Drug Resistance in Epilepsy: Clinical Impact, Potential Mechanisms, and New Innovative Treatment Options

Wolfgang Löscher, Heidrun Potschka, Sanjay M Sisodiya, Annamaria Vezzani, Wolfgang Löscher, Heidrun Potschka, Sanjay M Sisodiya, Annamaria Vezzani

Abstract

Epilepsy is a chronic neurologic disorder that affects over 70 million people worldwide. Despite the availability of over 20 antiseizure drugs (ASDs) for symptomatic treatment of epileptic seizures, about one-third of patients with epilepsy have seizures refractory to pharmacotherapy. Patients with such drug-resistant epilepsy (DRE) have increased risks of premature death, injuries, psychosocial dysfunction, and a reduced quality of life, so development of more effective therapies is an urgent clinical need. However, the various types of epilepsy and seizures and the complex temporal patterns of refractoriness complicate the issue. Furthermore, the underlying mechanisms of DRE are not fully understood, though recent work has begun to shape our understanding more clearly. Experimental models of DRE offer opportunities to discover, characterize, and challenge putative mechanisms of drug resistance. Furthermore, such preclinical models are important in developing therapies that may overcome drug resistance. Here, we will review the current understanding of the molecular, genetic, and structural mechanisms of ASD resistance and discuss how to overcome this problem. Encouragingly, better elucidation of the pathophysiological mechanisms underpinning epilepsies and drug resistance by concerted preclinical and clinical efforts have recently enabled a revised approach to the development of more promising therapies, including numerous potential etiology-specific drugs ("precision medicine") for severe pediatric (monogenetic) epilepsies and novel multitargeted ASDs for acquired partial epilepsies, suggesting that the long hoped-for breakthrough in therapy for as-yet ASD-resistant patients is a feasible goal. SIGNIFICANCE STATEMENT: Drug resistance provides a major challenge in epilepsy management. Here, we will review the current understanding of the molecular, genetic, and structural mechanisms of drug resistance in epilepsy and discuss how the problem might be overcome.

Copyright © 2020 by The Author(s).

Figures

Fig. 1.
Fig. 1.
Introduction of antiseizure drugs (ASDs) to the market from 1853 to 2019. Licensing varied from country to country. We give here the year of first licensing or the first mention of clinical use in a country of Europe, the United States, or Japan. We have not included all derivatives of listed ASDs nor ASDs used solely for treatment of status epilepticus. The first generation of ASDs, entering the market from 1857 to 1958, includes potassium bromide, phenobarbital, and a variety of drugs that were mainly derived by modification of the barbiturate structure, including phenytoin, primidone, trimethadione, and ethosuximide. The second-generation ASDs, including carbamazepine, valproate, and the benzodiazepines, which were introduced between 1960 and 1975, differed chemically from the barbiturates. The era of the third-generation ASDs started in the 1980s with “rational” (target-based) developments such as progabide, vigabatrin, and tiagabine, i.e., drugs that were designed to selectively target a mechanism that was thought to be critical for the occurrence of epileptic seizures. The figure also illustrates the impact of preclinical seizure models on ASD development. The use of seizure models for drug screening started with the experiments performed by Merritt and Putnam in the 1930s, who used an electroshock seizure model in cats, leading to the discovery of phenytoin. Subsequently, the electroshock model was adapted to rodents and, together with chemical seizure models, used for drug screening in diverse laboratories, leading to discovery of various additional ASDs. In 1975, the NIH/NINDS ASP was established in the United States as part of a larger Antiepileptic Drug Development program to promote industry interest in ASD development. Since its start, the seizure tests have been performed at a contract facility based at the University of Utah, using three rodent models, i.e., the maximal electroshock seizure (MES) test, the pentylenetetrazole (PTZ) seizure test, and the rotarod test for assessing neurotoxicity. Later, other seizure models were added. The seizure tests were performed on a blinded and confidential basis and at no cost to the ASP participants, thus providing opportunities for researchers from academia and industry in the United States and abroad to submit compounds for screening in a battery of well established rodent seizure models. Approximately 32,000 compounds from more than 600 participants from 38 countries have been screened by this program, and the ASP has contributed to bringing nine currently available ASDs to market since 1990 (Kehne et al., 2017). More recently (2016), the ASP has been renamed Epilepsy Therapy Screening Program (ETSP) with the refocused mission to identify novel agents that will help address the considerable remaining unmet medical needs in epilepsy, particularly ASD-resistant seizures (Kehne et al., 2017). Figure modified from Löscher and Schmidt (2011). For further details, see text and Löscher et al. (2013a).
Fig. 2.
Fig. 2.
Pharmacoresistant epilepsy workflow for the Epilepsy Therapy Screening Program (ETSP). The figure has been provided by John Kehne and slightly modified for consistency with the text of this review. For details, see text and Kehne et al. (2017) and Wilcox et al. (2020).
Fig. 3.
Fig. 3.
Various potential mechanisms of ASD resistance or factors predicting poor outcome have been implicated in patients with epilepsy and animal models of medically resistant seizures, indicating that intrinsic or acquired resistance to ASDs is a multifactorial phenomenon. Based on these findings, a number of hypotheses of ASD resistance, including the target, transporter, network, intrinsic severity, and genetic variant hypotheses, have been suggested (see text). These hypotheses are not mutually exclusive but may be relevant for the same patient, thus complicating any strategy to counteract or reverse pharmacoresistance. Modified from Löscher et al. (2013a).
Fig. 4.
Fig. 4.
Differences between ASD responders and nonresponders in two animal models of DRE. For comparison, alterations associated with ASD resistance in patients are shown. Those alterations that occur both in the models and in patients are highlighted by the colored boxes. For details, see Löscher (2011), Löscher et al. (2013a), and Löscher (2016).
Fig. 5.
Fig. 5.
Selection and characterization of ASD responders and nonresponders by phenobarbital in a rat model of TLE in which spontaneous recurrent seizures (SRS) develop following sustained electrical stimulation of the basolateral amygdala. (A) Schematic illustration of selection of drug-resistant and drug-responsive epileptic rats by prolonged administration of phenobarbital. (B) Effect of phenobarbital (PB) on SRS. About 5 months after the electrically induced SE, SRS were recorded over a period of 2 weeks before onset of PB treatment (predrug control), followed by drug treatment of 2 weeks and then a 2-week postdrug control period. All data are shown as means ± S.E.M. The graphs in (B) show 1) average seizure data from 33 epileptic rats from three prospective experiments, 2) respective data from 20 responders, 3) data from 13 nonresponders, and 4) average plasma concentration of PB from the blood samples taken at the end of the treatment period. The shaded area indicates the therapeutic plasma concentration range of PB. In the responder group, PB significantly suppressed SRS compared with the pre- and postdrug periods (*P < 0.001). Note the higher average frequency of SRS in nonresponders versus responders. (C) Pgp expression in brain capillary endothelial cells of responders and nonresponders. Significant differences are indicated by asterisk (*P < 0.05). (D) Coadministration of PB and the Pgp inhibitor tariquidar lead to a significant (*P < 0.05) suppression of SRS in PB nonresponders. Three different doses of tariquidar (10, 15, and 20 mg/kg) were used, demonstrating a dose-dependent effect. Data are from Brandt et al. (2004), Volk and Löscher (2005), Brandt et al. (2006), Bethmann et al. (2007), and Brandt and Löscher (2014).
Fig. 6.
Fig. 6.
Schematic representation of the evidence-based pathologic links between inflammatory mediators and mechanisms of drug resistance. Inflammatory mediators (including but not limited to cytokines) may contribute to drug-resistant seizures mainly by three (nonmutually exclusive) pathways: 1) the induction of BBB dysfunction by promoting breakdown of tight junctions or inducing transocytosis, aberrant angiogenesis generating “leaky” vessels, and oxidative stress. The inflammatory phenotype of astrocytes is pivotal for these actions to take place, and reciprocally, BBB permeability changes may promote the expression of inflammatory molecules in astrocytes. This vicious cycle contributes to recurrent seizures, cell loss, and maladaptive neuronal network plasticity, therefore contributing to increase the “intrinsic severity” of the disease. Morever, BBB dysfunction will enhance albumin brain extravasation into the brain parenchyma and potentially increase the “buffering” effect of albumin binding to drugs, thus decreasing functionally relevant unbound drug levels at brain target sites. 2) Another mechanism is the induction of Pgp in endothelial cells, and likely in perivascular astrocytes, by specific inflammatory pathways involving COX2-PGE2-EP1R and the IL-1beta-IL-1R1 axis, thus contributing to the transporter hypothesis of drug resistance. 3) Inflammatory mediators can also induce post-translational modifications in voltage-gated and receptor-operated ion channels resulting in less responsive ASD targets, which may contribute to the pharmacodynamic (target) hypothesis of drug resistance. Details and references are reported in the main text.

References

    1. Abbott NJ. (2000) Inflammatory mediators and modulation of blood-brain barrier permeability. Cell Mol Neurobiol 20:131–147.
    1. Abbott NJ. (2013) Blood-brain barrier structure and function and the challenges for CNS drug delivery. J Inherit Metab Dis 36:437–449.
    1. Asadi-Pooya AA, Razavizadegan SM, Abdi-Ardekani A, Sperling MR. (2013) Adjunctive use of verapamil in patients with refractory temporal lobe epilepsy: a pilot study. Epilepsy Behav 29:150–154.
    1. Avemary J, Salvamoser JD, Peraud A, Rémi J, Noachtar S, Fricker G, Potschka H. (2013) Dynamic regulation of P-glycoprotein in human brain capillaries. Mol Pharm 10:3333–3341.
    1. Balestrini S, Sisodiya SM. (2018) Pharmacogenomics in epilepsy. Neurosci Lett 667:27–39.
    1. Balosso S, Maroso M, Sanchez-Alavez M, Ravizza T, Frasca A, Bartfai T, Vezzani A. (2008) A novel non-transcriptional pathway mediates the proconvulsive effects of interleukin-1beta. Brain 131:3256–3265.
    1. Bankstahl M, Bankstahl JP, Löscher W. (2012) Inter-individual variation in the anticonvulsant effect of phenobarbital in the pilocarpine rat model of temporal lobe epilepsy. Exp Neurol 234:70–84.
    1. Bankstahl M, Bankstahl JP, Löscher W. (2013) Pilocarpine-induced epilepsy in mice alters seizure thresholds and the efficacy of antiepileptic drugs in the 6-Hertz psychomotor seizure model. Epilepsy Res 107:205–216.
    1. Baraban S, Löscher W. (2014) What new modeling approaches will help us identify promising drug treatments? Adv Exp Med Biol 813:283–294.
    1. Bar-Klein G, Cacheaux LP, Kamintsky L, Prager O, Weissberg I, Schoknecht K, Cheng P, Kim SY, Wood L, Heinemann U, et al. (2014) Losartan prevents acquired epilepsy via TGF-β signaling suppression. Ann Neurol 75:864–875.
    1. Bar-Klein G, Lublinsky S, Kamintsky L, Noyman I, Veksler R, Dalipaj H, Senatorov VV, Jr., Swissa E, Rosenbach D, Elazary N, et al. (2017) Imaging blood-brain barrier dysfunction as a biomarker for epileptogenesis. Brain 140:1692–1705.
    1. Barkovich AJ, Dobyns WB, Guerrini R. (2015) Malformations of cortical development and epilepsy. Cold Spring Harb Perspect Med 5:a022392.
    1. Barton ME, Klein BD, Wolf HH, White HS. (2001) Pharmacological characterization of the 6 Hz psychomotor seizure model of partial epilepsy. Epilepsy Res 47:217–227.
    1. Bauer B, Hartz AM, Pekcec A, Toellner K, Miller DS, Potschka H. (2008) Seizure-induced up-regulation of P-glycoprotein at the blood-brain barrier through glutamate and cyclooxygenase-2 signaling. Mol Pharmacol 73:1444–1453.
    1. Bauer M, Karch R, Zeitlinger M, Liu J, Koepp M, Asselin M-C, Sisodiya S, Hainfellner J, Wadsak W, Mitterhauser M, et al. (2014) In vivo P-glycoprotein function before and after epilepsy surgery. Neurology 83:1326–1331.
    1. Ben-Ari Y, Khalilov I, Kahle KT, Cherubini E. (2012) The GABA excitatory/inhibitory shift in brain maturation and neurological disorders. Neuroscientist 18:467–486.
    1. Berendt M, Farquhar R, Mandigers P, Pakozdy A, Bhatti S, De Risio L, Fischer A, Long S, Matiasek K, Muñana K, et al. (2015) International veterinary epilepsy task force consensus report on epilepsy definition, classification and terminology in companion animals. BMC Vet Res 11:182.
    1. Berg AT, Langfitt J, Shinnar S, Vickrey BG, Sperling MR, Walczak T, Bazil C, Pacia SV, Spencer SS. (2003) How long does it take for partial epilepsy to become intractable? Neurology 60:186–190.
    1. Bethmann K, Brandt C, Löscher W. (2007) Resistance to phenobarbital extends to phenytoin in a rat model of temporal lobe epilepsy. Epilepsia 48:816–826.
    1. Bethmann K, Fritschy JM, Brandt C, Löscher W. (2008) Antiepileptic drug resistant rats differ from drug responsive rats in GABA A receptor subunit expression in a model of temporal lobe epilepsy. Neurobiol Dis 31:169–187.
    1. Bialer M, Johannessen SI, Levy RH, Perucca E, Tomson T, White HS. (2013) Progress report on new antiepileptic drugs: a summary of the Eleventh Eilat Conference (EILAT XI). Epilepsy Res 103:2–30.
    1. Bialer M, Johannessen S, Koepp M, Levy R, Perucca E, Tomson T, White H. (2018) Progress report on new antiepileptic drugs: A summary of the Fourteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIV). I. Drugs in preclinical and early clinical development. Epilepsia 59:1811–1841.
    1. Blanco MM, dos Santos JG, Jr., Perez-Mendes P, Kohek SR, Cavarsan CF, Hummel M, Albuquerque C, Mello LE. (2009) Assessment of seizure susceptibility in pilocarpine epileptic and nonepileptic Wistar rats and of seizure reinduction with pentylenetetrazole and electroshock models. Epilepsia 50:824–831.
    1. Borlot F, Wither RG, Ali A, Wu N, Verocai F, Andrade DM. (2014) A pilot double-blind trial using verapamil as adjuvant therapy for refractory seizures. Epilepsy Res 108:1642–1651.
    1. Brandt C, Bethmann K, Gastens AM, Löscher W. (2006) The multidrug transporter hypothesis of drug resistance in epilepsy: proof-of-principle in a rat model of temporal lobe epilepsy. Neurobiol Dis 24:202–211.
    1. Brandt C, Löscher W. (2014) Antiepileptic efficacy of lamotrigine in phenobarbital-resistant and -responsive epileptic rats: a pilot study. Epilepsy Res 108:1145–1157.
    1. Brandt C, Volk HA, Löscher W. (2004) Striking differences in individual anticonvulsant response to phenobarbital in rats with spontaneous seizures after status epilepticus. Epilepsia 45:1488–1497.
    1. Brigo F, Striano P, Balagura G, Belcastro V. (2018) Emerging drugs for the treatment of Dravet syndrome. Expert Opin Emerg Drugs 23:261–269.
    1. Brodie MJ. (2016) Pharmacological treatment of drug-resistant epilepsy in adults: a practical guide. Curr Neurol Neurosci Rep 16:82.
    1. Broekaart DWM, Anink JJ, Baayen JC, Idema S, de Vries HE, Aronica E, Gorter JA, van Vliet EA. (2018) Activation of the innate immune system is evident throughout epileptogenesis and is associated with blood-brain barrier dysfunction and seizure progression. Epilepsia 59:1931–1944.
    1. Brooks-Kayal AR, Shumate MD, Jin H, Rikhter TY, Coulter DA. (1998) Selective changes in single cell GABA(A) receptor subunit expression and function in temporal lobe epilepsy. Nat Med 4:1166–1172.
    1. Brown WC, Schiffman DO, Swinyard EA, Goodman LS. (1953) Comparative assay of an antiepileptic drugs by psychomotor seizure test and minimal electroshock threshold test. J Pharmacol Exp Ther 107:273–283.
    1. Brückl TM, Uhr M. (2016) ABCB1 genotyping in the treatment of depression. Pharmacogenomics 17:2039–2069.
    1. Chen Z, Brodie MJ, Liew D, Kwan P. (2018) Treatment outcomes in patients with newly diagnosed epilepsy treated with established and new antiepileptic drugs: a 30-year longitudinal cohort study. JAMA Neurol 75:279–286.
    1. Chouchi M, Kaabachi W, Klaa H, Tizaoui K, Turki IB, Hila L. (2017) Relationship between ABCB1 3435TT genotype and antiepileptic drugs resistance in Epilepsy: updated systematic review and meta-analysis. BMC Neurol 17:32.
    1. Chung FS, Santiago JS, Jesus MF, Trinidad CV, See MF. (2016) Disrupting P-glycoprotein function in clinical settings: what can we learn from the fundamental aspects of this transporter? Am J Cancer Res 6:1583–1598.
    1. Clarkson B, LaFrance-Corey R, Kahoud R, Farias-Moeller R, Payne E, Howe C. (2019) Functional deficiency in endogenous interleukin-1 receptor antagonist in patients with febrile infection-related epilepsy syndrome. Ann Neurol 85:526–537.
    1. Cohen I, Navarro V, Clemenceau S, Baulac M, Miles R. (2002) On the origin of interictal activity in human temporal lobe epilepsy in vitro. Science 298:1418–1421.
    1. Coulter DA. (2000) Mossy fiber zinc and temporal lobe epilepsy: pathological association with altered “epileptic” gamma-aminobutyric acid A receptors in dentate granule cells. Epilepsia 41 (Suppl 6):S96–S99.
    1. Coulter DA. (2001) Epilepsy-associated plasticity in gamma-aminobutyric acid receptor expression, function, and inhibitory synaptic properties. Int Rev Neurobiol 45:237–252.
    1. Cucullo L, Hossain M, Rapp E, Manders T, Marchi N, Janigro D. (2007) Development of a humanized in vitro blood-brain barrier model to screen for brain penetration of antiepileptic drugs. Epilepsia 48:505–516.
    1. Dalic L, Cook MJ. (2016) Managing drug-resistant epilepsy: challenges and solutions. Neuropsychiatr Dis Treat 12:2605–2616.
    1. Demarest ST, Brooks-Kayal A. (2018) From molecules to medicines: the dawn of targeted therapies for genetic epilepsies. Nat Rev Neurol 14:735–745.
    1. Deng X, Shao Y, Xie Y, Feng Y, Wu M, Wang M, Chen Y. (2019) MicroRNA-146a-5p downregulates the expression of P-glycoprotein in rats with lithium-pilocarpine-induced status epilepticus. Biol Pharm Bull 42:744–750.
    1. DeSena AD, Do T, Schulert GS. (2018) Systemic autoinflammation with intractable epilepsy managed with interleukin-1 blockade. J Neuroinflammation 15:38.
    1. Devinsky O, Vezzani A, O’Brien TJ, Jette N, Scheffer IE, De Curtis M, Perucca P. (2018) Epilepsy. Nat Rev Dis Primers 4, doi: 10.1038/nrdp.2018.24.
    1. Dilena R, Mauri E, Aronica E, Bernasconi P, Bana C, Cappelletti C, Carrabba G, Ferrero S, Giorda R, Guez S, et al. (2019) Therapeutic effect of Anakinra in the relapsing chronic phase of febrile infection-related epilepsy syndrome. Epilepsia Open 4:344–350.
    1. Doeser A, Dickhof G, Reitze M, Uebachs M, Schaub C, Pires NM, Bonifácio MJ, Soares-da-Silva P, Beck H. (2015) Targeting pharmacoresistant epilepsy and epileptogenesis with a dual-purpose antiepileptic drug. Brain 138:371–387.
    1. Du X, Parent JM. (2015) Using patient-derived induced pluripotent stem cells to model and treat epilepsies. Curr Neurol Neurosci Rep 15:71.
    1. Dudvarski Stankovic N, Teodorczyk M, Ploen R, Zipp F, Schmidt MHH. (2016) Microglia-blood vessel interactions: a double-edged sword in brain pathologies. Acta Neuropathol 131:347–363.
    1. Duveau V, Pouyatos B, Bressand K, Bouyssieres C, Chabrol T, Roche Y, Depaulis A, Roucard C. (2016) Differential effects of antiepileptic drugs on focal seizures in the intrahippocampal kainate mouse model of mesial temporal lobe epilepsy. CNS Neurosci Ther 22:497–506.
    1. Eastman CL, Verley DR, Fender JS, Stewart TH, Nov E, Curia G, D’Ambrosio R. (2011) Antiepileptic and antiepileptogenic performance of carisbamate after head injury in the rat: blind and randomized studies. J Pharmacol Exp Ther 336:779–790.
    1. Ellerkmann RK, Remy S, Chen J, Sochivko D, Elger CE, Urban BW, Becker A, Beck H. (2003) Molecular and functional changes in voltage-dependent Na(+) channels following pilocarpine-induced status epilepticus in rat dentate granule cells. Neuroscience 119:323–333.
    1. Elmore M, Najafi A, Koike M, Dagher N, Spangenberg E, Rice R, Kitazawa M, Matusow B, Nguyen H, West B, et al. (2014) Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82:380–397.
    1. Epi25 Collaborative. Electronic address: s.berkovic@unimelb.edu.au; Epi25 Collaborative (2019) Ultra-rare genetic variation in the epilepsies: a whole-exome sequencing study of 17,606 individuals. Am J Hum Genet 105:267–282.
    1. Fang M, Xi ZQ, Wu Y, Wang XF. (2011) A new hypothesis of drug refractory epilepsy: neural network hypothesis. Med Hypotheses 76:871–876.
    1. Feldmann M, Asselin MC, Liu J, Wang S, McMahon A, Anton-Rodriguez J, Walker M, Symms M, Brown G, Hinz R, et al. (2013) P-glycoprotein expression and function in patients with temporal lobe epilepsy: a case-control study. Lancet Neurol 12:777–785.
    1. Ferrari CC, Depino AM, Prada F, Muraro N, Campbell S, Podhajcer O, Perry VH, Anthony DC, Pitossi FJ. (2004) Reversible demyelination, blood-brain barrier breakdown, and pronounced neutrophil recruitment induced by chronic IL-1 expression in the brain. Am J Pathol 165:1827–1837.
    1. Frederiksen K, Lu D, Yang J, Jensen H, Bastlund J, Larsen P, Liu H, Crestey F, Dekermendjian K, Badolo L, et al. (2017) A small molecule activator of Nav 1.1 channels increases fast-spiking interneuron excitability and GABAergic transmission in vitro and has anti-convulsive effects in vivo. Eur J Neurosci 46:1887–1896.
    1. French JA, Koepp M, Naegelin Y, Vigevano F, Auvin S, Rho JM, Rosenberg E, Devinsky O, Olofsson PS, Dichter MA. (2017) Clinical studies and anti-inflammatory mechanisms of treatments. Epilepsia 58 (Suppl 3):69–82.
    1. Friedman A, Heinemann U. (2012) Role of blood-brain barrier dysfunction in epileptogenesis. SourceJasper’s Basic Mechanisms of the Epilepsies, National Center for Biotechnology Information (US), Bethesda, MD.
    1. Frigerio F, Flynn C, Han Y, Lyman K, Lugo JN, Ravizza T, Ghestem A, Pitsch J, Becker A, Anderson AE, et al. (2018) Neuroinflammation alters integrative properties of rat hippocampal pyramidal cells. Mol Neurobiol 55:7500–7511.
    1. Frigerio F, Frasca A, Weissberg I, Parrella S, Friedman A, Vezzani A, Noé FM. (2012) Long-lasting pro-ictogenic effects induced in vivo by rat brain exposure to serum albumin in the absence of concomitant pathology. Epilepsia 53:1887–1897.
    1. Fromm MF. (2004) Importance of P-glycoprotein at blood-tissue barriers. Trends Pharmacol Sci 25:423–429.
    1. Galanopoulou AS, Moshé SL. (2015) Pathogenesis and new candidate treatments for infantile spasms and early life epileptic encephalopathies: a view from preclinical studies. Neurobiol Dis 79:135–149.
    1. Galovic M, Baudracco I, Wright-Goff E, Pillajo G, Nachev P, Wandschneider B, Woermann F, Thompson P, Baxendale S, McEvoy AW, et al. (2019) Association of piriform cortex resection with surgical outcomes in patients with temporal lobe epilepsy. JAMA Neurol 76:690–700.
    1. Gambardella A, Labate A, Mumoli L, Lopes-Cendes I, Cendes F. (2017) Role of pharmacogenomics in antiepileptic drug therapy: current status and future perspectives. Curr Pharm Des 23:5760–5765.
    1. Ghosh C, Puvenna V, Gonzalez-Martinez J, Janigro D, Marchi N. (2011) Blood-brain barrier P450 enzymes and multidrug transporters in drug resistance: a synergistic role in neurological diseases. Curr Drug Metab 12:742–749.
    1. Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, Dahlin A, Evers R, Fischer V, Hillgren KM, et al. International Transporter Consortium (2010) Membrane transporters in drug development. Nat Rev Drug Discov 9:215–236.
    1. Golyala A, Kwan P. (2017) Drug development for refractory epilepsy: the past 25 years and beyond. Seizure 44:147–156.
    1. Goodkin HP, Yeh JL, Kapur J. (2005) Status epilepticus increases the intracellular accumulation of GABAA receptors. J Neurosci 25:5511–5520.
    1. Griffin A, Hamling KR, Hong S, Anvar M, Lee LP, Baraban SC. (2018) Preclinical animal models for Dravet syndrome: seizure phenotypes, comorbidities and drug screening. Front Pharmacol 9:573.
    1. Grone B, Baraban S. (2015) Animal models in epilepsy research: legacies and new directions. Nat Neurosci 18:339–343.
    1. Guo JU, Ma DK, Mo H, Ball MP, Jang MH, Bonaguidi MA, Balazer JA, Eaves HL, Xie B, Ford E, et al. (2011) Neuronal activity modifies the DNA methylation landscape in the adult brain. Nat Neurosci 14:1345–1351.
    1. Haenisch S, von Rüden EL, Wahmkow H, Rettenbeck ML, Michler C, Russmann V, Bruckmueller H, Waetzig V, Cascorbi I, Potschka H. (2016) miRNA-187-3p-Mediated regulation of the KCNK10/TREK-2 potassium channel in a rat epilepsy model. ACS Chem Neurosci 7:1585–1594.
    1. Haerian BS, Lim KS, Tan CT, Raymond AA, Mohamed Z. (2011) Association of ABCB1 gene polymorphisms and their haplotypes with response to antiepileptic drugs: a systematic review and meta-analysis. Pharmacogenomics 12:713–725.
    1. Hartz AM, Pekcec A, Soldner EL, Zhong Y, Schlichtiger J, Bauer B. (2017) P-gp protein expression and transport activity in rodent seizure models and human epilepsy. Mol Pharm 14:999–1011.
    1. Hitiris N, Mohanraj R, Norrie J, Sills GJ, Brodie MJ. (2007) Predictors of pharmacoresistant epilepsy. Epilepsy Res 75:192–196.
    1. Holtman L, van Vliet EA, Edelbroek PM, Aronica E, Gorter JA. (2010) Cox-2 inhibition can lead to adverse effects in a rat model for temporal lobe epilepsy. Epilepsy Res 91:49–56.
    1. Holtman L, van Vliet EA, van Schaik R, Queiroz CM, Aronica E, Gorter JA. (2009) Effects of SC58236, a selective COX-2 inhibitor, on epileptogenesis and spontaneous seizures in a rat model for temporal lobe epilepsy. Epilepsy Res 84:56–66.
    1. Hsiao J, Yuan TY, Tsai MS, Lu CY, Lin YC, Lee ML, Lin SW, Chang FC, Liu Pimentel H, Olive C, et al. (2016) Upregulation of haploinsufficient gene expression in the brain by targeting a long non-coding RNA improves seizure phenotype in a model of Dravet syndrome. EBioMedicine 9:257–277.
    1. Iannone LF, Preda A, Blottière HM, Clarke G, Albani D, Belcastro V, Carotenuto M, Cattaneo A, Citraro R, Ferraris C, et al. (2019) Microbiota-gut brain axis involvement in neuropsychiatric disorders. Expert Rev Neurother 19:1037–1050.
    1. Iori V, Iyer AM, Ravizza T, Beltrame L, Paracchini L, Marchini S, Cerovic M, Hill C, Ferrari M, Zucchetti M, et al. (2017) Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis 99:12–23.
    1. Jandová K, Päsler D, Antonio LL, Raue C, Ji S, Njunting M, Kann O, Kovács R, Meencke HJ, Cavalheiro EA, et al. (2006) Carbamazepine-resistance in the epileptic dentate gyrus of human hippocampal slices. Brain 129:3290–3306.
    1. Jeub M, Beck H, Siep E, Rüschenschmidt C, Speckmann EJ, Ebert U, Potschka H, Freichel C, Reissmüller E, Löscher W. (2002) Effect of phenytoin on sodium and calcium currents in hippocampal CA1 neurons of phenytoin-resistant kindled rats. Neuropharmacology 42:107–116.
    1. Jyonouchi H, Geng L. (2016) Intractable epilepsy (IE) and responses to anakinra, a human recombinant IL-1 receptor antagonist (IL-1Ra): case reports. J Clin Cell Immunol 7:456–460.
    1. Kahle KT, Staley KJ, Nahed BV, Gamba G, Hebert SC, Lifton RP, Mount DB. (2008) Roles of the cation-chloride cotransporters in neurological disease. Nat Clin Pract Neurol 4:490–503.
    1. Kalilani L, Sun X, Pelgrims B, Noack-Rink M, Villanueva V. (2018) The epidemiology of drug-resistant epilepsy: a systematic review and meta-analysis. Epilepsia 59:2179–2193.
    1. Kaminski RM, Matagne A, Patsalos PN, Klitgaard H. (2009) Benefit of combination therapy in epilepsy: a review of the preclinical evidence with levetiracetam. Epilepsia 50:387–397.
    1. Keezer MR, Sisodiya SM, Sander JW. (2016) Comorbidities of epilepsy: current concepts and future perspectives. Lancet Neurol 15:106–115.
    1. Kehne JH, Klein BD, Raeissi S, Sharma S. (2017) The National Institute of neurological disorders and stroke (NINDS) epilepsy therapy screening program (ETSP). Neurochem Res 42:1894–1903.
    1. Kelley MS, Jacobs MP, Lowenstein DH, NINDS Epilepsy Benchmark Stewards (2009) The NINDS epilepsy research benchmarks. Epilepsia 50:579–582.
    1. Kenney-Jung DL, Vezzani A, Kahoud RJ, LaFrance-Corey RG, Ho ML, Muskardin TW, Wirrell EC, Howe CL, Payne ET. (2016) Febrile infection-related epilepsy syndrome treated with anakinra. Ann Neurol 80:939–945.
    1. Kim SY, Senatorov VV, Jr., Morrissey CS, Lippmann K, Vazquez O, Milikovsky DZ, Gu F, Parada I, Prince DA, Becker AJ, et al. (2017) TGFβ signaling is associated with changes in inflammatory gene expression and perineuronal net degradation around inhibitory neurons following various neurological insults. Sci Rep 7:7711.
    1. Klein S, Bankstahl M, Löscher W. (2015) Inter-individual variation in the effect of antiepileptic drugs in the intrahippocampal kainate model of mesial temporal lobe epilepsy in mice. Neuropharmacology 90:53–62.
    1. Klement W, Blaquiere M, Zub E, deBock F, Boux F, Barbier E, Audinat E, Lerner-Natoli M, Marchi N. (2019) A pericyte-glia scarring develops at the leaky capillaries in the hippocampus during seizure activity. Epilepsia 60:1399–1411.
    1. Kobow K, Blümcke I. (2018) Epigenetics in epilepsy. Neurosci Lett 667:40–46.
    1. Kobow K, El-Osta A, Blümcke I. (2013) The methylation hypothesis of pharmacoresistance in epilepsy. Epilepsia 54 (Suppl 2):41–47.
    1. Koepp MJ. (2014) Neuroimaging of drug resistance in epilepsy. Curr Opin Neurol 27:192–198.
    1. Koepp MJ, Årstad E, Bankstahl JP, Dedeurwaerdere S, Friedman A, Potschka H, Ravizza T, Theodore WH, Baram TZ. (2017) Neuroinflammation imaging markers for epileptogenesis. Epilepsia 58 (Suppl 3):11–19.
    1. Koneval Z, Knox KM, White HS, Barker-Haliski M. (2018) Lamotrigine-resistant corneal-kindled mice: a model of pharmacoresistant partial epilepsy for moderate-throughput drug discovery. Epilepsia 59:1245–1256.
    1. König J, Müller F, Fromm MF. (2013) Transporters and drug-drug interactions: important determinants of drug disposition and effects. Pharmacol Rev 65:944–966.
    1. Kovács R, Heinemann U. (2014) Models in research of pharmacoresistant epilepsy: present and future in development of antiepileptic drugs. Curr Med Chem 21:689–703.
    1. Krauss GL, Klein P, Brandt C, Lee SK, Milanov I, Milovanovic M, Steinhoff BJ, Kamin M. (2020) Safety and efficacy of adjunctive cenobamate (YKP3089) in patients with uncontrolled focal seizures: a multicentre, double-blind, randomised, placebo-controlled, dose-response trial. Lancet Neurol 19:38–48.
    1. Kwan P, Arzimanoglou A, Berg AT, Brodie MJ, Allen Hauser W, Mathern G, Moshé SL, Perucca E, Wiebe S, French J. (2010) Definition of drug resistant epilepsy: consensus proposal by the ad hoc Task Force of the ILAE Commission on Therapeutic Strategies. Epilepsia 51:1069–1077.
    1. Kwan P, Schachter SC, Brodie MJ. (2011) Drug-resistant epilepsy. N Engl J Med 365:919–926.
    1. Lazarowski A, Czornyj L, Lubienieki F, Girardi E, Vazquez S, D’Giano C. (2007) ABC transporters during epilepsy and mechanisms underlying multidrug resistance in refractory epilepsy. Epilepsia 48 (Suppl 5):140–149.
    1. Leclercq K, Afrikanova T, Langlois M, De Prins A, Buenafe OE, Rospo CC, Van Eeckhaut A, de Witte PA, Crawford AD, Smolders I, et al. (2015) Cross-species pharmacological characterization of the allylglycine seizure model in mice and larval zebrafish. Epilepsy Behav 45:53–63.
    1. Leclercq K, Kaminski RM. (2015) Status epilepticus induction has prolonged effects on the efficacy of antiepileptic drugs in the 6-Hz seizure model. Epilepsy Behav 49:55–60.
    1. Leclercq K, Matagne A, Kaminski RM. (2014) Low potency and limited efficacy of antiepileptic drugs in the mouse 6 Hz corneal kindling model. Epilepsy Res 108:675–683.
    1. Leclercq K, Matagne A, Provins L, Klitgaard H, Kaminski R. (2020) Pharmacological profile of the antiepileptic drug candidate padsevonil - characterization in rodent seizure and epilepsy models. J Pharmacol Exp Ther 372:11–20.
    1. Lévesque M, Avoli M. (2013) The kainic acid model of temporal lobe epilepsy. Neurosci Biobehav Rev 37:2887–2899.
    1. Librizzi L, Noè F, Vezzani A, de Curtis M, Ravizza T. (2012) Seizure-induced brain-borne inflammation sustains seizure recurrence and blood-brain barrier damage. Ann Neurol 72:82–90.
    1. Liu JY, Thom M, Catarino CB, Martinian L, Figarella-Branger D, Bartolomei F, Koepp M, Sisodiya SM. (2012) Neuropathology of the blood-brain barrier and pharmaco-resistance in human epilepsy. Brain 135:3115–3133.
    1. Löscher W. (1997) Animal models of intractable epilepsy. Prog Neurobiol 53:239–258.
    1. Löscher W. (2002) Animal models of drug resistant epilepsy, in Mechanisms of Drug Resistance in Epilepsy: Lessons from Oncology (Ling V. 149–159, Wiley, Chichester.
    1. Löscher W. (2007) Drug transporters in the epileptic brain. Epilepsia 48 (Suppl 1):8–13.
    1. Löscher W. (2016) Fit for purpose application of currently existing animal models in the discovery of novel epilepsy therapies. Epilepsy Res 126:157–184.
    1. Löscher W. (2017a) Animal models of seizures and epilepsy: past, present, and future role for the discovery of antiseizure drugs. Neurochem Res 42:1873–1888.
    1. Löscher W. (2017b) The search for new screening models of pharmacoresistant epilepsy: is induction of acute seizures in epileptic rodents a suitable Approach? Neurochem Res 42:1926–1938.
    1. Löscher W. (2017c) Animal models of drug-refractory epilepsy, in Models of Seizures and Epilepsy (Pitkänen A, Buckmaster PS, Galanopoulou AS, Moshé SL. 743–760, Academic Press, London.
    1. Löscher W. (2011) Critical review of current animal models of seizures and epilepsy used in the discovery and development of new antiepileptic drugs. Seizure 20:359–368.
    1. Löscher W, Brandt C. (2010) High seizure frequency prior to antiepileptic treatment is a predictor of pharmacoresistant epilepsy in a rat model of temporal lobe epilepsy. Epilepsia 51:89–97.
    1. Löscher W, Friedman A. (2020) Structural, molecular and functional alterations of the blood-brain barrier during epileptogenesis and epilepsy: a cause, consequence or both? Int J Mol Sci 21:591.
    1. Löscher W, Klein P. (2020) The feast and famine: epilepsy treatment and treatment gaps in early 21st century. Neuropharma In press.
    1. Löscher W, Klitgaard H, Twyman RE, Schmidt D. (2013a) New avenues for anti-epileptic drug discovery and development. Nat Rev Drug Discov 12:757–776.
    1. Löscher W, Klotz U, Zimprich F, Schmidt D. (2009) The clinical impact of pharmacogenetics on the treatment of epilepsy. Epilepsia 50:1–23.
    1. Löscher W, Luna-Tortós C, Römermann K, Fedrowitz M. (2011) Do ATP-binding cassette transporters cause pharmacoresistance in epilepsy? Problems and approaches in determining which antiepileptic drugs are affected. Curr Pharm Des 17:2808–2828.
    1. Löscher W, Potschka H. (2002) Role of multidrug transporters in pharmacoresistance to antiepileptic drugs. J Pharmacol Exp Ther 301:7–14.
    1. Löscher W, Potschka H. (2005) Drug resistance in brain diseases and the role of drug efflux transporters. Nat Rev Neurosci 6:591–602.
    1. Löscher W, Puskarjov M, Kaila K. (2013b) Cation-chloride cotransporters NKCC1 and KCC2 as potential targets for novel antiepileptic and antiepileptogenic treatments. Neuropharmacology 69:62–74.
    1. Löscher W, Rogawski MA. (2012) How theories evolved concerning the mechanism of action of barbiturates. Epilepsia 53 (Suppl 8):12–25.
    1. Löscher W, Rundfeldt C, Hönack D. (1993) Pharmacological characterization of phenytoin-resistant amygdala-kindled rats, a new model of drug-resistant partial epilepsy. Epilepsy Res 15:207–219.
    1. Löscher W, Schmidt D. (2011) Modern antiepileptic drug development has failed to deliver: ways out of the current dilemma. Epilepsia 52:657–678.
    1. Löscher W, Schmidt D. (2006) Experimental and clinical evidence for loss of effect (tolerance) during prolonged treatment with antiepileptic drugs. Epilepsia 47:1253–1284.
    1. Löscher W, Schmidt D. (2016) Mechanisms of drug resistance and tolerance, in The Treatment of Epilepsy, 4th ed. (Shorvon S, Perucca E, Engel J. Jr eds) pp 92–102, Wiley Blackwell, Oxford.
    1. Loup F, Wieser HG, Yonekawa Y, Aguzzi A, Fritschy JM. (2000) Selective alterations in GABAA receptor subtypes in human temporal lobe epilepsy. J Neurosci 20:5401–5419.
    1. Lum GR, Olson CA, Hsiao EY. (2020) Emerging roles for the intestinal microbiome in epilepsy. Neurobiol Dis 135:104576.
    1. Lybrand Z, Goswami S, Hsieh J. (2019) Stem cells: a path towards improved epilepsy therapies. Neuropharmacology 107781.
    1. Mahler B, Carlsson S, Andersson T, Tomson T. (2018) Risk for injuries and accidents in epilepsy: a prospective population-based cohort study. Neurology 90:e779–e789.
    1. Mahringer A, Fricker G. (2016) ABC transporters at the blood-brain barrier. Expert Opin Drug Metab Toxicol 12:499–508.
    1. Mantegazza M, Broccoli V. (2019) SCN1A/NaV 1.1 channelopathies: mechanisms in expression systems, animal models, and human IPSC models. Epilepsia 60:S25–S38.
    1. Marchi N, Fan Q, Ghosh C, Fazio V, Bertolini F, Betto G, Batra A, Carlton E, Najm I, Granata T, et al. (2009) Antagonism of peripheral inflammation reduces the severity of status epilepticus. Neurobiol Dis 33:171–181.
    1. Marchi N, Guiso G, Rizzi M, Pirker S, Novak K, Czech T, Baumgartner C, Janigro D, Caccia S, Vezzani A. (2005) A pilot study on brain-to-plasma partition of 10,11-dyhydro-10-hydroxy-5H-dibenzo(b,f)azepine-5-carboxamide and MDR1 brain expression in epilepsy patients not responding to oxcarbazepine. Epilepsia 46:1613–1619.
    1. Marcon J, Gagliardi B, Balosso S, Maroso M, Noé F, Morin M, Lerner-Natoli M, Vezzani A, Ravizza T. (2009) Age-dependent vascular changes induced by status epilepticus in rat forebrain: implications for epileptogenesis. Neurobiol Dis 34:121–132.
    1. Margineanu DG, Klitgaard H. (2009) Mechanisms of drug resistance in epilepsy: relevance for antiepileptic drug discovery. Expert Opin Drug Discov 4:23–32.
    1. Maroso M, Balosso S, Ravizza T, Iori V, Wright CI, French J, Vezzani A. (2011) Interleukin-1β biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice. Neurotherapeutics 8:304–315.
    1. Matagne A, Klitgaard H. (1998) Validation of corneally kindled mice: a sensitive screening model for partial epilepsy in man. Epilepsy Res 31:59–71.
    1. Mesraoua B, Deleu D, Kullmann D, Shetty A, Boon P, Perucca E, Mikati M, Asadi-Pooya A. (2019) Novel therapies for epilepsy in the pipeline. Epilepsy Behav 97:282–290.
    1. Metcalf CS, Huff J, Thomson KE, Johnson K, Edwards SF, Wilcox KS. (2019) Evaluation of antiseizure drug efficacy and tolerability in the rat lamotrigine-resistant amygdala kindling model. Epilepsia Open 4:452–463.
    1. Metcalf CS, West PJ, Thomson KE, Edwards SF, Smith MD, White HS, Wilcox KS. (2017) Development and pharmacologic characterization of the rat 6 Hz model of partial seizures. Epilepsia 58:1073–1084.
    1. Miller-Delaney SF, Bryan K, Das S, McKiernan RC, Bray IM, Reynolds JP, Gwinn R, Stallings RL, Henshall DC. (2015) Differential DNA methylation profiles of coding and non-coding genes define hippocampal sclerosis in human temporal lobe epilepsy. Brain 138:616–631.
    1. Møller RS, Hammer TB, Rubboli G, Lemke JR, Johannesen KM. (2019) From next-generation sequencing to targeted treatment of non-acquired epilepsies. Expert Rev Mol Diagn 19:217–228.
    1. Morin-Brureau M, Lebrun A, Rousset MC, Fagni L, Bockaert J, de Bock F, Lerner-Natoli M. (2011) Epileptiform activity induces vascular remodeling and zonula occludens 1 downregulation in organotypic hippocampal cultures: role of VEGF signaling pathways. J Neurosci 31:10677–10688.
    1. Morris G, Reschke CR, Henshall DC. (2019) Targeting microRNA-134 for seizure control and disease modification in epilepsy. EBioMedicine 45:646–654.
    1. Mula M, Zaccara G, Galimberti CA, Ferrò B, Canevini MP, Mascia A, Mecarelli O, Michelucci R, Pisani LR, Specchio LM, et al. (2019) Validated outcome of treatment changes according to International League against Epilepsy criteria in adults with drug-resistant focal epilepsy. Epilepsia 60:1114–1123.
    1. Murray MI, Halpern MT, Leppik IE. (1996) Cost of refractory epilepsy in adults in the USA. Epilepsy Res 23:139–148.
    1. Namgoong JH, Bertoni C. (2016) Clinical potential of Ataluren in the treatment of Duchenne muscular dystrophy. Degener Neurol Neuromuscul 6:37–48.
    1. Narayanan J, Frech R, Walters S, Patel V, Frigerio R, Maraganore DM. (2016) Low dose verapamil as an adjunct therapy for medically refractory epilepsy - an open label pilot study. Epilepsy Res 126:197–200.
    1. National Research Council (US) Committee on A Framework for Developing a New Taxonomy of Disease (2011) Toward Precision Medicine: Building a Knowledge Network for Biomedical Research and a New Taxonomy of Disease, National Academies Press, Washington, DC.
    1. Naylor DE, Liu H, Wasterlain CG. (2005) Trafficking of GABA(A) receptors, loss of inhibition, and a mechanism for pharmacoresistance in status epilepticus. J Neurosci 25:7724–7733.
    1. Neuwelt EA, Bauer B, Fahlke C, Fricker G, Iadecola C, Janigro D, Leybaert L, Molnár Z, O’Donnell ME, Povlishock JT, et al. (2011) Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 12:169–182.
    1. Niquet J, Baldwin R, Suchomelova L, Lumley L, Naylor D, Eavey R, Wasterlain CG. (2016) Benzodiazepine-refractory status epilepticus: pathophysiology and principles of treatment. Ann N Y Acad Sci 1378:166–173.
    1. O’Brien FE, Dinan TG, Griffin BT, Cryan JF. (2012) Interactions between antidepressants and P-glycoprotein at the blood-brain barrier: clinical significance of in vitro and in vivo findings. Br J Pharmacol 165:289–312.
    1. Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY. (2018) The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell 173:1728–1741.e13.
    1. Orlandi A, Paolino MC, Striano P, Parisi P. (2018) Clinical reappraisal of the influence of drug-transporter polymorphisms in epilepsy. Expert Opin Drug Metab Toxicol 14:505–512.
    1. Parent JM, Anderson SA. (2015) Reprogramming patient-derived cells to study the epilepsies. Nat Neurosci 18:360–366.
    1. Pekcec A, Unkrüer B, Schlichtiger J, Soerensen J, Hartz AM, Bauer B, van Vliet EA, Gorter JA, Potschka H. (2009) Targeting prostaglandin E2 EP1 receptors prevents seizure-associated P-glycoprotein up-regulation. J Pharmacol Exp Ther 330:939–947.
    1. Peng A, Qiu X, Lai W, Li W, Zhang L, Zhu X, He S, Duan J, Chen L. (2018) Altered composition of the gut microbiome in patients with drug-resistant epilepsy. Epilepsy Res 147:102–107.
    1. Pirker S, Schwarzer C, Czech T, Baumgartner C, Pockberger H, Maier H, Hauer B, Sieghart W, Furtinger S, Sperk G. (2003) Increased expression of GABA(A) receptor beta-subunits in the hippocampus of patients with temporal lobe epilepsy. J Neuropathol Exp Neurol 62:820–834.
    1. Polster T. (2019) Individualized treatment approaches: fenfluramine, a novel antiepileptic medication for the treatment of seizures in Dravet syndrome. Epilepsy Behav 91:99–102.
    1. Porter BE, Zhang G, Celix J, Hsu FC, Raol YH, Telfeian A, Gallagher PR, Coulter DA, Brooks-Kayal AR. (2005) Heterogeneous GABAA receptor suxWbunit expression in pediatric epilepsy patients. Neurobiol Dis 18:484–491.
    1. Porter RJ, Kupferberg HJ. (2017) The Anticonvulsant Screening Program of the National Institute of Neurological Disorders and Stroke, NIH: history and contributions to clinical care in the twentieth century and beyond. Neurochem Res 42:1889–1893.
    1. Postma T, Krupp E, Li XL, Post RM, Weiss SR. (2000) Lamotrigine treatment during amygdala-kindled seizure development fails to inhibit seizures and diminishes subsequent anticonvulsant efficacy. Epilepsia 41:1514–1521.
    1. Potschka H. (2010) Modulating P-glycoprotein regulation: future perspectives for pharmacoresistant epilepsies? Epilepsia 51:1333–1347.
    1. Potschka H. (2012) Role of CNS efflux drug transporters in antiepileptic drug delivery: overcoming CNS efflux drug transport. Adv Drug Deliv Rev 64:943–952.
    1. Potschka H. (2013) Animal and human data: where are our concepts for drug-resistant epilepsy going? Epilepsia 54 (Suppl 2):29–32.
    1. Potschka H, Fedrowitz M, Löscher W. (2003a) Multidrug resistance protein MRP2 contributes to blood-brain barrier function and restricts antiepileptic drug activity. J Pharmacol Exp Ther 306:124–131.
    1. Potschka H, Fedrowitz M, Löscher W. (2003b) Brain access and anticonvulsant efficacy of carbamazepine, lamotrigine, and felbamate in ABCC2/MRP2-deficient TR- rats. Epilepsia 44:1479–1486.
    1. Potschka H, Fischer A, von Rüden EL, Hülsmeyer V, Baumgärtner W. (2013) Canine epilepsy as a translational model? Epilepsia 54:571–579.
    1. Potschka H, Volk HA, Löscher W. (2004) Pharmacoresistance and expression of multidrug transporter P-glycoprotein in kindled rats. Neuroreport 15:1657–1661.
    1. Pressler RM, Boylan GB, Marlow N, Blennow M, Chiron C, Cross JH, de Vries LS, Hallberg B, Hellström-Westas L, Jullien V, et al. NEonatal seizure treatment with Medication Off-patent (NEMO) consortium (2015) Bumetanide for the treatment of seizures in newborn babies with hypoxic ischaemic encephalopathy (NEMO): an open-label, dose finding, and feasibility phase 1/2 trial. Lancet Neurol 14:469–477.
    1. Pugliatti M, Beghi E, Forsgren L, Ekman M, Sobocki P. (2007) Estimating the cost of epilepsy in Europe: a review with economic modeling. Epilepsia 48:2224–2233.
    1. Puskarjov M, Kahle KT, Ruusuvuori E, Kaila K. (2014) Pharmacotherapeutic targeting of cation-chloride cotransporters in neonatal seizures. Epilepsia 55:806–818.
    1. Ravizza T, Vezzani A. (2006) Status epilepticus induces time-dependent neuronal and astrocytic expression of interleukin-1 receptor type I in the rat limbic system. Neuroscience 137:301–308.
    1. Regesta G, Tanganelli P. (1999) Clinical aspects and biological bases of drug-resistant epilepsies. Epilepsy Res 34:109–122.
    1. Remy S, Beck H. (2006) Molecular and cellular mechanisms of pharmacoresistance in epilepsy. Brain 129:18–35.
    1. Remy S, Gabriel S, Urban BW, Dietrich D, Lehmann TN, Elger CE, Heinemann U, Beck H. (2003a) A novel mechanism underlying drug resistance in chronic epilepsy. Ann Neurol 53:469–479.
    1. Remy S, Urban BW, Elger CE, Beck H. (2003b) Anticonvulsant pharmacology of voltage-gated Na+ channels in hippocampal neurons of control and chronically epileptic rats. Eur J Neurosci 17:2648–2658.
    1. Riban V, Bouilleret V, Pham-Lê BT, Fritschy JM, Marescaux C, Depaulis A. (2002) Evolution of hippocampal epileptic activity during the development of hippocampal sclerosis in a mouse model of temporal lobe epilepsy. Neuroscience 112:101–111.
    1. Richards K, Milligan C, Richardson R, Jancovski N, Grunnet M, Jacobson L, Undheim E, Mobli M, Chow C, Herzig V, et al. (2018) Selective NaV1.1 activation rescues Dravet syndrome mice from seizures and premature death. Proc Natl Acad Sci USA 115:E8077–E8085.
    1. Richter S, Garner J, Wurbel H. (2009) Environmental standardization: cure or cause of poor reproducibility in animal experiments? Nat Methods 6:257–261.
    1. Rigau V, Morin M, Rousset MC, de Bock F, Lebrun A, Coubes P, Picot MC, Baldy-Moulinier M, Bockaert J, Crespel A, et al. (2007) Angiogenesis is associated with blood-brain barrier permeability in temporal lobe epilepsy. Brain 130:1942–1956.
    1. Rizzi M, Caccia S, Guiso G, Richichi C, Gorter JA, Aronica E, Aliprandi M, Bagnati R, Fanelli R, D’Incalci M, et al. (2002) Limbic seizures induce P-glycoprotein in rodent brain: functional implications for pharmacoresistance. J Neurosci 22:5833–5839.
    1. Rogawski MA. (2013) The intrinsic severity hypothesis of pharmacoresistance to antiepileptic drugs. Epilepsia 54 (Suppl 2):33–40.
    1. Rogawski MA, Johnson MR. (2008) Intrinsic severity as a determinant of antiepileptic drug refractoriness. Epilepsy Curr 8:127–130.
    1. Rogawski MA, Löscher W. (2004) The neurobiology of antiepileptic drugs. Nat Rev Neurosci 5:553–564.
    1. Rogawski MA, Löscher W, Rho JM. (2016) Mechanisms of action of antiseizure drugs and the ketogenic diet. Cold Spring Harb Perspect Med 6.
    1. Rojas A, Gueorguieva P, Lelutiu N, Quan Y, Shaw R, Dingledine R. (2014) The prostaglandin EP1 receptor potentiates kainate receptor activation via a protein kinase C pathway and exacerbates status epilepticus. Neurobiol Dis 70:74–89.
    1. Römermann K, Helmer R, Löscher W. (2015) The antiepileptic drug lamotrigine is a substrate of mouse and human breast cancer resistance protein (ABCG2). Neuropharmacology 93:7–14.
    1. Roseti C, Fucile S, Lauro C, Martinello K, Bertollini C, Esposito V, Mascia A, Catalano M, Aronica E, Limatola C, et al. (2013) Fractalkine/CX3CL1 modulates GABAA currents in human temporal lobe epilepsy. Epilepsia 54:1834–1844.
    1. Roseti C, van Vliet EA, Cifelli P, Ruffolo G, Baayen JC, Di Castro MA, Bertollini C, Limatola C, Aronica E, Vezzani A, et al. (2015) GABAA currents are decreased by IL-1β in epileptogenic tissue of patients with temporal lobe epilepsy: implications for ictogenesis. Neurobiol Dis 82:311–320.
    1. Rowley N, White H. (2010) Comparative anticonvulsant efficacy in the corneal kindled mouse model of partial epilepsy: correlation with other seizure and epilepsy models. Epilepsy Res 92:163–169.
    1. Rundfeldt C, Löscher W. (2014) The pharmacology of imepitoin: the first partial benzodiazepine receptor agonist developed for the treatment of epilepsy. Cns Drugs 28:29–43.
    1. Ryvlin P, Rheims S, Lhatoo SD. (2019) Risks and predictive biomarkers of sudden unexpected death in epilepsy patient. Curr Opin Neurol 32:205–212.
    1. Sa M, Singh R, Pujar S, D’Arco F, Desai N, Eltze C, Hughes E, Al Obaidi M, Eleftheriou D, Tisdall M, et al. (2019) Centromedian thalamic nuclei deep brain stimulation and anakinra treatment for FIRES - two different outcomes. Eur J Paediatr Neurol 23:749–754.
    1. Salar S, Maslarova A, Lippmann K, Nichtweiss J, Weissberg I, Sheintuch L, Kunz WS, Shorer Z, Friedman A, Heinemann U. (2014) Blood-brain barrier dysfunction can contribute to pharmacoresistance of seizures. Epilepsia 55:1255–1263.
    1. Sandoval KE, Witt KA. (2008) Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis 32:200–219.
    1. Saunders NR, Habgood MD, Møllgård K, Dziegielewska KM. (2016) The biological significance of brain barrier mechanisms: help or hindrance in drug delivery to the central nervous system? F1000 Res 5.
    1. Schaub C, Uebachs M, Beck H. (2007) Diminished response of CA1 neurons to antiepileptic drugs in chronic epilepsy. Epilepsia 48:1339–1350.
    1. Schlichtiger J, Pekcec A, Bartmann H, Winter P, Fuest C, Soerensen J, Potschka H. (2010) Celecoxib treatment restores pharmacosensitivity in a rat model of pharmacoresistant epilepsy. Br J Pharmacol 160:1062–1071.
    1. Schmidt D, Löscher W. (2005) Drug resistance in epilepsy: putative neurobiologic and clinical mechanisms. Epilepsia 46:858–877.
    1. Schmidt D, Löscher W. (2009) New developments in antiepileptic drug resistance: an integrative view. Epilepsy Curr 9:47–52.
    1. Siddiqui A, Kerb R, Weale ME, Brinkmann U, Smith A, Goldstein DB, Wood NW, Sisodiya SM. (2003) Association of multidrug resistance in epilepsy with a polymorphism in the drug-transporter gene ABCB1. N Engl J Med 348:1442–1448.
    1. Sillanpää M, Schmidt D. (2009) Early seizure frequency and aetiology predict long-term medical outcome in childhood-onset epilepsy. Brain 132:989–998.
    1. Sisodiya SM, Heffernan J, Squier MV. (1999) Over-expression of P-glycoprotein in malformations of cortical development. Neuroreport 10:3437–3441.
    1. Sisodiya SM, Lin WR, Harding BN, Squier MV, Thom M. (2002) Drug resistance in epilepsy: expression of drug resistance proteins in common causes of refractory epilepsy. Brain 125:22–31.
    1. Sills G, Rogawski M. (2020) Mechanisms of action of currently used antiseizure drugs. Neuropharmacology In press.
    1. Sisodiya SM. (2020) Epilepsy genetics and the precision medicine matrix. Lancet Neurol 19:29–30.
    1. Sisodiya SM. (2003) Mechanisms of antiepileptic drug resistance. Curr Opin Neurol 16:197–201.
    1. Smyth MD, Barbaro NM, Baraban SC. (2002) Effects of antiepileptic drugs on induced epileptiform activity in a rat model of dysplasia. Epilepsy Res 50:251–264.
    1. Srivastava AK, Alex AB, Wilcox KS, White HS. (2013) Rapid loss of efficacy to the antiseizure drugs lamotrigine and carbamazepine: a novel experimental model of pharmacoresistant epilepsy. Epilepsia 54:1186–1194.
    1. Srivastava P, van Eyll J, Godard P, Mazzuferi M, Delahaye-Duriez A, Van Steenwinckel J, Gressens P, Danis B, Vandenplas C, Foerch P, et al. (2018) A systems-level framework for drug discovery identifies Csf1R as an anti-epileptic drug target. Nat Commun 9:3561.
    1. Sun G, Sun X, Guan L. (2014) Association of MDR1 gene C3435T polymorphism with childhood intractable epilepsy: a meta-analysis. J Neural Transm (Vienna) 121:717–724.
    1. Swissa E, Serlin Y, Vazana U, Prager O, Friedman A. (2019) Blood-brain barrier dysfunction in status epileptics: Mechanisms and role in epileptogenesis. Epilepsy Behav 101:106285.
    1. Tang F, Hartz AMS, Bauer B. (2017) Drug-resistant epilepsy: multiple hypotheses, few answers. Front Neurol 8:301.
    1. Terrone G, Salamone A, Vezzani A. (2017) Inflammation and epilepsy: preclinical findings and potential clinical translation. Curr Pharm Des 23:5569–5576.
    1. Tishler DM, Weinberg KI, Hinton DR, Barbaro N, Annett GM, Raffel C. (1995) MDR1 gene expression in brain of patients with medically intractable epilepsy. Epilepsia 36:1–6.
    1. Töllner K, Wolf S, Löscher W, Gernert M. (2011) The anticonvulsant response to valproate in kindled rats is correlated with its effect on neuronal firing in the substantia nigra pars reticulata: a new mechanism of pharmacoresistance. J Neurosci 31:16423–16434.
    1. Uchida Y, Ohtsuki S, Katsukura Y, Ikeda C, Suzuki T, Kamiie J, Terasaki T. (2011) Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. J Neurochem 117:333–345.
    1. van Vliet EA, Aronica E, Gorter JA. (2015) Blood-brain barrier dysfunction, seizures and epilepsy. Semin Cell Dev Biol 38:26–34.
    1. van Vliet EA, Aronica E, Vezzani A, Ravizza T. (2018) Review: neuroinflammatory pathways as treatment targets and biomarker candidates in epilepsy: emerging evidence from preclinical and clinical studies. Neuropathol Appl Neurobiol 44:91–111.
    1. van Vliet EA, van Schaik R, Edelbroek PM, Voskuyl RA, Redeker S, Aronica E, Wadman WJ, Gorter JA. (2007) Region-specific overexpression of P-glycoprotein at the blood-brain barrier affects brain uptake of phenytoin in epileptic rats. J Pharmacol Exp Ther 322:141–147.
    1. van Vliet EA, Zibell G, Pekcec A, Schlichtiger J, Edelbroek PM, Holtman L, Aronica E, Gorter JA, Potschka H. (2010) COX-2 inhibition controls P-glycoprotein expression and promotes brain delivery of phenytoin in chronic epileptic rats. Neuropharmacology 58:404–412.
    1. Vezzani A, Balosso S, Ravizza T. (2019) Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat Rev Neurol 15:459–472.
    1. Viviani B, Bartesaghi S, Gardoni F, Vezzani A, Behrens MM, Bartfai T, Binaglia M, Corsini E, Di Luca M, Galli CL, et al. (2003) Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases. J Neurosci 23:8692–8700.
    1. Vezzani A, Conti M, De Luigi A, Ravizza T, Moneta D, Marchesi F, De Simoni MG. (1999) Interleukin-1beta immunoreactivity and microglia are enhanced in the rat hippocampus by focal kainate application: functional evidence for enhancement of electrographic seizures. J Neurosci 19:5054–5065.
    1. Vezzani A, Maroso M, Balosso S, Sanchez MA, Bartfai T. (2011) IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures. Brain Behav Immun 25:1281–1289.
    1. Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A, De Simoni MG, Sperk G, Andell-Jonsson S, Lundkvist J, et al. (2000) Powerful anticonvulsant action of IL-1 receptor antagonist on intracerebral injection and astrocytic overexpression in mice. Proc Natl Acad Sci USA 97:11534–11539.
    1. Vezzani A, Viviani B. (2015) Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology 96 (Pt A):70–82.
    1. Voelkl B, Vogt L, Sena E, Wurbel H. (2018) Reproducibility of preclinical animal research improves with heterogeneity of study samples. PLoS Biol 16:e2003693.
    1. Volk HA, Arabadzisz D, Fritschy JM, Brandt C, Bethmann K, Löscher W. (2006) Antiepileptic drug-resistant rats differ from drug-responsive rats in hippocampal neurodegeneration and GABA(A) receptor ligand binding in a model of temporal lobe epilepsy. Neurobiol Dis 21:633–646.
    1. Volk HA, Löscher W. (2005) Multidrug resistance in epilepsy: rats with drug-resistant seizures exhibit enhanced brain expression of P-glycoprotein compared with rats with drug-responsive seizures. Brain 128:1358–1368.
    1. Wahab A, Albus K, Gabriel S, Heinemann U. (2010) In search of models of pharmacoresistant epilepsy. Epilepsia 51 (Suppl 3):154–159.
    1. Wang J, Lin Z, Liu L, Xu H, Shi Y, Yi Y, He N, Liao W. (2017) Epilepsy-associated genes. Seizure 44:11–20.
    1. Watila MM, Balarabe SA, Ojo O, Keezer MR, Sander JW. (2018) Overall and cause-specific premature mortality in epilepsy: a systematic review. Epilepsy Behav 87:213–225.
    1. Weidner LD, Kannan P, Mitsios N, Kang SJ, Hall MD, Theodore WH, Innis RB, Mulder J. (2018) The expression of inflammatory markers and their potential influence on efflux transporters in drug-resistant mesial temporal lobe epilepsy tissue. Epilepsia 59:1507–1517.
    1. Weissberg I, Wood L, Kamintsky L, Vazquez O, Milikovsky DZ, Alexander A, Oppenheim H, Ardizzone C, Becker A, Frigerio F, et al. (2015) Albumin induces excitatory synaptogenesis through astrocytic TGF-β/ALK5 signaling in a model of acquired epilepsy following blood-brain barrier dysfunction. Neurobiol Dis 78:115–125.
    1. Wengert ER, Saga AU, Panchal PS, Barker BS, Patel MK. (2019) Prax330 reduces persistent and resurgent sodium channel currents and neuronal hyperexcitability of subiculum neurons in a mouse model of SCN8A epileptic encephalopathy. Neuropharmacology 158:107699.
    1. West PJ, Saunders GW, Billingsley P, Smith MD, White HS, Metcalf CS, Wilcox KS. (2018) Recurrent epileptiform discharges in the medial entorhinal cortex of kainate-treated rats are differentially sensitive to antiseizure drugs. Epilepsia 59:2035–2048.
    1. Wiebe S, Jette N. (2012) Pharmacoresistance and the role of surgery in difficult to treat epilepsy. Nat Rev Neurol 8:669–677.
    1. Wilcox K, West P, Metcalf C. (2020) The current approach of the epilepsy therapy screening program cntract site for identifying improved therapies for the treatment of pharmacoresistant seizures in epilepsy. Neuropharmacology In press.
    1. Wood M, Daniels V, Provins L, Wolff C, Kaminski R, Gillard M. (2020) Pharmacological profile of the novel antiepileptic drug candidate Padsevonil: interactions with synaptic vesicle 2 proteins and the GABAA receptor. J Pharmacol Exp Ther 372:1–10.
    1. Xu ZH, Wang Y, Tao AF, Yu J, Wang XY, Zu YY, Zhang SH, Chen Z. (2016) Interleukin-1 receptor is a target for adjunctive control of diazepam-refractory status epilepticus in mice. Neuroscience 328:22–29.
    1. Yang GY, Gong C, Qin Z, Liu XH, Lorris Betz A. (1999) Tumor necrosis factor alpha expression produces increased blood-brain barrier permeability following temporary focal cerebral ischemia in mice. Brain Res Mol Brain Res 69:135–143.
    1. Zaccara G, Mula M, Ferrò B, Consoli D, Elia M, Giallonardo AT, Iudice A, La Neve A, Meletti S, Tinuper P, et al. (2019) Do neurologists agree in diagnosing drug resistance in adults with focal epilepsy? Epilepsia 60:175–183.
    1. Zhang C, Kwan P, Zuo Z, Baum L. (2012) The transport of antiepileptic drugs by P-glycoprotein. Adv Drug Deliv Rev 64:930–942.
    1. Zibell G, Unkrüer B, Pekcec A, Hartz AM, Bauer B, Miller DS, Potschka H. (2009) Prevention of seizure-induced up-regulation of endothelial P-glycoprotein by COX-2 inhibition. Neuropharmacology 56:849–855.
    1. Ziobro J, Eschbach K, Sullivan JE, Knupp KG. (2018) Current treatment strategies and future treatment options for Dravet syndrome. Curr Treat Options Neurol 20:52.

Source: PubMed

Подписаться