1,632
Views
70
CrossRef citations to date
0
Altmetric
Review

Therapeutic drug monitoring of antiepileptic drugs: current status and future prospects

, &
Pages 227-238 | Received 13 Nov 2019, Accepted 29 Jan 2020, Published online: 13 Feb 2020

References

  • Patsalos PN, St Louis EK. The epilepsy prescriber’s guide to antiepileptic drugs. 3rd ed. Cambridge: Cambridge University Press; 2018.
  • 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.
  • Johannessen Landmark C, Johannessen SI, Tomson T. Host factors affecting antiepileptic drug delivery – pharmacokinetic variability. Adv Drug Deliv Rev. 2012a;64:896–910.
  • Johannessen Landmark C, Johannessen SI, Tomson T. Dosing strategies of antiepileptic drugs – from one dose to individualisation of treatment by implementation of therapeutic drug monitoring. Epileptic Disord. 2016;18(4):367–383.
  • Johannessen SI, Battino D, Berry DJ, et al. Therapeutic drug monitoring of the newer antiepileptic drugs. Ther Drug Monit. 2003;25:347–363.
  • 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.
  • Patsalos PN, Zugman M, Lake C, et al. Serum protein binding of 25 antiepileptic drugs in a routine clinical setting: a comparison of free non-protein-bound concentrations. Epilepsia. 2017;58(7):1234–1243.
  • Patsalos PN, Berry DJ. Therapeutic drug monitoring of antiepileptic drugs by use of saliva. Ther Drug Monit. 2013;35:4–29.
  • Baftiu A, Feet SA, Larsson PG, et al. Utilization of antiepileptic drugs in the elderly vs younger patients with epilepsy and psychiatric comorbidity. Epilepsy Res. 2018;139:35–42.
  • Chen Z, Brodie MJ, Liew D, et al. Treatment outcomes with newly diagnosed epilepsy treated with established and new antiepileptic drugs: a 30-year longitudinal cohort study. JAMA Neurol. 2018;75(3):279–286.
  • Kwan P, Brodie MJ. Early identification of refractory epilepsy. N Engl J Med. 2000;342:314–319.
  • 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(3):551–563.
  • Nunes VD, Sawyer L, Neilson J, et al. Diagnosis and management of the epilepsies in adults and children: summary of updated NICE guidance. BMJ. 2012;26:e344:281.
  • Hadjiloizou SM, Bourgeois BF. Antiepileptic drug treatment in children. Expert Rev Neurother. 2007;7:179–193.
  • Italiano D, Perucca E. Clinical pharmacokinetics of new-generation antiepileptic drugs at the extremes of age: an update. Clin Pharmacokinet. 2013;8:627–645.
  • Verotti A, Iapadre G, Di Donato G, et al. Pharmacokinetic considerations for anti-epileptic drugs in children. Exp Opin Drug Metab Toxicol. 2019;15(3):199–211.
  • Tomson T, Landmark CJ, Battino D. Antiepileptic drug treatment in pregnancy: changes in drug disposition and their clinical implications. Epilepsia. 2013;54:405–414.
  • Patel SI, Pennell PB. Management of epilepsy during pregnancy: an update. Ther Adv Neurol Disord. 2016;9(2):118–129.
  • Tomson T, Battino D. Pharmacokinetics and therapeutic drug monitoring of newer antiepileptic drugs during pregnancy and the puerperium. Clin Pharmacokinet. 2007a;46:209–219.
  • Petrenaite V, Sabers A, Hansen-Schwartz J. Individual changes in lamotrigine plasma concentrations during pregnancy. Epilepsy Res. 2005;65:185–188.
  • Pennell PB, Peng L, Newport DJ, et al. Lamotrigine in pregnancy. Clearance, therapeutic drug monitoring, and seizure frequency. Neurology. 2008;70:2130–2136.
  • Tomson T, Palm R, Källén K, et al. Pharmacokinetics of levetiracetam during pregnancy, delivery, in the neonatal period, and lactation. Epilepsia. 2007b;48:1111–1116.
  • Mazzucchelli I, Onat FY, Özkara C, et al. Changes in the disposition of oxcarbazepine and its metabolites during pregnancy and the puerperium. Epilepsia. 2006;47(3):504–509.
  • Öhman I, Vitols S, Luef G, et al. Topiramate kinetics during delivery, in the neonatal period, and lactation: preliminary observations. Epilepsia. 2002;43:1157–1160.
  • Westin AA, Nakken KO, Johannessen SI, et al. Serum concentration/dose ratio of topiramate during pregnancy. Epilepsia. 2009;50:480–485.
  • Reimers A, Helde G, Becser Andersen N, et al. Zonisamide serum concentrations during pregnancy. Epilepsy Res. 2018a;144:25–29.
  • Öhman I, Vitols S, Tomson T. Pharmacokinetics of gabapentin during delivery, in the neonatal period, and lactation: does a fetal accumulation occur during pregnancy? Epilepsia. 2005;46:1621–1624.
  • Gerard EE, Meador KJ. An update on maternal use of antiepileptic medications in pregnancy and neurodevelopment outcomes. J Pediatr Genet. 2015;4(2):94–110.
  • EMA. Visited March 28th, 2019. https://www.ema.europa.eu/en/medicines/human/referrals/valproate-related-substances.
  • Tomson T, Battino D, Bonizzoni E, et al. Comparative risk of major congenital malformations with eight different antiepileptic drugs: a prospective cohort study of the EURAP registry. Lancet Neurol. 2018;17(6):530–538.
  • Johannessen Landmark C, Burns ML, Baftiu A, et al. Pharmacokinetic variability of valproate in women of childbearing. Epilepsia. 2017;58(10):e142–e146.
  • Johannessen Landmark C, Farmen AH, Burns ML, et al. Pharmacokinetic variability of valproate during pregnancy – implications for the use of therapeutic drug monitoring. Epilepsy Res. 2018;141:31–37.
  • Johannessen SI, Johannessen Landmark C. Antiepileptic drug interactions – basic principles and clinical implications. Curr Neuropharm. 2010;8:254–267.
  • Johannessen Landmark C, Patsalos PN. Drug interactions involving the new second- and third-generation antiepileptic drugs. Expert Rev Neurother. 2010;10:119–140.
  • Patsalos PN. Drug interactions with the newer antiepileptic drugs (AEDs)-part 1: pharmacokinetic and pharmacodynamic interactions between AEDs. Clin Pharmacokinet. 2013a;52:927–966.
  • Patsalos PN. Drug interactions with the newer antiepileptic drugs (AEDs)-part 2: pharmacokinetic and pharmacodynamic interactions between AEDs and drugs used to treat non-epilepsy disorders. Clin Pharmacokinet. 2013b;52:1045–1061.
  • Patsalos PN. Antiepileptic drug interactions: a clinical guide. 3rd ed. Switzerland: Springer; 2016.
  • Hole K, Wollmann BM, Nguyen C, et al. Comparison of CYP3A4-inducing capacity of enzyme-inducing antiepileptic drugs using 4β-hydroxycholesterol as biomarker. Ther Drug Monit. 2018;40(4):463–468.
  • Bertilsson L, Höjer B, Tybring G, et al. Autoinduction of carbamazepine metabolism in children examined by a stable isotope technique. Clin Pharmacol Ther. 1980;27:83–88.
  • Kudriakova TB, Sirota LA, Rozova GI, et al. Autoinduction and steady-state pharmacokinetics of carbamazepine and its major metabolites. Br J Clin Pharmacol. 1992;33:611–615.
  • 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. 2012b;34(4):440–445.
  • Koristkova B, Grundmann M, Brozmanova H, et al. Lamotrigine drug interactions in combination therapy and the influence of therapeutic drug monitoring on clinical outcomes in paediatric patients. Basic Clin Pharmacol Toxicol. 2019;125(1):26–33.
  • De Leon J, Spina E, Diaz FJ. Clobazam therapeutic drug monitoring: a comprehensive review of the literature with proposals to improve future studies. Ther Drug Monit. 2013;35(1):30–47.
  • Burns ML, Baftiu A, Opdal MS, et al. Therapeutic drug monitoring of clobazam and its metabolite-Impact of age and comedication on pharmacokinetic variability. Ther Drug Monit. 2016;38(3):350–357.
  • Karouni M, Henning O, Larsson PG, et al. Psychiatric comorbidity in patients with refractory epilepsy. Epi Behav. 2013;29:77–81.
  • Spina E, Pisani F, de Leon J. Clinically significant pharmacokinetic drug interactions of antiepileptic drugs with new antidepressants and new antipsychotics. Pharmacol Res. 2016;106:72–86.
  • Asconapé JJ. Pharmacokinetic considerations with the use of antiepileptic drugs in patients with HIV and organ transplants. Curr Neurol Neurosci Rep. 2018;18:89.
  • Sabers A, Öhman I, Christensen J, et al. Oral contraceptives reduce lamotrigine plasma levels. Neurology. 2003;61:570–571.
  • Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not progestogens, reduces lamotrigine serum concentrations. Epilepsia. 2005;46:1414–1417.
  • Galgani A, Palleria C, Iannone LF, et al. Pharmacokinetic interactions of clinical interest between direct oral anticoagulants and antiepileptic drugs. Front Neurol. 2018;9:1067.
  • Löscher W, Klotz U, Zimprich F, et al. The clinical impact of pharmacogenetics on the treatment of epilepsy. Epilepsia. 2009;50(1):1–23.
  • Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry. 2018;51:9–62.
  • Bozina N, Sporis IS, Bozina T, et al. Pharmacogenomics and the treatment of epilepsy: what do we know?. Pharmacogenomics. 2019;20(15):1093–1101.
  • Smith RL, Haslemo T, Refsum H, et al. Impact of age, gender and CYP2C9/2C19 genotypes on dose-adjusted steady-state serum concentrations of valproic acid – a large-scale study based on naturalistic therapeutic drug monitoring data. Eur J Clin Pharmacol. 2016;72(9):1099–1104.
  • Smith RL, Haslemo T, Chan HF, et al. Clinically relevant effect of UGT1A4*3 on lamotrigine serum concentration is restricted to postmenopausal women – a study matching therapeutic drug monitoring and genotype data from 534 patients. Ther Drug Monit. 2018;40(5):567–571.
  • Tang F, Hartz AMS, Bauer B. Drug-resistant epilepsy: multiple hypotheses, few answers. Front Neurol. 2017;8:301. eCollection 2017.
  • Mirza N, Vaseiva O, Appleton R. An integrative in silico system for predicting dysregulated genes in the human epileptic focus: application to SLC transporters. Epilepsia. 2016;57:1467–1474.
  • Franco V, Baruffi K, Fransesca Crema TM, et al. Determination of perampanel in dried blood spots; applicability to therapeutic drug monitoring. Ther Drug Monit. 2019 Jul 29. Epub ahead of print.
  • Velghe S, Deprez S, Stove CP. Fully automated therapeutic drug monitoring of antiepileptic drugs making use of dried blood spots. J Chromatogr A. 2019;1601:95–103.
  • Wilhelm AJ, den Burger JC, Swart EL. Therapeutic drug monitoring by dried blood spot: progress to date and future directions. Clin Pharmacokinet. 2014;53(11):961–973.
  • Shah NM, Hawwa AF, Millership JS, et al. Adherence to antiepileptic medicines in children: a multiple-methods assessment involving dried blood spot sampling. Epilepsia. 2013;54:1020–1027.
  • Linder C, Neideman M, Wide K, et al. Dried blood spot self-sampling by guardians of children with epilepsy is feasible; comparison with plasma for multiple antiepileptic drugs. Ther Drug Monit. 2019;41(4):509–518.
  • Linder C, Wide K, Walander M, et al. Comparison between dried blood spot and plasma sampling for therapeutic drug monitoring of antiepileptic drugs in children with epilepsy: a step towards home sampling. Clin Biochem. 2017;50(7–8):418–424.
  • D’Urso A, Cangemi G, Barco S, et al. LC-MS/MS-based quantification of 9 antiepileptic drugs from a dried sample spot device. Ther Drug Monit. 2019;41(3):331–339.
  • D’Urso A, Rudge J, Patsalos PN, et al. Volumetric absorptive microsampling: a new sampling tool. Ther Drug Monit. 2019;41(5):681–692.
  • Johannessen SI, Gerna M, Bakke J, et al. CSF concentrations and serum protein binding of carbamazepine and carbamazepine-10,11-epoxide in epileptic patients. Br J Clin Pharmacol. 1976;3:575–582.
  • Bertilsson L, Tomson T. Clinical pharmacokinetics and pharmacological effects of carbamazepine and carbamazepine-10,11-epoxide. An Update Clin Pharmacokinet. 1986;11:177–198.
  • Pisani F, Caputo M, Fazio A, et al. Interaction of carbamazepine-10-11-epoxide, an active metabolite of carbamazepine, with valproate: a pharmacokinetic study. Epilepsia. 1990;31:339–342.
  • Burianová I, Borecka K. Routine therapeutic monitoring of the active metabolite of carbamazepine: is it really necessary? Clin Biochem. 2015;48:866–869.
  • Stockis A, Sargentini-Maier ML, Brodie MJ. Pharmacokinetic interaction of brivaracetam on carbamazepine in adult patients with epilepsy, with and without valproate co-administration. Epilepsy Res. 2016;128:163–168.
  • Reimers A, Andsnes Berg J, Larsen Burns M, et al. Reference ranges for antiepileptic drugs revisited: a practical approach to establish national guidelines. Drug Des Devel Ther. 2018b;12:271–280.
  • Specht U, Elsner H, May TW, et al. Postictal serum levels of antiepileptic drugs for detection of noncompliance. Epilepsy Behav. 2003;4(5):487–495.
  • Samsonsen C, Reimers A, Bråthen G, et al. Nonadherence to treatment causing acute hospitalizations in people with epilepsy: an observational, prospective study. Epilepsia. 2014;55(11):e125–128.
  • Touw DJ, Neef C, Thomson AH, et al. Cost-effectiveness of therapeutic drug monitoring: a systematic review. Ther Drug Monit. 2005;27:10–17.
  • Eadie MJ. Plasma antiepileptic drug monitoring in a neurological practice: a 25-year experience. Ther Drug Monit. 1994;16:458–468.
  • Eadie MJ. The role of therapeutic drug monitoring in improving the cost effectiveness of anticonvulsant therapy. Clin Pharmacokinet. 1995;29:29–35.
  • Rane CT, Dalvi SS, Gogtay NJ, et al. A pharmacoeconomic analysis of the impact of therapeutic drug monitoring in adult patients with generalized tonic-clonic epilepsy. Br J Clin Pharmacol. 2001;52:193–195.
  • Salih MR, Bahari MB, Shafie AA, et al. Cost-effectiveness analysis for the use of serum antiepileptic drug level monitoring in children with structural-metabolic epilepsy. Epilepsy Res. 2013;104:151–157.
  • Hussain SA, Ortendahl JD, Bentley TGK, et al. The economical burden of caregiving in epilepsy: an estimate based on a survey of US caregivers. Epilepsia. 2020 Jan 17. Epub ahead of print.
  • Perucca E. Is there a role for therapeutic drug monitoring of new anticonvulsants? Clin Pharmacokinet. 2000;38:191–204.
  • Woo E, Chan YM, Yu YL, et al. If a well-stabilized epileptic patient has a subtherapeutic antiepileptic drug level, should the dose be increased? A randomized prospective study. Epilepsia. 1988;29:129–139.
  • Buchthal F, Svensmark O, Schiller PJ. Clinical and electroencephalographic correlations with serum levels of diphenylhydantoin. Arch Neurol. 1960;2:624–630.
  • Schmidt D, Haenel F. Therapeutic plasma levels of phenytoin, phenobarbital, and carbamazepine: individual variation in relation to seizure frequency and type. Neurology. 1984;34:1252–1255.
  • Schmidt D, Einicke I, Haenel FT. The influence of seizure type on the efficacy of plasma concentrations of phenytoin, phenobarbital, and carbamazepine. Arch Neurol. 1986;43:263–265.
  • Gidal BE, Ferry J, Majid O, et al. Concentration-effect relationships with perampanel in patients with pharmacoresistant partial-onset seizures. Epilepsia. 2013;54:1490–1497.
  • Svendsen T, Brodtkorb E, Reimers A, et al. Pharmacokinetic variability, efficacy and tolerability of eslicarbazepine acetate – a national approach to the evaluation of therapeutic drug monitoring data and clinical outcome. Epilepsy Res. 2017a;129:125–131.
  • Svendsen T, Brodtkorb E, Baftiu A, et al. Therapeutic drug monitoring of lacosamide in Norway: focus on pharmacokinetic variability, efficacy and tolerability. Neurochem Res. 2017b;42(7):2077–2083.
  • Steinhoff BJ, Hübers E, Kurth C, et al. Plasma concentration and clinical effects of perampanel – the Kork experience. Seizure. 2019;28:18–22.
  • Burns ML, Nikanorova M, Baftiu A, et al. Pharmacokinetic variability and clinical use of lacosamide in children and adolescents in Denmark and Norway. Ther Drug Monit. 2019a;41(3):340–347.
  • 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.
  • May TW, Helmer R, Bien CG, et al. Influence of dose and antiepileptic comedication on lacosamide serum concentrations in patients with epilepsy of different ages. Ther Drug Monit. 2018;40(5):620–627.
  • Johannessen Landmark C, Larsson PG, Rytter E, et al. Antiepileptic drugs in epilepsy and other disorders: a population-based study of prescriptions. Epilepsy Res. 2009;87:31–39.
  • Baftiu A, Johannessen Landmark C, Rudberg Rusten I, et al. Safety aspects of utilization of antiepileptic drugs in epilepsy vs. non-epilepsy disorders. Eur J Clin Pharmacol. 2016;72(10):1245–1254.
  • Burns ML, Kinge E, Stokke Opdahl M, et al. Therapeutic drug monitoring of gabapentin in various indications. Acta Neurol Scand. 2019b;130(5):446–454.
  • Conway JM, Collins JF, Macias FM, et al. Factors in variability of serial gabapentin concentrations in elderly patients with epilepsy. Pharmacotherapy. 2017;37(10):1197–1203.
  • Bengtsson F. Therapeutic drug monitoring of psychotropic drugs. TDM “nouveau”. Ther Drug Monit. 2004;26:145–151.
  • World Health Organization. Adherence to long term therapies – evidence for action. World Health Organization 2003. Available from: http://apps.who.int/iris/bitstream/handle/10665/42682/9241545992.pdf
  • Davis KL, Candrilli SD, Edin HM. Prevalence and cost of nonadherence with antiepileptic drugs in an adult managed care population. Epilepsia. 2008;49:446–454.
  • Henning O, Johannessen Landmark C, Nakken KO, et al. Nonadherence to treatment regimens in epilepsy from the patients’ perspective and predisposing factors: differences between intentional and non-intentional lack of adherence. Epilepsia. 2019a;60(5):e58–e62.
  • Johannessen Landmark C, Fløgstad I, Syvertsen M, et al. Treatment with AEDs and challenges for patients with JME. Epi Behav. 2019;98:110–116.
  • Henning O, Lossius MI, Lima MH, et al. Refractory epilepsy and nonadherence to drug treatment. Epilepsia Open. 2019b;4:618–623.
  • Carpentier N, Jonas J, Frismand S, et al. Direct evidence of nonadherence to antiepileptic medication in refractory focal epilepsy. Epilepsia. 2013;54(1):e20–e23.
  • Lunardi M, Lin K, Walz R, et al. Single antiepileptic drug levels do not predict adherence and nonadherence. Acta Neurol Scand. 2019;139(2):199–203.
  • Johannessen Landmark C, Fløgstad I, Baftiu A, et al. Long term therapeutic drug monitoring of AEDs in patients with JME. Epilepsy Res. 2019;155:106148.
  • Mevaag M, Henning O, Baftiu A, et al. Discrepancies between physicians’ prescriptions and patients’ use of antiepileptic drugs. Acta Neurol Scand. 2017;135:80–87.
  • Holtkamp M, Theodore WH. Generic antiepileptic drugs – safe or harmful in patients with epilepsy? Epilepsia. 2018;59(7):1273–1281.
  • Kwan P, Arzimanoglou A, Berg AT, et al. Definition of drug resistant epilepsy: consensus proposal by the ad hoc task force of the ILAE commission on therapeutic strategies. Epilepsia. 2010;51:1069–1077.
  • Hao X, Goldberg D, Kelly K, et al. Uncontrolled epilepsy is not necessarily the same as drug-resistant epilepsy: differences between populations with newly diagnosed epilepsy and chronic epilepsy. Epilepsy Behav. 2013;29(1):4–6.
  • Bialer M, Johannessen SI, Koepp MJ, et al. Progress report on new antiepileptic drugs: a summary of the fourteenth Eilat conference on new antiepileptic drugs and devices (EILAT XiV) II. Drugs in more advanced clinical development. Epilepsia. 2018;59(10):1842–1866.
  • Tomson T, Dahl M, Kimland E. Therapeutic monitoring of antiepileptic drugs for epilepsy. Cochrane Database Syst Rev. 2007c;1:CD002216.
  • Fröscher W, Eichelbaum M, Gugler R, et al. A prospective randomised trial on the effect of monitoring plasma anticonvulsant levels in epilepsy. J Neurol. 1981;224:193–201.
  • Jannuzzi G, Cian P, Fattore C, et al. A multicenter randomized controlled trial on the clinical impact of therapeutic drug monitoring in patients with newly diagnosed epilepsy. Epilepsia. 2000;41:222–230.
  • Thangaratinam S, Marlin N, Newton S, et al. AntiEpileptic drug monitoring in PREgnancy (EMPiRE): a double-blind randomized trial on effectiveness and acceptability of monitoring strategies. Health Technol Assess. 2018;22(23):1–152.
  • Chen Z, Liew D, Kwan P. Real-world efficiency of pharmacogenetic screening for carbamazepine-induced severe cutaneous adverse reactions. PLoS One. 2014;9(5):e96990.
  • Tassaneeyakul W, Prabmeechai N, Sukasem C, et al. Associations between HLA class I and cytochrome P450 2C9 genetic polymorphisms and phenytoin-related severe cutaneous adverse reactions in a Thai population. Pharmacogenet Genomics. 2016;26(5):225–234.
  • Chouchi M, Kaabachi W, Tizaoui K, et al. The HLA-B*15:02 polymorphism and Tegretol®-induced serious cutaneous reactions in epilepsy: an updated systematic review and meta-analysis. Rev Neurol (Paris). 2018;174(5):278–291.

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.