abnormalities, are unclear at present. As the pathophysiology is unravelled, this may lead to the development of novel therapies and improved outcomes for patients with these syndromes.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Brodie MJ. Tolerability and safety of commonly used antiepileptic drugs in adolescents and adults: a clinician’s overview. CNS Drugs. 2017;31:135–47.

    Article  CAS  PubMed  Google Scholar 

  2. Scheffer IE, Berkovic S, Capovilla G, et al. ILAE classification of the epilepsies: position paper of the ILAE Commission for Classification and Terminology. Epilepsia. 2017;58:512–21.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Striano P, Nobile C. The genetic basis of juvenile myoclonic epilepsy. Lancet Neurol. 2018;17:493–5.

    Article  PubMed  Google Scholar 

  4. Glauser TA, Cnaan A, Shinnar S, et al. Ethosuximide, valproic acid and lamotrigine in childhood absence epilepsy. New Engl J Med. 2010;362:790–9.

    Article  CAS  PubMed  Google Scholar 

  5. Wallace SI. Myoclonus and epilepsy in childhood: a review of treatment with valproate, ethosuximide, lamotrigine and zonisamide. Epilepsy Res. 1998;29:147–54.

    Article  CAS  PubMed  Google Scholar 

  6. Ethosuximide; indications and dose. In: National Institute for Health and Care Excellence. British National Formulary. https://bnf.nice.org.uk/drug/ethosuximide.html#indicationsAndDoses. Accessed 15 Dec 2019.

  7. Glauser T, Ben-Menachem E, Bourgeois B, et al. Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 2013;54:551–63.

    Article  CAS  PubMed  Google Scholar 

  8. Berkovic SF, Knowlton RC, Leroy RF, et al. Placebo-controlled study of levetiracetam in idiopathic generalised epilepsy. Neurology. 2007;69:1751–60.

    Article  CAS  PubMed  Google Scholar 

  9. French JA, Krauss G, Wechsler R, et al. Adjunctive perampanel for treatment of drug-resistant primary generalized tonic-clonic seizures in patients with idiopathic generalized epilepsy: a double-blind randomized placebo-controlled phase III trial. Neurology. 2015;85:950–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yamauchi T, Aikawa H. Efficacy of zonisamide: our experience. Seizure. 2004;(Suppl. 1):S41–8.

  11. Marinas A, Villaneuva V, Giraldez BG, et al. Efficacy and tolerability of zonisamide in idiopathic generalized epilepsy. Epileptic Disord. 2009;11:61–6.

    Article  PubMed  Google Scholar 

  12. Stephen LJ, Kelly K, Wilson EA, Parker P, Brodie MJ. A prospective audit of adjunctive zonisamide in an everyday clinical setting. Epilepsy Behav. 2010;17:455–60.

    Article  PubMed  Google Scholar 

  13. Velizarova R, Crespel A, Genton P. Zonisamide for refractory juvenile absence epilepsy. Epilepsy Res. 2014;7:1263–6.

    Article  CAS  Google Scholar 

  14. Janz D, Christian W. Impulsive petit mal. Deutsche Zeitschrift für Nervenheilkunde. 1957;176:346–88.

    Google Scholar 

  15. Shorvon SD, Bermejo PE, Gibbs AA, Huberfeld G, Kalviainen R. Antiepileptic drug treatment of generalized tonic-clonic seizures: an evaluation of regulatory data and five criteria for drug selection. Epilepsy Behav. 2018;82:91–103.

    Article  PubMed  Google Scholar 

  16. Striano P, Belcastro V. Treating mycolonic epilepsy in children: state of the art. Exp Opin Pharmacother. 2013;14:1355–61.

    Article  CAS  Google Scholar 

  17. Kasteleijn-Nolst Trenite DG, Genton P, et al. Evaluation of brivaracetam, a novel SV2A ligand, in the photosensitivity model. Neurology. 2007;69:1027–34.

    Article  CAS  PubMed  Google Scholar 

  18. Matagne A, Margineau DG, Kenda B, et al. Anti-convulsive and anti-epileptic properties of brivaracetam (ucb34714), a high-affinity ligand for the synaptic vesicle protein, SV2A. Br J Pharmacol. 2008;154:1662–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kwan P, Trinka E, Van Paesschen W, Rektor I, Johnson ME, Lu S. Adjunctive brivaracetam for uncontrolled focal and generalized epilepsies: results of a phase III, double-blind, randomized, placebo-controlled, flexible-dose trial. Epilepsia. 2014;55:38–46.

    Article  CAS  PubMed  Google Scholar 

  20. Berg AT, Berkovic SF, Brodie MJ, et al. Revised terminology and concepts for organisation of the epilepsies: report of the ILAE Commission on Classification and Terminology. Epilepsia. 2015;51:676–85.

    Article  Google Scholar 

  21. Balagura G, Lapadre G, Verrotti A, Striano P. Moving beyond sodium valproate: choosing the right anti-epileptic drug in children. Exp Opin Pharmacother. 2019;20:1449–56.

    Article  CAS  Google Scholar 

  22. Kwan P, Sills GJ, Brodie MJ. The mechanisms of action of commonly used antiepileptic drugs. Pharmacol Ther. 2001;90:21–34.

    Article  CAS  PubMed  Google Scholar 

  23. Tomson T, Battino D, Perucca E. Valproic acid after five decades of use in epilepsy: time to reconsider the indications of a time-honoured drug. Lancet Neurol. 2016;15:210–8.

    Article  CAS  PubMed  Google Scholar 

  24. Tomson T, Battino D, Bonizzoni E, Craig J, Lindhout D, Perucca E. Comparative risk of major congenital malformations with eight different antiepileptic drugs: a prospective cohort study of the EURAP registry. Lancet Neurol. 2018;17:530–8.

    Article  CAS  PubMed  Google Scholar 

  25. Patsalos PN, Perucca E. Clinically important drug interactions in epilepsy: interactions between antiepileptic drugs and other drugs. Lancet Neurol. 2003;2:473–81.

    Article  CAS  PubMed  Google Scholar 

  26. Machado RA, Garcia VF, Astencio AG, Cuartas VB. Efficacy and tolerability of lamotrigine in juvenile myoclonic epilepsy: a prospective, unblinded randomized controlled trial. Seizure. 2013;22:846–55.

    Article  PubMed  Google Scholar 

  27. Brodie MJ. Modern management of juvenile myoclonic epilepsy. Expert Rev Neurother. 2016;16:681–8.

    Article  CAS  PubMed  Google Scholar 

  28. Brodie MJ, Yuen AW. Lamotrigine substitution study: evidence for synergism with sodium valproate? 105 Study Group. Epilepsy Res. 1997;26:423–32.

    Article  CAS  PubMed  Google Scholar 

  29. Brodie MJ. Sodium channel blockers in the treatment of epilepsy. CNS Drugs. 2017;31:527–34.

    Article  CAS  PubMed  Google Scholar 

  30. Guberman AH, Besag FM, Brodie MJ, et al. Lamotrigine associated rash: risk benefit comorbidities in adults and children. Epilepsia. 1999;40:985–91.

    Article  CAS  PubMed  Google Scholar 

  31. Gaffield ME, Culwell KR, Lee CR. The use of hormonal contraception among women taking anticonvulsant therapy. Contraception. 2011;83:16–29.

    Article  CAS  PubMed  Google Scholar 

  32. Perucca EA. Pharmacological and clinical review on topiramate, a new antiepileptic drug. Pharmacol Res. 1997;35:241–56.

    Article  CAS  PubMed  Google Scholar 

  33. Lee S, Sziklas V, Andermann F, et al. The effect of adjunctive topiramate on cognitive function in patients with epilepsy. Epilepsia. 2003;44:339–47.

    Article  CAS  PubMed  Google Scholar 

  34. Mula M, Trimble MR, Lhatoo SD, Sander JW. Topiramate and psychiatric adverse events in patients with epilepsy. Epilepsia. 2003;44:659–63.

    Article  PubMed  Google Scholar 

  35. Veiby G, Daltveit AK, Engelsen BA, Gilhus NE. Fetal growth restriction and birth defects with newer and older antiepileptic drugs during pregnancy. J Neurol. 2014;261:579–88.

    Article  CAS  PubMed  Google Scholar 

  36. Brodie MJ, Mintzer S, Pack AM, Gidal BE, Vecht CJ, Schmidt D. Enzyme induction with antiepileptic drugs: cause for concern? Epilepsia. 2013;54:11–27.

    Article  CAS  PubMed  Google Scholar 

  37. Rosenfield WE, Doose DR, Walker SA, Nayak RK. Effect of topiramate on the pharmacokinetics of an oral contraceptive containing norethindrone and ethinyloestradiol. Epilepsia. 1997;38:317–23.

    Article  Google Scholar 

  38. Kwan P, Brodie MJ. Emerging drugs for epilepsy. Expert Opin Emerg Drugs. 2007;12:407–22.

    Article  CAS  PubMed  Google Scholar 

  39. White JR, Walczak TS, Leppik IE, et al. Discontinuation of levetiracetam because of behavioural side effects. Neurology. 2003;61:1218–21.

    Article  CAS  PubMed  Google Scholar 

  40. Hirsch LJ, Arif H, Buchsbaum R, et al. Effect of age and comedication on levetiracetam pharmacokinetics and tolerability. Epilepsia. 2007;48:1351–9.

    Article  CAS  PubMed  Google Scholar 

  41. Brodie MJ. Perampanel. In: Shorvon S, Perucca E, Engel J, editors. The treatment of epilepsy. 4th ed. Hoboken: Wiley-Blackwell; 2016. p. 546–54.

    Google Scholar 

  42. Steinhoff BJ, Ben-Menachem E, Ryvlin P, et al. Efficacy and safety of adjunctive perampanel for the treatment of refractory partial seizures: a pooled analysis of three phase III studies. Epilepsia. 2013;54:1481–9.

    Article  CAS  PubMed  Google Scholar 

  43. Brodie MJ, Stephen LJ. Prospective audit with adjunctive perampanel: preliminary observations in focal epilepsy. Epilepsy Behav. 2016;54:100–3.

    Article  PubMed  Google Scholar 

  44. Ettinger AB, Lopresti A, Yang H, et al. Psychiatric and behavioural adverse events in randomized clinical studies of the non-competitive AMPA receptor antagonist perampanel. Epilepsia. 2015;56:1252–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gidal BE, Laurenza A, Hussein Z, et al. Perampanel efficacy and tolerability with enzyme-inducing AEDs in patients with epilepsy. Neurology. 2015;84:1–9.

    Article  CAS  Google Scholar 

  46. Rock DM, Macdonald RL, Taylor CP. Blockade of sustained repetitive action potentials in cultured spinal cord neurons by zonisamide (AD 810, CI 912), a novel anticonvulsant. Epilepsy Res. 1989;3:138–43.

    Article  CAS  PubMed  Google Scholar 

  47. Kito M, Machara M, Watanabe K. Mechanisms of T-type calcium channel blockade by zonisamide. Seizure. 1996;5:115–9.

    Article  CAS  PubMed  Google Scholar 

  48. Piedad J, Rickards H, Besag FG, Cavana AE. Beneficial and adverse psychotropic effects of antiepileptic drugs in patients with epilepsy. CNS Drugs. 2012;26:319–35.

    Article  CAS  PubMed  Google Scholar 

  49. Wroe S. Zonisamide and renal calculi in patients with epilepsy: how big an issue? Curr Med Res Opin. 2007;23:1765–73.

    Article  CAS  PubMed  Google Scholar 

  50. Low PA, James S, Peschel T, Leon R, Rothstein A. Zonisamide and associated oligohydrosis and hypothermia. Epilepsy Res. 2004;62:27–34.

    Article  CAS  PubMed  Google Scholar 

  51. Goren MJ, Onat F. Ethosuximide: from bench to bedside. CNS Drugs. 2007;13:224–39.

    Article  CAS  Google Scholar 

  52. Brodie MJ, Kwan P. Current position of phenobarbital in epilepsy and its future. Epilepsia. 2012;53(Suppl. 8):40–6.

    Article  CAS  PubMed  Google Scholar 

  53. Zaccara G, Perucca E. Interactions between antiepileptic drugs, and between antiepileptic drugs and other drugs. Epileptic Disord. 2014;16:403–32.

    Google Scholar 

  54. Pernea M, Sutcliffe AG. Clobazam and its use in epilepsy. Pediatr Rep. 2016;8:34–8.

    Article  CAS  Google Scholar 

  55. Sankar R. GABA (A) receptor physiology and its relationship to the mechanism of action of the 1,5-benzodiazepine clobazam. CNS Drugs. 2012;26:229–44.

    Article  CAS  PubMed  Google Scholar 

  56. Ng Y, Collins SD. Clobazam. Neurotherapeutics. 2007;4:138–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Gauthier AC, Mattson RH. Clobazam: a safe, efficacious and newly rediscovered therapeutic for epilepsy. CNS Neurosci Ther. 2015;21:543–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Browne T. Clonazepam. N Engl J Med. 1978;299:812–6.

    Article  CAS  PubMed  Google Scholar 

  59. Brodie MJ, Chung S, Wade A, et al. Clobazam and clonazepam use in epilepsy: results from a UK database incident user cohort study. Epilepsy Res. 2016;123:68–74.

    Article  CAS  PubMed  Google Scholar 

  60. Grabowska-Grzb A, Jedrzcejczak J, Naganska E, Fizer U. Risk factors for depression in people with epilepsy. Epilepsy Behav. 2006;8:411–7.

    Article  Google Scholar 

  61. Stephen LJ, Brodie MJ. Brivaracetam: a novel antiepileptic drug for focal-onset seizure. Therap Adv Neurol Dis. 2017;11:1–10.

    Google Scholar 

  62. Gillard M, Fuks B, Leclercq K, Matagne A. Binding characteristics of brivaracetam, a selective, high affinity SV2A ligand in rat, mouse and human brain: relationship to anticonvulsant properties. Br J Pharmacol. 2011;664:34–44.

    Google Scholar 

  63. Yates SL, Fakhoury T, Lang W, Eckhardt K, Borghs S, D’Souza J. An open-label, prospective, exploratory study of patients with epilepsy switching from levetiracetam to brivaracetam. Epilepsy Behav. 2015;52:165–8.

    Article  PubMed  Google Scholar 

  64. Zhu L, Chen D, Tao C, Xu D, Chen S, Liu L. The adverse event profile of brivaracetam: a meta-analysis of randomized controlled trials. Seizure. 2017;45:7–16.

    Article  PubMed  Google Scholar 

  65. D’Souza J, Perucca E. Brivaracetam. In: Shorvon S, Perucca E, Engel J, editors. The treatment of epilepsy. 4th ed. Chichester: Wiley; 2015. p. 418–30.

    Chapter  Google Scholar 

  66. Marson AG, Al-Kharusi AM, Alwaidh M, et al. The SANAD study of effectiveness of valproate, lamotrigine, or topiramate for generalised and unclassifiable epilepsy: an unblinded randomised controlled trial. Lancet. 2007;369:1016–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rosenow F, Schade-Brittinger C, Burchardi N, et al. The LaLiMo Trial: lamotrigine compared with levetiracetam in the initial 26 weeks of monotherapy for focal and generalised epilepsy: an open-label, prospective, randomised controlled multicenter study. J Neurol Neurosurg Psychiatry. 2012;83:1093–8.

    Article  PubMed  Google Scholar 

  68. Coppola G, Piccorossi A, Operto FF, Verrotti A. Anticonvulsant drugs for generalized tonic-clonic epilepsy. Exp Opin Pharmacother. 2017;18:925–36.

    Article  CAS  Google Scholar 

  69. Glauser TA, Cnaan A, Shinnar S, et al. Ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy: initial monotherapy outcomes at 12 months. Epilepsia. 2013;54:141–55.

    Article  CAS  PubMed  Google Scholar 

  70. Brigo F, Igwe SC, Lattanzi S. Ethosuximide, sodium valproate or lamotrigine for absence seizures in children and adolescents. Cochrane Database Syst Rev. 2019;2:CD003032.

  71. Nolan D, Lester S, Rau S, et al. Clinical use and efficacy of levetiracetam for absence epilepsies. J Child Neurol. 2018;34:94–8.

    Article  PubMed  Google Scholar 

  72. Levy A, Chen R. Myoclonus: pathophysiology and treatment options. Curr Treat Options Neurol. 2016;18:21.

    Article  PubMed  Google Scholar 

  73. Zhang Y, Chen J, Ren J, et al. Clinical features and treatment outcomes of juvenile myoclonic epilepsy patients. Epilepsia Open. 2019;4:302–8.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Velizarova R, Crespel A, Genton P, Serafini A, Gelisse P. Zonisamide for refractory juvenile absence epilepsy. Epilepsy Res. 2014;108:1263–6.

    Article  CAS  PubMed  Google Scholar 

  75. Steinig I, von Podwils F, Möddel G, et al. Postmarketing experience with brivaracetam in the treatment of epilepsies: a multicenter cohort study from Germany. Epilepsia. 2017;58:1208–16.

    Article  CAS  PubMed  Google Scholar 

  76. Grande-Martin A, Sopelana-Garay D, Pardal-Fernandez J, et al. Exceptional response to brivaracetam in a patient with refractory idiopathic generalized epilepsy and absence seizures. Epileptic Disord. 2018;20:60–4.

    PubMed  Google Scholar 

  77. Strzelczyk A, Kay L, Bauer S, et al. Use of brivaracetam in genetic generalized epilepsies and for acute, intravenous treatment of absence status epilepticus. Epilepsia. 2018;59:1549–56.

    Article  CAS  PubMed  Google Scholar 

  78. Reif PS, Männer A, Willems LM, et al. Intravenous lacosamide for treatment of absence status epilepticus in genetic generalized epilepsy: a case report and review of literature. Acta Neurol Scand. 2018;138:259–62.

    Article  CAS  PubMed  Google Scholar 

  79. Camfield CS, Striano P, Camfield PR. Epidemiology of juvenile myoclonic epilepsy. Epilepsy Behav. 2013;28(Suppl. 1):S15–7.

    Article  PubMed  Google Scholar 

  80. Maguire M, Marson AG, Ramaratnam S. Epilepsy (generalised). Clin Evid. 2012;2:1201.

    Google Scholar 

  81. Covanis A, Gupta AK, Jeavons PM. Sodium valproate: monotherapy and polytherapy. Epilepsia. 1982;23:693–720.

    Article  CAS  PubMed  Google Scholar 

  82. Calleja S, Salas-Puig J, Ribacoba R, et al. Evolution of juvenile myoclonic epilepsy treated from the outset with sodium valproate. Seizure. 2001;10:424–7.

    Article  CAS  PubMed  Google Scholar 

  83. Hitiris N, Brodie MJ. Evidence based treatment of idiopathic generalized epilepsies with older antiepileptic drugs. Epilepsia. 2005;4(Suppl. 9):149–53.

    Article  Google Scholar 

  84. Mendriatta MM, Aggarwal P. Clinical expression and EEG features of patients with juvenile myoclonic epilepsy (JME) from North India. Seizure. 2002;11:431–6.

    Article  Google Scholar 

  85. Chowdhury A, Brodie MJ. Pharmacological outcomes in juvenile myoclonic epilepsy over a 30 year period: support for sodium valproate. Epilepsy Res. 2016;119:62–6.

    Article  CAS  PubMed  Google Scholar 

  86. Gelisse P, Genton P, Thomas P, et al. Clinical factors of drug resistance in juvenile myoclonic epilepsy. J Neurol Neurosurg Psychiatry. 2001;70:240–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Jayalakshmi S, Vooturi S, Bana AK, et al. Factors associated with lack of response to valproic acid monotherapy in juvenile myoclonic epilepsy. Seizure. 2014;23:527–32.

    Article  PubMed  Google Scholar 

  88. Silvennoinen K, de Lange N, Zagaglia S, et al. Comparative effectiveness of antiepileptic drugs in juvenile myoclonic epilepsy. Epilepsia Open. 2019;4:420–30.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Levisohn PM, Holland KD. Topiramate or valproate in patients with juvenile myoclonic epilepsy: a randomized open-label comparison. Epilepsy Behav. 2007;10:547–52.

    Article  PubMed  Google Scholar 

  90. Park KM, Kim SH, Nho SK, et al. A randomized open-label observational study to compare the efficacy and tolerability between topiramate and valproate in juvenile myoclonic epilepsy. J Clin Neurosci. 2013;20:1079–82.

    Article  CAS  PubMed  Google Scholar 

  91. Liu J, Wang LN, Wang YP. Topiramate for juvenile myoclonic epilepsy. Cochrane Database Syst Rev. 2019;1:CD010008. https://doi.org/10.1002/14651858.cd010008.pub4.

  92. Noachter S, Andermann E, Meyisck P, et al. Levetiracetam for the treatment of idiopathic generalised epilepsy with myoclonic seizures. Neurology. 2008;70:607–16.

    Article  CAS  Google Scholar 

  93. Rosenfeld WE, Benbadis S, Edrich P, et al. Levetiracetam as add on therapy for idiopathic generalized epilepsy syndromes with onset during adolescence: analysis of two randomized double-blind placebo-controlled studies. Epilepsy Res. 2009;85:72–80.

    Article  CAS  PubMed  Google Scholar 

  94. Specchio LM, Gambardella A, Giallonardo AT, et al. Open label, long-term pragmatic study of levetiracetam in the treatment of juvenile myoclonic epilepsy. Epilepsy Res. 2006;17:32–9.

    Article  CAS  Google Scholar 

  95. Stephen LJ, Kelly K, Parker P, et al. Levetiracetam monotherapy outcomes from an epilepsy clinic. Seizure. 2011;20:554–7.

    Article  PubMed  Google Scholar 

  96. Biton V, Bourgeois BF; YTC/YTCE Study Investigators. Topiramate in patients with juvenile myoclonic epilepsy. Arch Neurol. 2005;62:170–8.

  97. De Araujo Filho GM, Pascalicchio TF, Lin K, Sousa PS, Yacubian MT. Neuropsychiatric profiles of patients with juvenile myoclonic epilepsy treated with valproate or topiramate. Epilepsy Behav. 2006;3:606–9.

    Article  Google Scholar 

  98. Stephen LJ, Sills GJ, Brodie MJ. Topiramate in refractory epilepsy: a prospective, observational study. Epilepsia. 2000;41:977–80.

    Article  CAS  PubMed  Google Scholar 

  99. Kothare SV, Valencia I, Khurana DS, et al. Efficacy and tolerability of zonisamide in juvenile myoclonic epilepsy. Epileptic Disord. 2004;6:267–70.

    PubMed  Google Scholar 

  100. Afra P, Adamolekum B. Lacosamide treatment of juvenile myoclonic epilepsy. Seizure. 2012;21:202–4.

    Article  PubMed  Google Scholar 

  101. Zangaladze A, Skidmore C. Lacosamide use in refractory idiopathic refractory primary generalized epilepsy. Epilepsy Behav. 2012;23:79–80.

    Article  PubMed  Google Scholar 

  102. Wechsler RT, Yates SL, Messenheimer J, Leroy R, Beller C, Doty P. Lacosamide for uncontrolled primary generalized tonic-clonic seizures: an open-label study with 59-week extension. Epilepsy Res. 2017;130:13–20.

    Article  CAS  PubMed  Google Scholar 

  103. Sodemann U, Moller HS, Blaabjerg M, Beier CP. Successful treatment of refractory absence status epilepticus with lacosamide. J Neurol. 2014;261:2025–7.

    Article  PubMed  Google Scholar 

  104. D’Orsi G, Pacillo T, Trivisano M, Pascarella MG, Ferrara MA, Specchio LM. Lacosamide in absence status epilepticus. Seizure. 2014;23:397–8.

    Article  PubMed  Google Scholar 

  105. Obeid T, Panayiotopoulos CP. Clonazepam in juvenile myoclonic epilepsy. Epilepsia. 1989;30:603–6.

    Article  CAS  PubMed  Google Scholar 

  106. Resor SR, Resor LD. Chronic acetazolamide monotherapy in the treatment of juvenile myoclonic epilepsy. Neurology. 1990;40:1677–81.

    Article  PubMed  Google Scholar 

  107. Ng M, Devinsky O. Vagus nerve stimulation for refractory idiopathic generalised epilepsy. Seizure. 2004;13:176–8.

    Article  PubMed  Google Scholar 

  108. Holmes MD, Silbergeld DL, Drouhard D, Wilensky JA, Ojemann L. Effect of vagus nerve stimulation on adults with pharmacoresistant generalized epilepsy syndromes. Seizure. 2004;13:340–5.

    Article  PubMed  Google Scholar 

  109. Biraben A, Allain H, Scarabin JM, et al. Exacerbation of juvenile myoclonic epilepsy with lamotrigine. Neurology. 2000;55:1757–8.

    Article  Google Scholar 

  110. Scottish Intercollegiate Guideline Network. Epilepsy and women’s health. In: Diagnosis and management of epilepsy in adults (SIGN publication no 143). May 2015. http://www.sign.ac.uk/assets/sign143.pdf. Accessed 15 Dec 2019.

  111. National Institute for Health and Care Excellence. Epilepsies: diagnosis and management. Clinical guideline. January 2012. https://www.nice.org.uk/guidance/cg137/resources/epilepsies-diagnosis-and-management-pdf-35109515407813. Accessed 15 Dec 2019.

  112. Gayatri NA. Aggravation of epilepsy by anti-epileptic drugs. Dev Med Child Neurol. 2006;48:394–8.

    Article  CAS  PubMed  Google Scholar 

  113. Hamano S-I, Mochizuki M, Monikawa T. Phenobarbital-induced atypical absence seizure in benign childhood epilepsy with centrotemporal spikes. Seizure. 2002;11:201–4.

    Article  PubMed  Google Scholar 

  114. Sazgar M, Bourgeois B. Aggravation of epilepsy by antiepileptic drugs. Pediatr Neurol. 2005;4:227–34.

    Article  Google Scholar 

  115. Stephen LJ, Forsyth M, Kelly K, Brodie MJ. Antiepileptic drug combinations: have newer agents altered clinical outcomes? Epilepsy Res. 2012;98:194–8.

    Article  CAS  PubMed  Google Scholar 

  116. Cacao G, Parra J, Mannan S, Sisodiya S, Sander J. Juvenile myoclonic epilepsy refractory to treatment in a tertiary referral centre. Epilepsy Behav. 2018;82:81–6.

    Article  PubMed  Google Scholar 

  117. Trinka E, Baumgartner S, Unterberger I, Unterrainer J, Luef G, Haberlandt E, et al. Long-term prognosis for childhood and juvenile absence epilepsy. J Neurol. 2004;251:1235–41.

    Article  PubMed  Google Scholar 

  118. Danhofer P, Brazdil M, Oslejskova H, Kuba R. Long-term seizure outcome in patients with juvenile absence epilepsy: a retrospective study in a tertiary referral center. Seizure. 2014;23:443–7.

    Article  PubMed  Google Scholar 

  119. Yacubian EM. Juvenile myoclonic epilepsy: challenges on its 60th anniversary. Seizure. 2017;44:48–52.

    Article  PubMed  Google Scholar 

  120. Burneo JG. SUDEP: let’s talk about it. Neurology. 2019;93:1–2.

    Article  Google Scholar 

  121. Covanis A. Photosensitivity in idiopathic generalized epilepsies. Epilepsia. 2005;46(Suppl. 9):67–72.

    Article  PubMed  Google Scholar 

  122. Asadi-Pooya AA, Hashemzehi Z, Emami M. Predictors of seizure control in patients with juvenile myoclonic epilepsy (JME). Seizure. 2014;23:889–91.

    Article  PubMed  Google Scholar 

  123. Gűrer R, Aydin S, Őzkara C. Outcomes of low dose valproic acid treatment in patients with juvenile myoclonic epilepsy. Seizure. 2019;70:43–8.

    Article  PubMed  Google Scholar 

  124. Giorgi FS, Guida M, Caciagli L, et al. Social cognition in juvenile myoclonic epilepsy. Epilepsy Res. 2016;128:61–7.

    Article  PubMed  Google Scholar 

  125. Somayajula S, Vooturi S, Jayalakshmi S. Psychiatric disorders among 165 patients with juvenile myoclonic epilepsy in India and association with clinical and sociodemographic variables. Epilepsy Behav. 2015;53:37–42.

    Article  PubMed  Google Scholar 

  126. Almane DN, Jones JE, McMillan T, et al. The timing, nature and range of neurobehavioral comorbidities in juvenile myoclonic epilepsy. Pediatr Neurol. 2019;101:47–52.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Kanner AM. Most antidepressant drugs are safe for patients with epilepsy at therapeutic doses: a review of the evidence. Epilepsy Behav. 2016;61:282–6.

    Article  PubMed  Google Scholar 

  128. FDA Drug Safety Communication. Valproate antiseizure products contraindicated for migraine prevention in pregnant women due to decreased IQ scores in exposed children. https://www.fda.gov/Drugs/DrugSafety/ucm350684.htm. Accessed 15 Dec 2019.

  129. European Medicines Agency. Valproate and related substances. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/referrals/Valproate_and_related_substances/human_referral_prac_000032.jsp&mid=WC0b01ac05805c516f. Accessed 15 Dec 2019.

  130. Hernández-Diaz S, Smith CR, Shen A, et al.; North American AED Pregnancy Registry; North American AED Pregnancy Registry. Comparative safety of antiepileptic drugs during pregnancy. Neurology. 2012;78:1692–9.

  131. Campbell E, Kennedy F, Russell A, et al. Malformation risks of antiepileptic drug monotherapies in pregnancy: updated results from the UK and Ireland Epilepsy and Pregnancy Registers. J Neurol Neurosurg Psychiatry. 2014;85:1029–34.

    Article  CAS  PubMed  Google Scholar 

  132. Baker GA, Bromley RL, Briggs M, et al. IQ at 6 years after in utero exposure to antiepileptic drugs: a controlled cohort study. Neurology. 2015;84:382–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Weston J, Bromley R, Jackson CF, et al. Monotherapy treatment of epilepsy in pregnancy: congenital malformation outcomes in the child. Cochrane Database Syst Rev. 2016;11:CD010224. https://doi.org/10.1002/14651858.cd010224.pub2.

  134. Veroniki AA, Rios P, Cogo E, et al. Comparative safety of antiepileptic drugs for neurological development in children exposed during pregnancy and breast feeding: a systematic review and network meta-analysis. BMJ Open. 2017;7:e017248. https://doi.org/10.1136/bmjopen-2017-017248.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Lacey AS, Pickrell WO, Thomas RH, Kerr MP, White CP, Rees MI. Educational attainment of children born to mothers with epilepsy. J Neurol Neurosurg Psychiatry. 2018;89:736–40.

    Article  PubMed  Google Scholar 

  136. Bromley RL, Weston J, Adab N. Treatment for epilepsy in pregnancy: neurodevelopmental outcomes in the child. Cochrane Database Syst Rev. 2014;10;CD010236.

  137. Christensen J, Gronborg TK, Sorensen MJ, et al. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013;309:1696–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Bromley RL, Baker GA. Fetal antiepileptic drug exposure and cognitive outcomes. Seizure. 2017;44:225–31.

    Article  PubMed  Google Scholar 

  139. Mostacci B, Bisulli F, Poluzzi E, et al. Emilia-Romagna study on pregnancy and exposure to antiepileptic drugs (ESPEA): a population-based study on prescription patterns, pregnancy outcomes and fetal health. J Neurol Neurosurg Psychiatry. 2018;89:983–8.

    Article  PubMed  Google Scholar 

  140. Bech LF, Polcwiartek C, Kragholm K, et al. In utero exposure to antiepileptic drugs is associated with learning disabilities among offspring. J Neurol Neurosurg Psychiatry. 2018;89:1324–31.

    Article  PubMed  Google Scholar 

  141. Christensen J, Pedersen L, Sun Y, et al. Association of prenatal exposure to valproate and other antiepileptic drugs with risk for attention-deficit/hyperactivity disorder in offspring. JAMA Network Open. 2019;2(1):e186606.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Angus-Leppan H, Liu R. Weighing the risks of valproate in women who could become pregnant. BMJ. 2018;361:53–5.

    Google Scholar 

  143. Tomson T, Battino D, Perucca E. Teratogenicity of antiepileptic drugs. Curr Opin Neurol. 2019;32:246–52.

    Article  CAS  PubMed  Google Scholar 

  144. Tomson T, Battino D, Bromley R, Kochen S, Meador K, Pennell P, et al. Management of epilepsy in pregnancy: a report from the International League Against Epilepsy Task Force on Women and Pregnancy. Epileptic Disord. 2019;21:1–21.

    Google Scholar 

  145. Rohracher A, Brigo F, Hofler J, et al. Perampanel for the treatment of primary generalized tonic-clonic seizures in idiopathic generalized epilepsy. Expert Opin Pharmacother. 2016;17:1403–11.

    Article  CAS  PubMed  Google Scholar 

  146. Stephen LJ, Harden C, Tomson T, Brodie MJ. Management of epilepsy in women. Lancet Neurol. 2019;18:481–91.

    Article  CAS  PubMed  Google Scholar 

  147. Bromley RL, Weston J, Marson AG. Maternal use of antiepileptic agents during pregnancy and major congenital malformations in children. JAMA. 2017;318:1700–1.

    Article  PubMed  Google Scholar 

  148. Voinescu PE, Park S, Chen IQ, et al. Antiepileptic drug clearances during pregnancy and clinical implications for women with epilepsy. Neurology. 2018;91:e1228–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Pennell PB, Peng L, Newport DJ, Ritchie JC, Koganti A, Holley DK, et al. Lamotrigine in pregnancy: clearance, therapeutic drug monitoring and seizure frequency. Neurology. 2008;70:2130–6.

    Article  CAS  PubMed  Google Scholar 

  150. Baykan B, Martinez-Juarez IE, Altindag EA, Camfield CS, Camfield PR. Lifetime prognosis of juvenile myoclonic epilepsy. Epilepsy Behav. 2013;28:S18–24.

    Article  PubMed  Google Scholar 

  151. Höfler J, Unterberger I, Dobesberger J, Kuchukhidze G, Walser G, Trinka E. Seizures outcome in 175 patients with juvenile myoclonic epilepsy: a long-term observational study. Epilepsy Res. 2014;108:1817–24.

    Article  PubMed  Google Scholar 

  152. Healy L, Moran M, Singhal S, et al. Relapse after treatment withdrawal of antiepileptic drugs for juvenile absence epilepsy and juvenile myoclonic epilepsy. Seizure. 2018;59:116–22.

    Article  PubMed  Google Scholar 

  153. Vorderwulbecke BJ, Kirschbaum A, Merkle H, Senf P, Holtkamp M. Discontinuing antiepileptic drugs in long-standing idiopathic generalised epilepsy. J Neurol. 2019;266(10):2554–9.

    Article  PubMed  Google Scholar 

  154. Holtkamp M, Kowski AB, Merkle H, Janz D. Long-term outcome in epilepsy with grand mal on awakening: forty years of follow-up. Ann Neurol. 2014;75:298–302.

    Article  PubMed  Google Scholar 

  155. Senf P, Schmitz B, Holtkamp M, Janz D. Prognosis of juvenile myoclonic epilepsy 45 years after onset: seizure outcome and predictors. Neurology. 2013;81:2128–33.

    Article  PubMed  Google Scholar 

  156. Keezer M, Sisodiya S, Sander J. Comorbidities of epilepsy: current concepts and future perspectives. Lancet Neurol. 2016;15:106–15.

    Article  PubMed  Google Scholar 

  157. O’Muircheartaigh J, Vollmar C, Barker GJ, et al. Focal structural changes and cognitive dysfunction in juvenile myoclonic epilepsy. Neurology. 2011;76:34–40.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Yogarajah M, Mula M. Social cognition, psychiatric comorbidities and quality of life in adults with epilepsy. Epilepsy Behav. 2019;100:106321.

    Article  PubMed  Google Scholar 

  159. Rauchenzauner M, Hagn C, Walch R, Baumann M, Haberlandt E, Frühwirth M, Rostasy K. Quality of life and fitness in children and adolescents with epilepsy (EpiFit). Neuropediatrics. 2017;48:161–5.

    Article  PubMed  Google Scholar 

  160. Gupta S, Kwan P, Faught E, Tsong W, Forsythe A, Ryvlin P. Understanding the burden of idiopathic generalised epilepsy in the United States, Europe and Brazil: an analysis for the National Health and Wellness Survey. Epilepsy Behav. 2016;55:146–56.

    Article  PubMed  Google Scholar 

  161. Panayiotopolous CP. Epilepsy with GTCS only. A clinical guide to tpileptic syndromes and their treatment. 2nd ed. London: Springer; 2007. p. 343–5.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Linda J. Stephen.

Ethics declarations

Funding

No sources of funding were received for the preparation of this article.

Conflict of interest

Linda J. Stephen has received personal fees from Eisai, outside of the submitted work. Martin J. Brodie has received personal speaker and/or consultancy fees from UCB, Eisai, Xenon and Arvelle Therapeutics outside of the submitted work.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stephen, L.J., Brodie, M.J. Pharmacological Management of the Genetic Generalised Epilepsies in Adolescents and Adults. CNS Drugs 34, 147–161 (2020). https://doi.org/10.1007/s40263-020-00698-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-020-00698-5

Navigation