Skip to main content
Log in

The Search for New Screening Models of Pharmacoresistant Epilepsy: Is Induction of Acute Seizures in Epileptic Rodents a Suitable Approach?

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Epilepsy, a prevalent neurological disease characterized by spontaneous recurrent seizures (SRS), is often refractory to treatment with anti-seizure drugs (ASDs), so that more effective ASDs are urgently needed. For this purpose, it would be important to develop, validate, and implement new animal models of pharmacoresistant epilepsy into drug discovery. Several chronic animal models with difficult-to-treat SRS do exist; however, most of these models are not suited for drug screening, because drug testing on SRS necessitates laborious video-EEG seizure monitoring. More recently, it was proposed that, instead of monitoring SRS, chemical or electrical induction of acute seizures in epileptic rodents may be used as a surrogate for testing the efficacy of novel ASDs against refractory SRS. Indeed, several ASDs were shown to lose their efficacy on acute seizures, when such seizures were induced by pentylenetetrazole (PTZ) in epileptic rather than nonepileptic rats, whereas this was not observed when using the maximal electroshock seizure test. Subsequent studies confirmed the loss of anti-seizure efficacy of valproate against PTZ-induced seizures in epileptic mice, but several other ASDs were more potent against PTZ in epileptic than nonepileptic mice. This was also observed when using the 6-Hz model of partial seizures in epileptic mice, in which the potency of levetiracetam, in particular, was markedly increased compared to nonepileptic animals. Overall, these observations suggest that performing acute seizure tests in epileptic rodents provides valuable information on the pharmacological profile of ASDs, in particular those with mechanisms inherent to disease-induced brain alterations. However, it appears that further work is needed to define optimal approaches for acute seizure induction and generation of epileptic/drug refractory animals that would permit reliable screening of new ASDs with improved potential to provide seizure control in patients with pharmacoresistant epilepsy.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Abbreviations

AED:

Antiepileptic drug

ASD:

Anti-seizure drug

ASP:

Anticonvulsant Screening Program

BUM5:

N,N-dimethylaminoethylester of bumetanide

CC:

Convulsant current

ED50 :

Median effective dose

EEG:

Electroencephalogram

ETSP:

Epilepsy Therapy Screening Program

GAERS:

Genetic absence epilepsy rats from strasbourg

GEPR:

Genetically epilepsy-prone rat

MES:

Maximal electroshock seizure

MEST:

MES threshold

NINDS:

National Institute of Neurological Disorders and Stroke

NKCC:

N-K-2Cl cotransporter

PTZ:

Pentylenetetrazole

SE:

Status epilepticus

SRS:

Spontaneous recurrent seizures

SV2A:

Synaptic vesicle protein 2A

TLE:

Temporal lobe epilepsy

References

  1. Krall RL, Penry JK, Kupferberg HJ, Swinyard EA (1978) Antiepileptic drug development: I. History and a program for progress. Epilepsia 19:393–408

    Article  CAS  PubMed  Google Scholar 

  2. Krall RL, Penry JK, White BG, Kupferberg HJ, Swinyard EA (1978) Antiepileptic drug development: II. Anticonvulsant drug screening. Epilepsia 19:409–428

    Article  CAS  PubMed  Google Scholar 

  3. Gladding GD, Kupferberg HJ, Swinyard EA (1985) Antiepileptic drug development program. In: Frey H-H, Janz D (eds) Antiepileptic drugs. Springer, Berlin, pp 341–350

    Google Scholar 

  4. White HS, Wolf HH, Woodhead JH, Kupferberg HJ (1998) The National Institutes of Health Anticonvulsant Drug Development Program: screening for efficacy. Adv Neurol 76:29–39

    CAS  PubMed  Google Scholar 

  5. Wilcox KS, Dixon-Salazar T, Sills GJ, Ben Menachem E, White HS, Porter RJ, Dichter MA, Moshe SL, Noebels JL, Privitera MD, Rogawski MA (2013) Issues related to development of new antiseizure treatments. Epilepsia 54(Suppl 4):24–34

    Article  PubMed  PubMed Central  Google Scholar 

  6. French JA, White HS, Klitgaard H, Holmes GL, Privitera MD, Cole AJ, Quay E, Wiebe S, Schmidt D, Porter RJ, Arzimanoglou A, Trinka E, Perucca E (2013) Development of new treatment approaches for epilepsy: unmet needs and opportunities. Epilepsia 54(Suppl 4):3–12

    Article  CAS  PubMed  Google Scholar 

  7. Löscher W, Schmidt D (2011) Modern antiepileptic drug development has failed to deliver: ways out of the current dilemma. Epilepsia 52:657–678

    Article  PubMed  Google Scholar 

  8. Löscher W (2016) Fit for purpose application of currently existing animal models in the discovery of novel epilepsy therapies. Epilepsy Res. doi:10.1016/j.eplepsyres.2016.05.016

    Google Scholar 

  9. 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

    Article  PubMed  Google Scholar 

  10. White HS, Smith-Yockman M, Srivastava A, Wilcox KS (2006) Therapeutic assays for the identification and characterization of antiepileptic and antiepileptogenic drugs. In: Pitkänen A, Schwartzkroin PA, Moshé SL (eds) Models of seizures and epilepsy. Elsevier, Amsterdam, pp 539–549

    Chapter  Google Scholar 

  11. Blanco MM, Dos SJ 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

    Article  CAS  PubMed  Google Scholar 

  12. Leite JP, Garcia-Cairasco N, Cavalheiro EA (2002) New insights from the use of pilocarpine and kainate models. Epilepsy Res 50:93–103

    Article  CAS  PubMed  Google Scholar 

  13. Curia G, Longo D, Biagini G, Jones RS, Avoli M (2008) The pilocarpine model of temporal lobe epilepsy. J Neurosci Methods 172:143–157

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Levesque M, Avoli M, Bernard C (2016) Animal models of temporal lobe epilepsy following systemic chemoconvulsant administration. J Neurosci Methods 260:45–52

    Article  PubMed  Google Scholar 

  15. Leite JP, Cavalheiro EA (1995) Effects of conventional antiepileptic drugs in a model of spontaneous recurrent seizures in rats. Epilepsy Res 20:93–104

    Article  CAS  PubMed  Google Scholar 

  16. Glien M, Brandt C, Potschka H, Löscher W (2002) Effects of the novel antiepileptic drug levetiracetam on spontaneous recurrent seizures in the rat pilocarpine model of temporal lobe epilepsy. Epilepsia 43:350–357

    Article  CAS  PubMed  Google Scholar 

  17. 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

    Article  CAS  PubMed  Google Scholar 

  18. Stables JP, Bertram E, Dudek FE, Holmes G, Mathern G, Pitkänen A, White HS (2003) Therapy discovery for pharmacoresistant epilepsy and for disease-modifying therapeutics: summary of the NIH/NINDS/AES models II workshop. Epilepsia 44:1472–1478

    Article  PubMed  Google Scholar 

  19. 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

    Article  CAS  PubMed  Google Scholar 

  20. 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

    Article  CAS  PubMed  Google Scholar 

  21. White HS (1997) Clinical significance of animal seizure models and mechanism of action studies of potential antiepileptic drugs. Epilepsia 38:S9–S17

    Article  CAS  PubMed  Google Scholar 

  22. Löscher W, Schmidt D (1988) Which animal models should be used in the search for new antiepileptic drugs? A proposal based on experimental and clinical considerations. Epilepsy Res 2:145–181

    Article  PubMed  Google Scholar 

  23. Töllner K, Twele F, Löscher W (2016) Evaluation of the pentylenetetrazole seizure threshold test in epileptic mice as surrogate model for drug testing against pharmacoresistant seizures. Epilepsy Behav 57:95–104

    Article  PubMed  Google Scholar 

  24. 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

    Article  CAS  PubMed  Google Scholar 

  25. Erker T, Brandt C, Töllner K, Schreppel P, Twele F, Schidlitzki A, Löscher W (2016) The bumetanide prodrug BUM5, but not bumetanide, potentiates the anti-seizure effect of phenobarbital in adult epileptic mice. Epilepsia 57:698–705

    Article  CAS  PubMed  Google Scholar 

  26. Kaila K, Price TJ, Payne JA, Puskarjov M, Voipio J (2014) Cation-chloride cotransporters in neuronal development, plasticity and disease. Nat Rev Neurosci 15:637–654

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li X, Zhou J, Chen Z, Chen S, Zhu F, Zhou L (2008) Long-term expressional changes of Na+ -K+ -Cl- co-transporter 1 (NKCC1) and K+ -Cl- co-transporter 2 (KCC2) in CA1 region of hippocampus following lithium-pilocarpine induced status epilepticus (PISE). Brain Res 1221:141–146

    Article  CAS  PubMed  Google Scholar 

  28. Cleary RT, Sun H, Huynh T, Manning SM, Li Y, Rotenberg A, Talos DM, Kahle KT, Jackson M, Rakhade SN, Berry G, Jensen FE (2013) Bumetanide enhances phenobarbital efficacy in a rat model of hypoxic neonatal seizures. PLoS One 8:e57148

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Töllner K, Brandt C, Töpfer M, Brunhofer G, Erker T, Gabriel M, Feit PW, Lindfors J, Kaila K, Löscher W (2014) A novel prodrug-based strategy to increase effects of bumetanide in epilepsy. Ann Neurol 75:550–562

    Article  PubMed  Google Scholar 

  30. Brandt C, Nozadze M, Heuchert N, Rattka M, Löscher W (2010) Disease-modifying effects of phenobarbital and the NKCC1 inhibitor bumetanide in the pilocarpine model of temporal lobe epilepsy. J Neurosci 30:8602–8612

    Article  CAS  PubMed  Google Scholar 

  31. Löscher W, Hönack D (1991) Responses to NMDA receptor antagonists altered by epileptogenesis. Trends Pharmacol Sci 12:52

    Article  PubMed  Google Scholar 

  32. Hönack D, Löscher W (1995) Kindling increases the sensitivity of rats to adverse effects of certain antiepileptic drugs. Epilepsia 36:763–771

    Article  PubMed  Google Scholar 

  33. Löscher W, Schmidt D (1994) Strategies in antiepileptic drug development: is rational drug design superior to random screening and structural variation? Epilepsy Res 17:95–134

    Article  PubMed  Google Scholar 

  34. Klitgaard H, Matagne A, Lamberty Y (2002) Use of epileptic animals for adverse effect testing. Epilepsy Res 50:55–65

    Article  CAS  PubMed  Google Scholar 

  35. Löscher W, Klitgaard H, Twyman RE, Schmidt D (2013) New avenues for antiepileptic drug discovery and development. Nat Rev Drug Discov 12:757–776

    Article  PubMed  Google Scholar 

  36. Toman JEP (1951) Neuropharmacologic Considerations in Psychic Seizures. Neurology 1:444–460

    Article  CAS  PubMed  Google Scholar 

  37. Brown WC, Schiffman DO, Swinyard EA, Goodman LS (1953) Comparative assay of antiepileptic drugs by “pychomotor” seizure test and minimal electroshock threshold test. J Pharmacol Exp Ther 107:273–283

    CAS  PubMed  Google Scholar 

  38. Barton ME, Klein BD, Wolf HH, White HS (2001) Pharmacological characterization of the 6 Hz psychomotor seizure model of partial epilepsy. Epilepsy Res 47:217–228

    Article  CAS  PubMed  Google Scholar 

  39. Leclercq K, Kaminski RM (2015) Genetic background of mice strongly influences treatment resistance in the 6 Hz seizure model. Epilepsia 56:310–318

    Article  CAS  PubMed  Google Scholar 

  40. 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

    Article  PubMed  Google Scholar 

  41. Deutsch SI, Mastropaolo J, Riggs RL, Rosse RB (1997) The antiseizure efficacies of MK-801, phencyclidine, ketamine, and memantine are altered selectively by stress. Pharmacol Biochem Behav 58:709–712

    Article  CAS  PubMed  Google Scholar 

  42. Reddy DS, Rogawski MA (2002) Stress-induced deoxycorticosterone-derived neurosteroids modulate GABA(A) receptor function and seizure susceptibility. J Neurosci 22:3795–3805

    CAS  PubMed  Google Scholar 

  43. Joels M (2009) Stress, the hippocampus, and epilepsy. Epilepsia 50:586–597

    Article  PubMed  Google Scholar 

  44. Riban V, Bouilleret V, Pham L, 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

    Article  CAS  PubMed  Google Scholar 

  45. 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

    Article  CAS  PubMed  Google Scholar 

  46. 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

    Article  CAS  PubMed  Google Scholar 

  47. Pontes JCC, Lima TZ, Queiroz CM, Cinini SM, Blanco MM, Mello LE (2016) Seizures triggered by pentylenetetrazol in marmosets made chronically epileptic with pilocarpine show greater refractoriness to treatment. Epilepsy Res 126:16-25

    Article  CAS  PubMed  Google Scholar 

  48. Perez-Mendes P, Blanco MM, Calcagnotto ME, Cinini SM, Bachiega J, Papoti D, Covolan L, Tannus A, Mello LE (2011) Modeling epileptogenesis and temporal lobe epilepsy in a non-human primate. Epilepsy Res 96:45–57

    Article  CAS  PubMed  Google Scholar 

  49. Smith M, Wilcox KS, White HS (2007) Discovery of antiepileptic drugs. Neurother 4:12–17

    Article  CAS  Google Scholar 

  50. Czuczwar SJ, Turski L, Turski W, Kleinrok Z (1981) Effects of some antiepileptic drugs in pentetrazol-induced convulsions in mice lesioned with kainic acid. Epilepsia 22:407–414

    Article  CAS  PubMed  Google Scholar 

  51. Czuczwar SJ, Turski L, Kleinrok Z (1982) Anticonvulsant action of phenobarbital, diazepam, carbamazepine, and diphenylhydantoin in the electroshock test in mice after lesion of hippocampal pyramidal cells with intracerebroventricular kainic acid. Epilepsia 23:377–382

    Article  CAS  PubMed  Google Scholar 

  52. Goddard GV, McIntyre DC, Leech CK (1969) A permanent change in brain function resulting from daily electrical stimulation. Exp Neurol 25:295–330

    Article  CAS  PubMed  Google Scholar 

  53. Sato M, Racine RJ, McIntyre DC (1990) Kindling: basic mechanisms and clinical validity. Electroenceph Clin Neurophysiol 76:459–472

    Article  CAS  PubMed  Google Scholar 

  54. Löscher W (1997) Animal models of intractable epilepsy. Prog Neurobiol 53:239–258

    Article  PubMed  Google Scholar 

  55. Löscher W, Jäckel R, Czuczwar SJ (1986) Is amygdala kindling in rats a model for drug-resistant partial epilepsy? Exp Neurol 93:211–226

    Article  PubMed  Google Scholar 

  56. Löscher W, Hönack D (1993) Profile of ucb L059, a novel anticonvulsant drug, in models of partial and generalized epilepsy in mice and rats. Eur J Pharmacol 232:147–158

    Article  PubMed  Google Scholar 

  57. Klitgaard, H., and Verdru, P. 2007. Levetiracetam: the first SV2A ligand for the treatment of epilepsy. Expert Opin Drug Discov 2:1537–1545.

    Article  CAS  PubMed  Google Scholar 

  58. Klitgaard H (2001) Levetiracetam: the preclinical profile of a new class of antiepileptic drugs? Epilepsia 42(Suppl 4):13–18

    Article  PubMed  Google Scholar 

  59. Löscher W (2002) Animal models of epilepsy for the development of antiepileptogenic and disease-modifying drugs. A comparison of the pharmacology of kindling and post-status epilepticus models of temporal lobe epilepsy. Epilepsy Res 50:105–123

    Article  PubMed  Google Scholar 

  60. White HS, Wolf HH, Swinyard EA, Skeen GA, Sofia RD (1992) A neuropharmacological evaluation of felbamate as a novel anticonvulsant. Epilepsia 33:564–572

    Article  CAS  PubMed  Google Scholar 

  61. Matagne A, Klitgaard H (1998) Validation of corneally kindled mice: a sensitive screening model for partial epilepsy in man. Epilepsy Res 31:59–71

    Article  CAS  PubMed  Google Scholar 

  62. Potschka H, Löscher W (1999) Corneal kindling in mice: behavioral and pharmacological differences to conventional kindling. Epilepsy Res 37:109–120

    Article  CAS  PubMed  Google Scholar 

  63. Rowley NM, White HS (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

    Article  CAS  PubMed  Google Scholar 

  64. 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

    Article  CAS  PubMed  Google Scholar 

  65. Jobe PC, Mishra PK, Ludvig N, Dailey JW (1991) Scope and contribution of genetic models to an understanding of the epilepsies. Crit Rev Neurobiol 6:183–220

    CAS  PubMed  Google Scholar 

  66. Löscher W (1984) Genetic animal models of epilepsy as a unique resource for the evaluation of anticonvulsant drugs. A review. Methods Find Exp Clin Pharmacol 6:531–547

    PubMed  Google Scholar 

  67. Löscher W, Meldrum BS (1984) Evaluation of anticonvulsant drugs in genetic animal models of epilepsy. Fed Proc 43:276–284

    PubMed  Google Scholar 

  68. Naquet RG, Valin A (1998) Experimental models of reflex epilepsy. Adv Neurol 75:15–28

    CAS  PubMed  Google Scholar 

  69. Seyfried TN, Todorova MT, Poderycki MJ (1999) Experimental models of multifactorial epilepsies: the EL mouse and mice susceptible to audiogenic seizures. Adv Neurol 79:279–290

    CAS  PubMed  Google Scholar 

  70. De Sarro G, Russo E, Citraro R, Meldrum BS (2015) Genetically epilepsy-prone rats (GEPRs) and DBA/2 mice: two animal models of audiogenic reflex epilepsy for the evaluation of new generation AEDs. Epilepsy Behav. doi:10.1016/j.yebeh.2015.06.030

    PubMed  Google Scholar 

  71. Chapman AG, Croucher MJ, Meldrum BS (1984) Evaluation of anticonvulsant drugs in DBA/2 mice with sound-induced seizures. Arzneimittelforschung 34:1261–1270

    CAS  PubMed  Google Scholar 

  72. Ferraro TN, Golden GT, Snyder R, Laibinis M, Smith GG, Buono RJ, Berrettini WH (1998) Genetic influences on electrical seizure threshold. Brain Res 813:207–210

    Article  CAS  PubMed  Google Scholar 

  73. Chaix Y, Ferraro TN, Lapouble E, Martin B (2007) Chemoconvulsant-induced seizure susceptibility: toward a common genetic basis? Epilepsia 48(Suppl 5):48–52

    Article  PubMed  Google Scholar 

  74. Dailey JW, Jobe PC (1985) Anticonvulsant drugs and the genetically epilepsy-prone rat. Fed Proc 44:2640–2644

    CAS  PubMed  Google Scholar 

  75. Striano S, Coppola A, del Gaudio L, Striano P (2012) Reflex seizures and reflex epilepsies: old models for understanding mechanisms of epileptogenesis. Epilepsy Res 100:1–11

    Article  PubMed  Google Scholar 

  76. Depaulis A, David O, Charpier S (2016) The genetic absence epilepsy rat from Strasbourg as a model to decipher the neuronal and network mechanisms of generalized idiopathic epilepsies. J Neurosci Methods 260:159–174

    Article  PubMed  Google Scholar 

  77. Brailowsky S, Montiel T, Boehrer A, Marescaux C, Vergnes M (1999) Susceptibility to focal and generalized seizures in Wistar rats with genetic absence-like epilepsy. Neuroscience 93:1173–1177

    Article  CAS  PubMed  Google Scholar 

  78. Eskazan E, Onat FY, Aker R, Oner G (2002) Resistance to propagation of amygdaloid kindling seizures in rats with genetic absence epilepsy. Epilepsia 43:1115–1119

    Article  PubMed  Google Scholar 

  79. Frankel WN (2009) Genetics of complex neurological disease: challenges and opportunities for modeling epilepsy in mice and rats. Trends Genet 25:361–367

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Otto JF, Singh NA, Dahle EJ, Leppert MF, Pappas CM, Pruess TH, Wilcox KS, White HS (2009) Electroconvulsive seizure thresholds and kindling acquisition rates are altered in mouse models of human KCNQ2 and KCNQ3 mutations for benign familial neonatal convulsions. Epilepsia 50:1752–1759

    Article  CAS  PubMed  Google Scholar 

  81. Baraban SC, Löscher W (2014) What new modeling approaches will help us identify promising drug treatments? Adv Exp Med Biol 813:283–294

    Article  PubMed  Google Scholar 

  82. Grone BP, Baraban SC (2015) Animal models in epilepsy research: legacies and new directions. Nat Neurosci 18:339–343

    Article  CAS  PubMed  Google Scholar 

  83. Bialer, M., White, H.S (2010) Key factors in the discovery and development of new antiepileptic drugs. Nat Rev Drug Discov 9:68–82

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The author thanks Michael A. Rogawski, Henrik Klitgaard, Graeme Sills, Stanislaw Jerzy Czuczwar, Luiz E. Mello, Kathrin Töllner, Claudia Brandt, Marion Bankstahl, and Manuela Gernert for excellent comments on previous versions of the manuscript and Kathrin Töllner for contributing unpublished data from PTZ seizure threshold experiments in epileptic and naive mice. The author’s own studies were supported by grants from the Deutsche Forschungsgemeinschaft (Bonn, Germany) and funding from the European Union’s Seventh Framework Programme (FP7/2007–2013) under Grant Agreement no 602102 (EPITARGET).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wolfgang Löscher.

Additional information

This review is dedicated to my colleague and friend Dr. H. Steve White to acknowledge his outstanding contributions in anti-seizure drug discovery and development of novel models of drug-resistant seizures.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Löscher, W. 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 (2017). https://doi.org/10.1007/s11064-016-2025-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-016-2025-7

Keywords

Navigation