Pronounced antiseizure activity of the subtype-selective GABAA positive allosteric modulator darigabat in a mouse model of drug-resistant focal epilepsy

Rachel Gurrell, Philip Iredale, Alexis Evrard, Venceslas Duveau, Céline Ruggiero, Corinne Roucard, Rachel Gurrell, Philip Iredale, Alexis Evrard, Venceslas Duveau, Céline Ruggiero, Corinne Roucard

Abstract

Aim: Darigabat is an α2/3/5 subunit-selective positive allosteric modulator of GABAA receptors that has demonstrated broad-spectrum activity in several preclinical models of epilepsy as well as in a clinical photoepilepsy trial. The objective here was to assess the acute antiseizure effect of darigabat in the mesial temporal lobe epilepsy (MTLE) mouse model of drug-resistant focal seizures.

Methods: The MTLE model is generated by single unilateral intrahippocampal injection of low dose (1 nmole) kainic acid in adult mice, and subsequent epileptiform activity is recorded following implantation of a bipolar electrode under general anesthesia. After a period of epileptogenesis (~4 weeks), spontaneous and recurrent hippocampal paroxysmal discharges (HPD; focal seizures) are recorded using intracerebral electroencephalography. The number and cumulated duration of HPDs were recorded following administration of vehicle (PO), darigabat (0.3-10 mg kg-1 , PO), and positive control diazepam (2 mg kg-1 , IP).

Results: Darigabat dose-dependently reduced the expression of HPDs, demonstrating comparable efficacy profile to diazepam at doses of 3 and 10 mg kg-1 .

Conclusions: Darigabat exhibited a robust efficacy profile in the MTLE model, a preclinical model of drug-resistant focal epilepsy. A Phase II proof-of-concept placebo-controlled, adjunctive-therapy trial (NCT04244175) is ongoing to evaluate efficacy and safety of darigabat in patients with drug-resistant focal seizures.

Trial registration: ClinicalTrials.gov NCT04244175 NCT04686786.

Keywords: CVL-865; GABA; MTLE; darigabat; drug-resistant epilepsy; focal; gaba; seizure (total ≥5, ≤8).

Conflict of interest statement

RG and PI are or were employees of Cerevel Therapeutics at the time of this research and may own stock and/or stock options in the company.

© 2022 Cerevel Therapeutics, LLC. CNS Neuroscience & Therapeutics published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Example of a typical HPD in an EEG recording in a MTLE mouse selected in the study prior to compound administration. The HPD occurs spontaneously, with an obvious start and well‐defined stop over the basal EEG.
FIGURE 2
FIGURE 2
Time course of the effect of compound over time on number of HPDs. Time course of the effect of vehicle, darigabat (0.3, 3, 10 mg kg−1, PO), and diazepam (2 mg kg−1, IP) on the number of hippocampal paroxysmal discharges (HPD; mean ± SEM, n = 10) during baseline and post‐administration periods. The arrow indicates the timing of compound administration. #, ##, ###, ####: p < 0.05, 0.01, 0.001, 0.0001, respectively, as compared to vehicle using a two‐way ANOVA.
FIGURE 3
FIGURE 3
Effect of darigabat on number of HPDs. Dot plot (individual rodent) of hippocampal paroxysmal discharges (HPDs) during the 30‐ to 90‐min period after administration was measured and expressed as a % of the baseline HPD frequency (mean ± SEM, n = 10). #, ##, ###: p < 0.05, 0.01, and 0.001, respectively, as compared to vehicle using a one‐way ANOVA. Diazepam was administered 2 mg kg−1, IP.

References

    1. Loscher W. Fit for purpose application of currently existing animal models in the discovery of novel epilepsy therapies. Epilepsy Res. 2016;126:157‐184.
    1. Rho JM, White HS. Brief history of anti‐seizure drug development. Epilepsia Open. 2018;3(s2):114‐119.
    1. Duveau V, Pouyatos B, Bressand K, et al. Differential effects of antiepileptic drugs on focal seizures in the intrahippocampal kainite mouse model of mesial temporal lobe epilepsy. CNS Neurosci Ther. 2016;22:497‐506.
    1. Riban V, Bouilleret V, Pham‐Le B, Fritschy J. Evolution of hippocampal epileptic activity during the development of hippocampal sclerosis in a mouse model of temporal lobe epilepsy. Neurosci. 2002;112:101‐111.
    1. 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(6):1069‐1077.
    1. Stables JP, Bertram E, Dudek FE, et al. Therapy discovery for pharmacoresistant epilepsy and for disease‐modifying therapeutics: summary of the NIH/NINDS/AES models II workshop. Epilepsia. 2003;44:1472‐1478.
    1. McKernan RM, Whiting PJ. Which GABAA‐receptor subtypes really occur in the brain? Trends in Neurosci. 1996;19(4):139‐143.
    1. Rudolph U, Crestani F, Benke D, et al. Benzodiazepine actions mediated by specific gamma‐aminobutyric acid (a) receptor subtypes. Nature. 1999;401(6755):796‐800.
    1. McKernan RM, Rosahl TW, Reynolds DS, et al. Sedative but not anxiolytic properties of benzodiazepines are mediated by the GABAA receptor α1 subtype. Nat Neurosci. 2000;3(6):587‐592.
    1. Fradley RL, Guscott MR, Bull S, et al. Differential contribution of GABAA receptor subtypes to the anticonvulsant efficacy of benzodiazepine site ligands. Psychopharm. 2007;21(4):384‐391.
    1. Atack JR, Wafford KA, Tye SJ, et al. TPA023 [7‐(1,1‐dimethylethyl)‐6‐(2‐ethyl‐2H‐1,2,4‐triazol‐3‐ylmethoxy)‐3‐(2‐fluorophenyl )‐1,2,4‐triazolo[4,3‐b]pyridazine], an agonist selective for α2‐ and α3‐containing GABAA receptors, is a nonsedating anxiolytic in rodents and primates. J Pharmacol Exp Ther. 2006;316(1):410‐422.
    1. Crestani F, Low K, Keist R, et al. Molecular targets for the myorelaxant action of diazepam. Mol Pharmacol. 2001;59(3):442‐445.
    1. Dias R, Sheppard WF, Fradley RL, et al. Evidence for a significant role of α3‐containing GABAA receptors in mediating the anxiolytic effects of benzodiazepines. J Neurosci. 2005;25(46):10682‐10688.
    1. Stephens DN, King SL, Lambert JJ, Belelli D, Duka T. GABAA receptor subtype involvement in addictive behaviour. Genes Brain Behav. 2017;16(1):149‐184.
    1. Tan KR, Rudolph U, Luscher C. Hooked on benzodiazepines: GABAA receptor subtypes and addiction. Trends in Neurosci. 2011;34(4):188‐197.
    1. Cheng T, Wallace DM, Ponteri B, Tuli M. Valium without dependence? Individual GABAA receptor subtype contribution toward benzodiazepine addiction, tolerance, and therapeutic effects. Neuropsychiatr Dis Treat. 2018;14:1351‐1361.
    1. Vinkers CH, Olivier B. Mechanisms underlying tolerance after long‐term benzodiazepine use: a future for subtype‐selective GABAA receptor modulators? Adv Pharmacol Sci. 2012;416864:1‐19.
    1. Nickolls SA, Gurrell R, van Amerongen G, et al. Pharmacology in translation; the preclinical and early clinical profile of the novel α2/3 functionally selective GABAA receptor positive allosteric modulator PF‐06372865. Br J Pharmacol. 2018;175:708‐725.
    1. Owen RM, Blakemore D, Cao L, et al. Design and identification of a novel, functionally subtype selective GABAA positive allosteric modulator (PF‐06372865). J Med Chem. 2019;62(12):5773‐5796.
    1. Duveau V, Roucard C. A mesiotemporal lobe epilepsy mouse model. Neurochem Res. 2017;42:1919‐1925.
    1. Gurrell R, Gorman D, Whitlock M, et al. Photosensitive epilepsy: robust clinical efficacy of a selective GABA potentiator. Neurology. 2019;92(15):e1786‐e1795.
    1. Gurrell R, Dua P, Feng G, et al. A randomised, placebo‐controlled clinical trial with the α2/3/5 subunit selective GABAA positive allosteric modulator PF‐06372865 in patients with chronic low back pain. Pain. 2018;159(9):1742‐1751.
    1. Gurrell R, Whitlock M, Wei H, Shen Z, Ogden A. Safety, tolerability, and pharmacokinetics of multiple repeated oral doses of the α2/3/5‐subtype selective GABAA‐positive allosteric modulator PF‐06372865 in healthy volunteers. Clinical Pharmacol Drug Devel. 2021;10:756‐764. doi:10.1002/cpdd.912
    1. Simen A, Whitlock M, Ruolun Q, et al. An 8‐week, randomized, phase 2, double‐blind, sequential parallel‐group comparison study of two dose levels of PF‐06372865 compared to placebo as an adjunctive treatment in outpatients with inadequate response to standard of care for generalized anxiety disorder. J Clin Psychopharmacol. 2018;39(1):20‐27.
    1. Kleijn E, Rossum JM, Muskens ET, Rijntjes NV. Pharmacokinetics of diazepam in dogs, mice and humans. Acta Pharmacol Toxicol. 2009;29:109‐127.
    1. Atack JR. GABAA receptor alpha2/alpha3 subtype‐selective modulators as potential nonsedating anxiolytics. Curr Top Behav Neurosci. 2010;2:331‐360.
    1. Nickolls, S. , Mace, H. , Fish, R. , Edye M., Gurrell R., Ivarsson M., Pitcher T., Tanimoto‐Mori S., Richardson D., Sweatman C., Nicholson J., Ward C., Jinks J., Bell C., Young K., Rees H., Moss A., Kinloch R., McMurray G. A comparison of the α2/3/5 selective positive allosteric modulators L‐838,417 and TPA023 in nonclinical models of inflammatory and neuropathic pain. Adv Pharmacol Sci, 2011;608912, 2011, 1, 12.
    1. Duveau V, Buhl DL, Evrard A, et al. Pronounced antiepileptic activity of the subtype‐selective GABAA‐positive allosteric modulator PF‐06372865 in the GAERS absence epilepsy model. CNS Neurosci Ther. 2019;25(2):255‐260.
    1. French J, Krauss G, Kasteleijn D, et al. Effects of marketed antiepileptic drugs and placebo in the human photosensitivity screening protocol. Neurotherapeutics. 2014;11:412‐418.
    1. Yuen ES, Sims JR. How predictive are photosensitive epilepsy models as proof of principle trials for epilepsy? Seizure. 2014;23(6):490‐493.
    1. Leclercq K, Matagne A, Provins L, Klitgaard H, Kaminski RM. Pharmacological profile of the novel antiepileptic drug candidate padsevonil: characterization in rodent seizure and epilepsy models. J Pharmacol Exp Ther. 2020;372(1):11‐20.
    1. Loscher W. Single‐target versus multi‐target drugs versus combinations of drugs with multiple targets: preclinical and clinical evidence for the treatment or prevention of epilepsy. Front Pharmacol. 2021;12:730257.
    1. Wilcox KS, West PJ, Metcalf CS. The current approach of the epilepsy therapy screening program contract site for identifying improved therapies for the treatment of pharmacoresistant seizures in epilepsy. Neuropharmacology. 2020;166:107811.

Source: PubMed

3
Prenumerera