Skip to main content
Log in

The Pharmacology of Imepitoin: The First Partial Benzodiazepine Receptor Agonist Developed for the Treatment of Epilepsy

  • Review Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

Although benzodiazepines (BZDs) offer a wide spectrum of antiepileptic activity against diverse types of epileptic seizures, their use in the treatment of epilepsy is limited because of adverse effects, loss of efficacy (tolerance), and development of physical and psychological dependence. BZDs act as positive allosteric modulators of the inhibitory neurotransmitter GABA by binding to the BZD recognition site (“BZD receptor”) of the GABAA receptor. Traditional BZDs such as diazepam or clonazepam act as full agonists at this site, so that one strategy to resolve the disadvantages of these compounds would be the development of partial agonists with lower intrinsic efficacy at the BZD site of the GABAA receptor. Several BZD site partial or subtype selective compounds, including bretazenil, abecarnil, or alpidem, have been developed as anxioselective anxiolytic drugs, but epilepsy was not a target indication for such compounds. More recently, the imidazolone derivatives imepitoin (ELB138) and ELB139 were shown to act as low-affinity partial agonists at the BZD site of the GABAA receptor, and imepitoin was developed for the treatment of epilepsy. Imepitoin displayed a broad spectrum of anticonvulsant activity in diverse seizure and epilepsy models at tolerable doses, and, as expected from its mechanism of action, lacked tolerance and abuse liability in rodent and primate models. The more favorable pharmacokinetic profile of imepitoin in dogs versus humans led to the decision to develop imepitoin for the treatment of canine epilepsy. Based on randomized controlled trials that demonstrated antiepileptic efficacy and high tolerability and safety in epileptic dogs, the drug was recently approved for this indication in Europe. Hopefully, the favorable profile of imepitoin for the treatment of epilepsy in dogs will reactivate the interest in partial BZD site agonists as new treatments for human 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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Chang BS, Lowenstein DH. Epilepsy. N Engl J Med. 2003;349:1257–66.

    Article  PubMed  Google Scholar 

  2. Chandler K. Canine epilepsy: what can we learn from human seizure disorders? Vet J. 2006;172:207–17.

    Article  PubMed  Google Scholar 

  3. Löscher W, Schmidt D. Modern antiepileptic drug development has failed to deliver: ways out of the current dilemma. Epilepsia. 2011;52:657–78.

    Article  PubMed  Google Scholar 

  4. Löscher W, Schmidt D. Experimental and clinical evidence for loss of effect (tolerance) during prolonged treatment with antiepileptic drugs. Epilepsia. 2006;47:1253–84.

    Article  PubMed  Google Scholar 

  5. Rogawski MA, Löscher W. The neurobiology of antiepileptic drugs. Nat Rev Neurosci. 2004;5:553–64.

    Article  CAS  PubMed  Google Scholar 

  6. Vinkers CH, Olivier B. Mechanisms underlying tolerance after long-term benzodiazepine use: a future for subtype-selective GABA(A) receptor modulators? Adv Pharmacol Sci. 2012;2012:416864.

    PubMed Central  PubMed  Google Scholar 

  7. Haefely W. Partial agonists of the benzodiazepine receptor: from animal data to results in patients. Adv Biochem Psychopharmacol. 1988;45:275–92.

    CAS  PubMed  Google Scholar 

  8. Stephens DN, Sarter M. Bidirectional nature of benzodiazepine receptor ligands extends to effects on vigilance. Psychopharmacol Ser. 1988;6:205–17.

    CAS  PubMed  Google Scholar 

  9. Haefely W, Martin JR, Schoch P. Novel anxiolytics that act as partial agonists at benzodiazepine receptors. Trends Pharmacol Sci. 1990;11:452–6.

    Article  PubMed  Google Scholar 

  10. Haefely W, Facklam M, Schoch P, et al. Partial agonists of benzodiazepine receptors for the treatment of epilepsy, sleep, and anxiety disorders. Adv Biochem Psychopharmacol. 1992;47:379–94.

    CAS  PubMed  Google Scholar 

  11. Costa E, Guidotti A. Benzodiazepines on trial: a research strategy for their rehabilitation. Trends Pharmacol Sci. 1996;17:192–200.

    Article  CAS  PubMed  Google Scholar 

  12. Stephens DN, Turski L, Jones GH, et al. Abecarnil: a novel anxiolytic with mixed full agonist/partial agonist properties in animal models of anxiety and sedation. In: Stephens DN, editor. Anxiolytic β-carbolines. Berlin: Springer; 1993. p. 79–95.

    Chapter  Google Scholar 

  13. Skolnick P. Anxioselective anxiolytics: on a quest for the Holy Grail. Trends Pharmacol Sci. 2012;33:611–20.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Löscher W, Hönack D, Scherkl R, et al. Pharmacokinetics, anticonvulsant efficacy and adverse effects of the β-carboline abecarnil, a novel ligand for benzodiazepine receptors, after acute and chronic administration in dogs. J Pharmacol Exp Ther. 1990;255:541–8.

    PubMed  Google Scholar 

  15. Löscher W. Abecarnil shows reduced tolerance development and dependence potential in comparison to diazepam: animal studies. In: Stephens DN, editor. Anxiolytic β-carbolines: from molecular biology to the clinic. Berlin: Springer; 1993. p. 96–112.

  16. Turski L, Stephens DN, Jensen LH, et al. Anticonvulsant action of the β-carboline abecarnil: studies in rodents and baboon, Papio papio. J Pharmacol Exp Ther. 1990;253:344–52.

    CAS  PubMed  Google Scholar 

  17. Sannerud CA, Ator NA, Griffiths RR. Behavioral pharmacology of abecarnil in baboons: self-injection, drug discrimination and physical dependence. Behav Pharmacol. 1992;3:507–16.

    Article  CAS  PubMed  Google Scholar 

  18. Rostock A, Tober C, Dost R, et al. AWD-131–138. Drugs Future. 1998;23:253–5.

    Article  CAS  Google Scholar 

  19. Sigel E, Baur R, Netzer R, et al. The antiepileptic drug AWD 131–138 stimulates different recombinant isoforms of the rat GABA(A) receptor through the benzodiazepine binding site. Neurosci Lett. 1998;245:85–8.

    Article  CAS  PubMed  Google Scholar 

  20. Grunwald C, Rundfeldt C, Lankau HJ, et al. Synthesis, pharmacology, and structure-activity relationships of novel imidazolones and pyrrolones as modulators of GABAA receptors. J Med Chem. 2006;49:1855–66.

    Article  CAS  PubMed  Google Scholar 

  21. Gasparic A. Investigations on biotransformation of AWD 131–138. Doctoral thesis. Faculty of Bioscience, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany; 2005.

  22. Löscher W, Potschka H, Rieck S, et al. Anticonvulsant efficacy of the low-affinity partial benzodiazepine receptor agonist ELB 138 in a dog seizure model and in epileptic dogs with spontaneously recurrent seizures. Epilepsia. 2004;45:1228–39.

    Article  PubMed  Google Scholar 

  23. Frey H-H, Löscher W. Pharmacokinetics of anti-epileptic drugs in the dog: a review. J Vet Pharmacol Ther. 1985;8:219–33.

    Article  CAS  PubMed  Google Scholar 

  24. Potschka H, Fischer A, von Rüden EL, et al. Canine epilepsy as a translational model? Epilepsia. 2013;54:571–9.

    Article  CAS  PubMed  Google Scholar 

  25. European Medicines Agency (EMA). Summary of product characteristics (SPC) for Pexion (imepitoin). 2013. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/veterinary/medicines/002543/vet_med_000268.jsp&mid=WC0b01ac058008d7a8.

  26. Langen B, Egerland U, Bernoster K, et al. Characterization in rats of the anxiolytic potential of ELB139 [1-(4-chlorophenyl)-4-piperidin-1-yl-1,5-dihydro-imidazol-2-on], a new agonist at the benzodiazepine binding site of the GABAA receptor. J Pharmacol Exp Ther. 2005;314:717–24.

    Article  CAS  PubMed  Google Scholar 

  27. Kupferberg HJ. Antiepileptic drug development program: a cooperative effort of government and industry. Epilepsia. 1989;30(Suppl 1):S51–6.

    Article  PubMed  Google Scholar 

  28. Rostock A, Tober C, Rundfeldt C, et al. D-23129: a new anticonvulsant with a broad spectrum activity in animal models of epileptic seizures. Epilepsy Res. 1996;23:211–23.

    Article  CAS  PubMed  Google Scholar 

  29. Rundfeldt C. The new anticonvulsant retigabine (D-23129) acts as an opener of K+ channels in neuronal cells. Eur J Pharmacol. 1997;336:243–9.

    Article  CAS  PubMed  Google Scholar 

  30. Rundfeldt C, Netzer R. The novel anticonvulsant retigabine activates M-currents in Chinese hamster ovary-cells transfected with human KCNQ2/3 subunits. Neurosci Lett. 2000;282:73–6.

    Article  CAS  PubMed  Google Scholar 

  31. Rogawski MA. KCNQ2/KCNQ3 K+ channels and the molecular pathogenesis of epilepsy: implications for therapy. Trends Neurosci. 2000;23:393–8.

    Article  CAS  PubMed  Google Scholar 

  32. Lankau HJ, Unverferth K, Grunwald C, et al. New GABA-modulating 1,2,4-oxadiazole derivatives and their anticonvulsant activity. Eur J Med Chem. 2007;42:873–9.

    Article  CAS  PubMed  Google Scholar 

  33. Unverferth K, Dorre R, Korner B, et al. Synthesis and anticonvulsant activity of 3-carbamoyl-4-aryl-isoquinolin-1(2H)-ones. Arch Pharm (Weinheim). 1991;324:809–14.

    Article  CAS  Google Scholar 

  34. Unverferth K, Engel J, Hofgen N, et al. Synthesis, anticonvulsant activity, and structure-activity relationships of sodium channel blocking 3-aminopyrroles. J Med Chem. 1998;41:63–73.

    Article  CAS  PubMed  Google Scholar 

  35. Lankau HJ, Menzer M, Rostock A, et al. 3-Amino- and 5-aminopyrazoles with anticonvulsant activity. Arch Pharm (Weinheim). 1999;332:219–21.

    Article  CAS  Google Scholar 

  36. Lankau HJ, Menzer M, Rostock A, et al. Synthesis and anticonvulsant activity of new 4-aminopyrazoles and 5-aminopyrazol-3-ones. Pharmazie. 1999;54:705–6.

    CAS  PubMed  Google Scholar 

  37. Rostock A, Tober C, Rundfeldt C, et al. AWD 140–190: a new anticonvulsant with a very good margin of safety. Epilepsy Res. 1997;28:17–28.

    Article  CAS  PubMed  Google Scholar 

  38. Langen B, Rundfeldt C, Dost R, et al., Inventors. Method of treating or preventing central nervous system disorders with compounds having selectivity for the alpha 3 subunit of the benzodiazepine receptor. Patent application WO 2005/004867 A2; published 2005.

  39. Heinecke K, Thiel W. Identity and physicochemical properties of 1-(4-chlorophenyl)-4-morpholino-imidazolin-2-one, AWD 131–138. Pharmazie. 2001;56:458–61.

    CAS  PubMed  Google Scholar 

  40. Rostock A, Tober C, Dost R, et al. AWD 131–138: anxiolytic and anticonvulsant activities without side effects in animals. Behav Pharmacol. 1998;9(Suppl 1):S79.

    Google Scholar 

  41. Tober C, Rostock A, Bartsch R. Anticonvulsant profile of AWD 131–138, a derivative of a series of imidazolinones. Naunyn-Schmiedeberg’s Arch Pharmacol. 1998;357(Suppl 4):R98.

    Google Scholar 

  42. Tober C, Rostock A, Bartsch R. AWD 131–138: a derivative of a series of imidazolinones with anticonvulsant activity. Naunyn-Schmiedeberg’s Arch Pharmacol. 1998; 358 Suppl 1:P35.

  43. Tober C, Rostock A, White HS, et al. Anticonvulsant activity of AWD 131–138 in genetic animal models of epilepsy. Naunyn-Schmiedeberg’s Arch Pharmacol. 1999; 359 Suppl:R97.

  44. McNamara JO, Byrne MC, Dasheiff RM, et al. The kindling model of epilepsy: a review. Prog Neurobiol. 1980;15:139–59.

    Article  CAS  PubMed  Google Scholar 

  45. Dost R, Langen B, Rundfeldt C. The α-3 subunit selective benzodiazepine agonist ELB139 does not induce tolerance in animal models for anxiety and epilepsy. Soc Neurosci Abstr. 2005;678.1.

  46. Tober C, Stark B, Bartsch R, et al. Effects of AWD 131–138 in the amygdala kindling model of focal epilepsy. Naunyn-Schmied Arch Pharmacol. 2000;361 Suppl:R98.

    Google Scholar 

  47. File SE, Lister RG. Do the reductions in social interaction produced by picrotoxin and pentylenetetrazole indicate anxiogenic actions? Neuropharmacology. 1984;23:793–6.

    Article  CAS  PubMed  Google Scholar 

  48. Rabe H, Kronbach C, Rundfeldt C, et al. The novel anxiolytic ELB139 displays selectivity to recombinant GABA(A) receptors different from diazepam. Neuropharmacology. 2007;52:796–801.

    Article  CAS  PubMed  Google Scholar 

  49. Langen B, Dost R, Rundfeldt C. Antipsychotic effect of the alpha-3 subunit selective benzodiazepine agonist ELB139 in rats. Pharmacopsychiatry. 2005;38:A135.

    Article  Google Scholar 

  50. Yasar S, Bergman J, Munzar P, et al. Evaluation of the novel antiepileptic drug, AWD 131–138, for benzodiazepine-like discriminative stimulus and reinforcing effects in squirrel monkeys. Eur J Pharmacol. 2003;465:257–65.

    Article  CAS  PubMed  Google Scholar 

  51. Rieck S, Rundfeldt C, Tipold A. Anticonvulsant activity and tolerance of ELB138 in dogs with epilepsy: a clinical pilot study. Vet J. 2006;172:86–95.

    Article  CAS  PubMed  Google Scholar 

  52. Frey H-H, Göbel W, Löscher W. Pharmacokinetics of primidone and its active metabolites in the dog. Arch Int Pharmacodyn Ther. 1979;242:14–30.

    CAS  PubMed  Google Scholar 

  53. Gasparic A, Schupke H, Olbrich M, et al. Morpholine ring oxidation of AWD 131–138, a novel anxiolytic and anticonvulsant, is catalysed by CYP1A1/2. Drug Metab Rev. 2001;33(Suppl. 1):89.

    Google Scholar 

  54. Whyatt RM, Garte SJ, Cosma G, et al. CYP1A1 messenger RNA levels in placental tissue as a biomarker of environmental exposure. Cancer Epidemiol Biomark Prev. 1995;4:147–53.

    CAS  Google Scholar 

  55. Rundfeldt C, Dost R, Löscher W, et al., Inventors. Use of dihydroimidazolones for the treatment of epilepsy in dogs. Patent application WO 2004/032938 A1; published 2004; granted European patent EP1553952B1. 2008.

  56. Rundfeldt C, Schlichthaar R, Grunwald M, et al. The α-3 subunit selective benzodiazepine ligand ELB139 is well tolerated without sedation in healthy male volunteers while exerting pharmacodynamic effects assessed as power spectrum changes in Fourier-transformed EEG. Soc Neurosci Abstr. 2005;678.15.

  57. European Medicines Agency (EMA). European Public Assessment Report (EPAR) for Pexion (imepitoin). 2012. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/veterinary/002543/WC500140842.pdf.

  58. Shihab N, Bowen J, Volk HA. Behavioral changes in dogs associated with the development of idiopathic epilepsy. Epilepsy Behav. 2011;21:160–7.

    Article  PubMed  Google Scholar 

  59. Löscher W, Schwartz-Porsche D, Frey H-H, et al. Evaluation of epileptic dogs as an animal model of human epilepsy. Arzneim-Forsch (Drug Res). 1985;35:82–7.

    Google Scholar 

  60. Leppik IE, Patterson EN, Coles LD, et al. Canine status epilepticus: a translational platform for human therapeutic trials. Epilepsia. 2011;52(Suppl 8):31–4.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  61. Steinmetz S, Tipold A, Löscher W. Epilepsy after head injury in dogs: a natural model of posttraumatic epilepsy. Epilepsia. 2013;54:580–8.

    Article  PubMed  Google Scholar 

  62. Kanner AM. The treatment of depressive disorders in epilepsy: what all neurologists should know. Epilepsia. 2013;54(Suppl 1):3–12.

    Article  CAS  PubMed  Google Scholar 

  63. Löscher W, Rogawski MA. How theories evolved concerning the mechanism of action of barbiturates. Epilepsia. 2012;53(Suppl 8):12–25.

    Article  PubMed  Google Scholar 

  64. Löscher W, Hoffmann K, Twele F, et al. The novel antiepileptic drug imepitoin compares favourably to other GABA-mimetic drugs in a seizure threshold model in mice and dogs. Pharmacol Res. 2013;77:39–46.

    Article  PubMed  Google Scholar 

  65. Rostock A, Tober C, Dost R, et al. AWD 131–138 is a potential novel anxiolytic without sedation and amnesia: a comparison with diazepam and buspirone. Naunyn-Schmied Arch Pharmacol. 1998;358(Suppl 1):R68.

    Google Scholar 

Download references

Acknowledgments

We thank Dr. Klaus Unverferth and Dr. Hans-Joachim Lankau (both previously AWD) for their kind support in describing the chemical synthesis, Dr. Antje Gasparic (previously AWD) for her support in retrieving data from her doctoral thesis, Dr. Michael A. Rogawski for providing Fig. 1, and Dr. Richard W. Olsen for advice relating to Fig. 1. The authors of this article confirm that they have full control of all primary data and certify that no funding has been received for the preparation of this manuscript. C. Rundfeldt was a former employee of AWD and Elbion (which developed imepitoin) and acted as a consultant for Boehringer Ingelheim. W. Löscher acted as a consultant for AWD, Elbion, and Boehringer Ingelheim during the development of imepitoin. The authors have no patent rights to the compounds described in this review.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wolfgang Löscher.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rundfeldt, C., Löscher, W. The Pharmacology of Imepitoin: The First Partial Benzodiazepine Receptor Agonist Developed for the Treatment of Epilepsy. CNS Drugs 28, 29–43 (2014). https://doi.org/10.1007/s40263-013-0129-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-013-0129-z

Keywords

Navigation