487
Views
17
CrossRef citations to date
0
Altmetric
Review

Pharmacokinetic considerations for anti-epileptic drugs in children

, , , , , , ORCID Icon & show all
Pages 199-211 | Received 10 Nov 2018, Accepted 24 Jan 2019, Published online: 07 Feb 2019

References

  • Guerrini R. Epilepsy in children. Lancet. 2006;367(9509):499–524.
  • Italiano D, Perrucca E. Clinical pharmacokinetics of new-generation antiepileptic drugs at the extremes of age: an update. Clin Pharmacokinet. 2013;52(8):627–645.
  • Tolou-Ghamari Z, Zare M, Habibabadi JM, et al. A quick review of carbamazepine pharmacokinetics in epilepsy from 1953 to 2012. J Res Med Sci. 2013;18(Suppl 1):S81–5.
  • Milovanovic JR, Jankovic SM. Factors influencing carbamazepine pharmacokinetics in children and adults: population pharmacokinetic analysis. Int J Clin Pharmacol Ther. 2011;49(7):428–436.
  • Brodie MJ, Mintzer S, Pack AM, et al. Enzyme induction with antiepileptic drugs: cause for concern? Epilepsia. 2013;54:11–27.
  • Djordjevic N, Jankovic SM, Milovanovic JR. Pharmacokinetics and pharmacogenetics of carbamazepine in children. Eur J Drug Metab Pharmacokinet. 2017;42(5):729–744.
  • Perucca E. Clinically relevant drug interactions with antiepileptic drugs. Br J Clin Pharmacol. 2006;61:246–255.
  • Riva R, Contin M, Albani F, et al. Free and total serum concentrations of carbamazepine and carbamazepine-10,11-epoxide in infancy and childhood. Epilepsia. 1985;26(4):320–322.
  • Brigo F, Leo A, Russo E, et al. Intravenous carbamazepine for the treatment of epilepsy. Expert Opin Pharmacother. 2018;19(7):743–747.
  • Tulloch JK, Carr RR, Ensom MH. A systematic review of the pharmacokinetics of antiepileptic drugs in neonates with refractory seizures. J Pediatr Pharmacol Ther. 2012;17(1):31–44.
  • Reimers A, Berg JA, Burns ML, et al. Reference ranges for antiepileptic drugs revisited: a practical approach to establish national guidelines. Drug Des Devel Ther. 2018;12:271–280.
  • Gören MZ, Onat F. Ethosuximide: from bench to bedside. CNS Drug Rev. 2007;13(2):224–239.
  • Brigo F, Igwe SC. Ethosuximide, sodium valproate or lamotrigine for absence seizures in children and adolescents. Cochrane Database Syst Rev. 2017;2:CD003032.
  • Battino D, Estienne M, Avanzini G. Clinical pharmacokinetics of antiepileptic drugs in paediatric patients. Part I: phenobarbital, primidone, valproic acid, ethosuximide and mesuximide. Clin Pharmacokinet. 1995;29(4):257–286.
  • Newton CR, Garcia HH. Epilepsy in poor regions of the world. Lancet. 2012;380(9848):1193–1201.
  • Czapiński P, Blaszczyk B, Czuczwar SJ. Mechanisms of action of antiepileptic drugs. Curr Top Med Chem. 2005;5(1):3–14.
  • Moffett BS, Weingarten MM, Galati M, et al. Phenobarbital population pharmacokinetics across the pediatric age spectrum. Epilepsia. 2018;59(7):1327–1333.
  • Anderson GD. Developmental pharmacokinetics. Semin Pediatr Neurol. 2010;17(4):208–213.
  • Koukouritaki SB, Manro JR, Marsh SA, et al. Developmental expression of human hepatic CYP2C9 and CYP2C19. J Pharm Exp Ther. 2004;308:965–974.
  • Pacifici GM. Clinical pharmacology of phenobarbital in neonates: effects, metabolism and pharmacokinetics. Curr Pediatr Rev. 2016;12(1):48–54.
  • Yukawa M, Yukawa E, Suematsu F, et al. Population pharmacokinetics of phenobarbital by mixed effect modelling using routine clinical pharmacokinetic data in Japanese neonates and infants: an update. J Clin Pharm Ther. 2011;36(6):704–710.
  • Grasela TH Jr, Donn SM. Neonatal population pharmacokinetics of phenobarbital derived from routine clinical data. Dev Pharmacol Ther. 1985;8(6):374–383.
  • Filippi L, la Marca G, Cavallaro G, et al. Phenobarbital for neonatal seizures in hypoxic ischemic encephalopathy: a pharmacokinetic study during whole body hypothermia. Epilepsia. 2011;52(4):794–801.
  • Shellhaas RA, Ng CM, Dillon CH, et al. Population pharmacokinetics of phenobarbital in infants with neonatal encephalopathy treated with therapeutic hypothermia. Pediatr Crit Care Med. 2013;14(2):194–202.
  • Shorvon S, Perucca E, Engel J. Phenytoin. In: The treatment of epilepsy. 3rd ed. Wiley-Blackwell; 2015:605–618.
  • Koren G, Brand N, Halkin H, et al. Kinetics of intravenous phenytoin in children. Pediatr Pharmacol. 1984;4(1):31–38.
  • Loscher W. Drug transporters in the epileptic brain. Epilepsia. 2007;48(suppl.1):8–13.
  • Franco V, Perucca E. CYP2C9 polymorphisms and phenytoin metabolism: implications for adverse effects. Expert Opin Drug Metab Toxicol. 2015;11(8):1269–1279.
  • Bauer LA, Blouin RA. Phenytoin Michaelis-Menten pharmacokinetics in Caucasian paediatric patients. ClinPharmacokinet. 1983;8:545–549.
  • Johnson TN, Rostami-Hodjegan A, Tucker GT. Prediction of the clearance of eleven drugs and associated variability in neonates, infants and children. Clin Pharmacokinet. 2006;45(9):931–956.
  • Leniger T, Wiemann M, Bingmann D, et al. Carbonic anhydrase inhibitor sulthiame reduces intracellular pH and epileptiform activity of hippocampal CA3 neurons. Epilepsia. 2002;43(5):469–474.
  • Tacke M, Borggraefe I, Gerstl L, et al. Effects of Levetiracetam and Sulthiame on EEG in benign epilepsy with centrotemporal spikes: A randomized controlled trial. Seizure. 2018;56:115–120.
  • Fejerman N, Caraballo R, Cersósimo R, et al. Sulthiame add-on therapy in children with focal epilepsies associated with encephalopathy related to electrical status epilepticus during slow sleep (ESES). Epilepsia. 2012;53(7):1156–1161.
  • Gorman KM, Shahwan A. Sulthiame revisited: treatment of refractory absence seizures. Epileptic Disord. 2016;18(3):329–333.
  • May TW, Korn-Merker E, Rambeck B, et al. Pharmacokinetics of sulthiame in epileptic patients. Ther Drug Monit. 1994;16(3):251–257.
  • Milburn-McNulty P, Powell G, Sills GJ, et al. Sulthiame monotherapy for epilepsy. Cochrane Database Syst Rev. 2014;3:CD010062.
  • Yamamoto Y, Takahashi Y, Imai K, et al. Interaction between sulthiame and clobazam: sulthiame inhibits the metabolism of clobazam, possibly via an action on CYP2C19. Epilepsy Behav. 2014;34:124–126.
  • 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(2):210–218.
  • Perucca E. Pharmacological and therapeutic properties of valproate. A summary after 35 years of clinical experience. CNS Drugs. 2002;16:695–714.
  • Ding J, Wang Y, Lin W, et al. A population pharmacokinetic model of valproic acid in pediatric patients with epilepsy: a non-linear pharmacokinetic model based on protein-binding saturation. Clin Pharmacokinet. 2015;54:305–317.
  • Ghodke-Puranik Y, Thorn CF, Lamba JK, et al. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics. 2013;23:236–241.
  • Xu S, Chen Y, Zhao M, et al. Population pharmacokinetics of valproic acid in epileptic children: effects of clinical and genetic factors. Eur J Pharm Sci. 2018;15(122):170–178.
  • Harty TP, Rogawski MA. Felbamate block of recombinant Nmethyl-D-aspartate receptors: selectivity for the NR2B subunit. Epilepsy Res. 2000;39:47–55.
  • Grosso S, Cordelli DM, Coppola G, et al. Efficacy and safety of felbamate in children under 4 years of age: a retrospective chart review. Eur J Neurol. 2008;15:940–946.
  • Pellock JM, Faught E, Leppik IE, et al. Felbamate: consensus of current clinical experience. Epilepsy Res. 2006;71:89–101.
  • DeSarro G, Ongini E, Bertorelli R, et al. Excitatory amino acid neurotransmission through both NMDA and non-NMDA receptors is involved in the anticonvulsant activity of felbamate in DBA/2 mice. Eur J Pharmacol. 1994;262:11–19.
  • Rho JM, Donevan SD, Rogawski MA. Mechanism of action of the anticonvulsant felbamate: opposing effects on N-methyl-D-aspartate and a-amino-butyric acid A receptors. Ann Neurol. 1994;35:229–236.
  • Felbamate Study Group in Lennox-Gastaut Syndrome. Efficacy of felbamate in childhood epileptic encephalopathy (Lennox–Gastaut syndrome). N Engl J Med. 1993;328:29–33.
  • Winkler SR, Luer MS. Antiepileptic drug review: part 2. Surg Neurol. 1998;49:566–568.
  • Thakkar K, Billa G, Rane J, et al. The rise and fall of felbamate as a treatment for partial epilepsy–aplastic anemia and hepatic failure to blame? Expert Rev Neurother. 2015;15:1373–1375.
  • Ward DL, Shumaker RC. Comparative bioavailability of felbamate in healthy men. Epilepsia. 1990;31:642.
  • Iapadre G, Balagura G, Zagaroli L, et al. Pharmacokinetics and drug interaction of antiepileptic drugs in children and adolescents. Paediatr Drugs. 2018.
  • Perucca E. Clinical pharmacokinetics of new-generation antiepileptic drugs at the extremes of age. Clin Pharmacokinet. 2006;45:351–363.
  • Shumaker RC, Fantel C, Kelton E, et al. Evaluation of the elimination of (14C) felbamate in healthy men. Epilepsia. 1990;31:642.
  • Macdonald RL, Greenfield LJ. Mechanisms of action of new antiepileptic drugs. Curr Opin Neurol. 1997;10:121–128.
  • Dibué-Adjei M, Kamp MA, Alpdogan S. Cav2.3 (R-Type) calcium channels are critical for mediating anticonvulsive and neuroprotective properties of lamotrigine in vivo. Cell Physiol Biochem. 2017;44:935–947.
  • La Roche SM, Helmers SL. The new antiepileptic drugs. Jama. 2004;291:605–614.
  • 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;54:551–563.
  • Hancock EC, Cross JH. Treatment of Lennox-Gastaut syndrome. Cochrane Database Syst Rev. 2013;28:CD003277.
  • Bartoli A, Guerrini R, Belmonte A, et al. The influence of dosage, age, and comedication on steady state plasma lamotrigine concentrations in epileptic children: a prospective study with preliminary assessment of correlations with clinical response. Ther Drug Monit. 1997;19:252–260.
  • Morris RG, Black AB, Harris AL, et al. Lamotrigine and therapeutic drug monitoring: retrospective survey following the introduction of a routine service. Br J Clin Pharmacol. 1998;46:547–551.
  • Patsalos PN, Berry DJ, Bourgeois BF, et al. Antiepileptic drugs–best practice guidelines for therapeutic drug monitoring: a position paper by the subcommission on therapeutic drug monitoring, ILAE Commission on Therapeutic Strategies. Epilepsia. 2008;49:1239–1276.
  • Rambeck B, Wolf P. Lamotrigine clinical pharmacokinetics. Clin Pharmacokinet. 1993;25:433–443.
  • Biton V. Pharmacokinetics, toxicology and safety of lamotrigine in epilepsy. Expert Opin Drug Metab Toxicol. 2006;2:1009–1018.
  • Chen C, Casale EJ, Duncan B, et al. Pharmacokinetics of lamotrigine in children in the absence of other antiepileptic drugs. Pharmacotherapy. 1999;19:437–441.
  • Hussein Z, Posner J. Population pharmacokinetics of lamotrigine monotherapy in patients with epilepsy: retrospective analysis of routine monitoring data. Br J Clin Pharmacol. 1997;43:457–464.
  • Deshpande LS, Delorenzo RJ. Mechanism of levetiracetam in the control of status epilepticus and epilepsy. Front Neurol. 2014;5:11.
  • Lukyanetz EA, Shkryl VM, Kostyuk PG. Selective blockade of N-type calcium channels by levetiracetam. Epilepsia. 2002;43:9–18.
  • Weijenberg A, Brouwer OF, Callenbach PM. Levetiracetam monotherapy in children with epilepsy: a systematic review. CNS Drugs. 2015;29:371–382.
  • Verrotti A, Prezioso G, Di Sabatino F. The adverse event profile of levetiracetam: a meta-analysis on children and adults. Seizure. 2015;31:49–55.
  • Patsalos PN. Pharmacokinetic profile of levetiracetam: toward ideal characteristics. Pharmacol Ther. 2000;85:77–85.
  • Allegaert K, Lewi L, Naulaers G, et al. Levetiracetam pharmacokinetics in neonates at birth. Epilepsia. 2006;47:1068–1069.
  • Glauser TA, Mitchell WG, Weinstock A, et al. Pharmacokinetics of levetiracetam in infants and young children with epilepsy. Epilepsia. 2007;48:1117–1122.
  • Walker MC, Patsalos PN. Clinical pharmacokinetics of new antiepileptic drugs. Pharmacol Ther. 1995;67:351–384.
  • Patsalos PN. Clinical pharmacokinetics of levetiracetam. Clin Pharmacokinet. 2004;43:707–724.
  • Pennell P B. 2005 AES annual course: evidence used to treat women with epilepsy. Epilepsia. 2006;47 Suppl 1:46–53.
  • McLean MJ, Schmutz M, Wamil AW, et al. Oxcarbazepine: mechanisms of action. Epilepsia. 1994;35:5–9.
  • Schmidt D, Elger CE. What is the evidence that oxcarbazepine and carbamazepine are distinctly different antiepileptic drugs? Epilepsy Behav. 2004;5:627–635.
  • Ambrosio AF, Silva AP, Malva JO, et al. Carbamazepine inhibits L-type Ca2+ channels in cultured rat hippocampus neurons stimulated with glutamate receptor antagonists. Neuropharmacology. 1999;38:1349–1359.
  • Stefani A, Pisani A, De Murtas M, et al. Action of GP47779, the active metabolite of oxcarbazepine, on corticostriatal system. II. Modulation of high-voltage-activated calcium currents. Epilepsia. 1995;336:997–1002.
  • May TW, Korn-Merker E, Rambeck B. Clinical pharmacokinetics of oxcarbazepine. Clin Pharmacokinet. 2003;42:1023–1042.
  • Flesch G, Tudor D, Souppart C, et al. Oxcarbazepine final market image tablet formulation bioequivalence study after single administration and at steady state in healthy subjects. Int J Clin Pharmacol Ther. 2002;40:524–532.
  • Schultz H, Feldman K, Faigle JW, et al. The metabolism of 14C-oxcarbazepine in man. Xenobiotica. 1986;16:769–778.
  • Lloyd P, Flesch G, Dieterle W. Clinical pharmacology and pharmacokinetics of oxcarbazepine. Epilepsia. 1994;35:10–13.
  • Tartara A, Galimberti CA, Manni R, et al. The pharmacoinkinetics of oxcarbazepine and its active metabolite 10-hydroxy-carbazepine in healthy subjects and in epileptic patients treated with phenobarbitone or valproic acid. Br J Clin Pharmacol. 1993;36:366–368.
  • van Heiningen PN, Eve MD, Oosterhuis B, et al. The influence of age on the pharmacokinetics of the antiepileptic agent oxcarbazepine. Clin Pharmacol Ther. 1991;50:410–419.
  • Grant SM, Faulds D. Oxcarbazepine: a review of its pharmacology and therapeutic potential in epilepsy, trigeminal neuralgia and affective disorders. Drugs. 1992;43:873–888.
  • May TW, Boor R, Mayer T, et al. Concentrations of stiripentol in children and adults with epilepsy: the influence of dose, age, and comedication. Ther Drug Monit. 2012;34(4):390–397.
  • Nabbout R, Chiron C. Stiripentol: an example of antiepileptic drug development in childhood epilepsies. Eur J Paediatr Neurol. 2012;16(Suppl 1):S13–7.
  • Nickels KC, Wirrell EC. Stiripentol in the management of epilepsy. CNS Drugs. 2017;31(5):405–416.
  • Peigne S, Rey E, Le Guern ME, et al. Reassessment of stiripentol pharmacokinetics in healthy adult volunteers. Epilepsy Res. 2014;108:909–916.
  • Peigné S, Chhun S, Tod M, et al. Population pharmacokinetics of stiripentol in paediatric patients with Dravet syndrome treated with stiripentol, valproate and clobazam combination therapy. Clin Pharmacokinet. 2018;57(6):739–748.
  • Bialer M, Johanessen SI, Kupferberg HJ, et al. Progress report on new antiepileptic drugs: a summary of the Sixth Eilat Conference (EILAT VI). Epilepsy Res. 2002;51:31–71.
  • Abtahi MA, Abtahi SH, Fazel F, et al. Topiramate and the vision: a systematic review. Clin Ophthalmol. 2012;6:117–131.
  • Easterling DE, Zakszewski T, Moyer MD, et al. Plasma pharmacokinetics of topiramate, a new anticonvulsant in humans. Epilepsia. 1988;29:662.
  • Doose DR, Walker SA, Gisclon LG, et al. Single-dose pharmacokinetics and affect of food on the bioavailability of topiramate, a novel antiepileptic drug. J Clin Pharmacol. 1996;36:884–891.
  • Doose DR, Streeter AJ. Topiramate: chemistry, biotransformation, and pharmacokinetics. In: Levy RH, Mattson RH, Meldrum BS, et al., editors. Antiepileptic drugs, 5th ed. Philadelphia: Lippincott Williams & Wilkins; 2002:727–734.
  • Sachdeo RC, Sachdeo SK, Walker SA, et al. Steady-state pharmacokinetics of topiramate and carbamazepine in patients with epilepsy during monotherapy and concomitant therapy. Epilepsia. 1996;37:774–780.
  • Britzi M, Perucca E, Soback S, et al. Pharmacokinetic and metabolic investigation of topiramate disposition in healthy subjects in the absence and in the presence of enzyme induction by carbamazepine. Epilepsia. 2005;46:378–384.
  • Johannessen Landmark C, Baftiu A, Tysse I, et al. Pharmacokinetic variability of four newer antiepileptic drugs, lamotrigine, levetiracetam, oxcarbazepine, and topiramate: a comparison of the impact of age and comedication. Ther Drug Monit. 2012;34:440–445.
  • Perucca E. The clinical pharmacokinetics of the new antiepileptic drugs. Epilepsia. 1999;40:7–13.
  • Garnett WR. Clinical pharmacology of topiramate: a review. Epilepsia. 2000;41:61–65.
  • Van Der Poest Clement EA, Sahin M, Peters JM. Vigabatrin for epileptic spasms and tonic seizures in Tuberous Sclerosis Complex. J Child Neurol. 2018 Jul;8:519–524.
  • Zhang B, Mc Daniel SS, Rensing NR, et al. Vigabatrin inhibits seizures and mTOR pathway activation in a mouse model of tuberous sclerosis complex. PLoS One. 2013;8(2):e57445.
  • Frisk-Holmberg M, Kerth P, Meyer P. Effect of food on the absorption of Vigabatrin. Br J Pharmacol. 1989;27(Suppl 1):23S–5S.
  • Battino D, Estienne M, Avanzini G. Clinical pharmacokinetics of antiepileptis drugs in paediatric patients. Part II: phenytoin, carbamazepine, sulthiame, lamotrigine, vigabatrin, oxcarbazepine and felbamate. Clin Pharmacokinet. 1995;29(5):341–369.
  • Nielsen JC, Kowalski KG, Karim A, et al. Population pharmacokinetics analysis of Vigabatrin in adults and children with epilepsy and children with infantile spasms. Clin Pharmacokinet. 2014;53:1019–1031.
  • Chang SY, Lin W-C. Determination of Vigabatrin by capillary electrophoresis with laser-induced fluorescence detection. J Chromatogr B Analyt Technol Biomed Life Sci. 2003;794(1):17–22.
  • Jacob S, Nair AB. An updated overview on therapeutic drug monitoring of recent antiepileptic drugs. Drugs R D. 2016;16(4):303–316.
  • Coppola G, Grosso S, Verrotti A, et al. Zonisamide in children and young adults with refractory epilepsy: an open label, multicenter Italian study. Epilepsy Res. 2009;83:112–116.
  • Biton V. Clinical pharmacology and mechanism of action of zonisamide. Clin Neuropharmacol. 2007;30(4):230–240.
  • Hoy SM. Zonisamide: a review of its use as adjunctive therapy in the management of partial seizures in pediatric patients aged ≥ 6 years. Pediatr Drugs. 2014;16:235–246.
  • Johannesen Landmark C, Patsalos PN. Drug interactions involving the new second- and third-generation antiepileptic drugs. Expert Rev Neurother. 2010;10(1):119–140.
  • Cross JH, Auvin S, Patten A, et al. Safety and tolerability of Zonisamide in paediatric patients with epilepsy. Off J Eur Paediatric Neurol Soc. 2014;(6):747–758.
  • Patsalos PN. Drug interactions with the newer antiepileptic drugs (AEDs) – part 1: pharmacokinetics and pharmacodynamic interactions between AEDs. Clin Pharmacokinet. 2013;52:927–966.
  • Landmark CJ, Johannessen SI, Tomson T. Dosing strategies for antiepileptic drugs: from a standard dose for all to individualised treatment by implementation of therapeutic drug monitoring. Epileptic Disord. 2016;18(4):367–383.
  • De Biase S, Valente M, Gigli GL, et al. Pharmacokinetic drug evaluation of Lacosamide for the treatment of partial-onset seizures. Expert Opin Drug Metab Toxicol. 2017;13(9):997–1005.
  • Rogawski MA, Tofighy A, White HS, et al. Current understanding of the mechanism of action of the antiepileptic drug lacosamide. Epilepsy Res. 2015;110:189–205.
  • Verrotti A, Loiacono G, Pizzolorusso A, et al. Lacosamide in pediatric and adult patients: comparison of efficacy and safety. Seizure. 2013;22:210–216.
  • Cawello W. Clinical pharmacokinetic and pharmacodynamic profile of lacosamide. Clin Pharmacokinet. 2015;54(9):901–914.
  • Cawello W, Mueller-Voessing C, Fichtner A. Pharmacokinetics of lacosamide and omeprazole coadministration in healthy volunteers: results from a phase 1, randomized, crossover trial. Clin Drug Investig. 2014;24(5):317–325.
  • Contin M, Albani F, Riva R, et al. Lacosamide therapeutic monitoring in patients with epilepsy: effect of concomitant antiepileptic drugs. Ther Drug Monit. 2013;35(6):849–852.
  • Rudzinski LA, Vélez-Ruiz NJ, Gedzelman ER, et al. New antiepileptic drugs: focus on ezogabine, clobazam, and perampanel. J Investig Med. 2016;64:1087–1101.
  • De Liso P, Moavero R, Coppola G, et al. Current role of perampanel in pediatric epilepsy. Ital J Pediatr. 2017;43(1):51.
  • Birò A, Stephani U, Tarallo T, et al. Effectiveness and tolerability of perampanel in children and adolescents with refract-ory epilepsies: first experiences. Neuropediatrics. 2015;46(2):110–116.
  • Schulze-Bonhage A. Perampanel for epilepsy with partial-onset seizures: a pharmacokinetic and pharmacodynamic evaluation. Expert Opin Drug Metab Toxicol. 2015;11(8):1329–1337.
  • Mudigoudar B, Weatherspoon S, Wheless JW. Emerging antiepileptic drugs for severe pediatric epilepsies. Semin Pediatr Neurol. 2016;23(2):167–179.
  • Patsalos PN, Spencer EP, Berry DJ. Therapeutic drug monitoring of antiepileptic drugs in epilepsy: a 2018 update. Ther Drug Monit. 2018;40(5):526–548.
  • Perucca E, Cloyd J, Critchley D, et al. Rufinamide: clinical pharmacokinetics and concentration-response relationships in patients with epilepsy. Epilepsia. 2008;49:1123–1141.
  • Mueller A, Boor R, Coppola G, et al. Low long-term efficacy and tolerability of add-on rufinamide in patients with Dravet syndrome. Epilepsy Behav. 2011;21:282–284.
  • Coppola G, Besaq F, Cusmai R, et al. Current role of rufinamide in the treatment of childhood epilepsy: literature review and treatment guidelines. Eur J Paediatr Neurol. 2014;18(6):685–690.
  • May TW, Boor R, Rambeck B, et al. Serum concentrations of rufinamide in children and adults with epilepsy: the influence of dose, age, and comedication. Ther Drug Monit. 2011;33:214–221.
  • La Marca G, Rosati A, Falchi M, et al. A pharmacokinetic study and correlation with clinical response of rufinamide in infants with epileptic encephalopathies. Pharmacology. 2013;91:275e80.
  • Yozawitz E, Stacey A, Pressler RM. Pharmacotherapy for seizures in neonates with hypoxic ischemic encephalopathy. Paediatr Drugs. 2017;19(6):553–567.
  • Orlandi A, Paolino MC, Striano P, et al. Clinical reappraisal of the influence of drug-transporter polymorphisms in epilepsy. Expert Opin Drug Metab Toxicol. 2018;14(5):505–512.
  • Loiacono G, Masci M, Zaccara G, et al. The treatment of neonatal seizures: focus on Levetiracetam. J Matern Fetal Neonatal Med. 2016;29(1):69–74.
  • Agrawal A, Banergee A. A review on pharmacokinetics of levetiracetam in neonates. Curr Drug Metab. 2017;18(8):727–734.
  • Pichini S, Papaseit E, Joya X. Pharmacokinetics and therapeutic drug monitoring of psychotropic drugs in pediatrics. Ther Drug Monit. 2009 Jun;31(3):283–318.
  • Brandt C, May TW. Extended-release drug formulations for the treatment of epilepsy. Expert Opin Pharmacother. 2018;19(8):843–850.
  • Legros B, Boon P, Ceulemans B, et al. Development of an electronic decision tool to support appropriate treatment choice in adult patients with epilepsy–Epi-Scope(®). Seizure. 2012;21(1):32–39.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.