Skip to main content

Advertisement

Log in

Pharmacokinetics and metabolism of 14C-levetiracetam, a new antiepileptic agent, in healthy volunteers

  • Pharmacokinetics and Disposition
  • Published:
European Journal of Clinical Pharmacology Aims and scope Submit manuscript

Abstract

The absorption, disposition and metabolism of levetiracetam, a new antiepileptic drug, have been investigated after a single oral dose of the 14C-labelled molecule administered to male healthy volunteers. As chiral inversion can occur during drug metabolism, the chiral inversion of levetiracetam and/or of its major metabolite produced by hydrolysis (the corresponding acid) was also investigated. Finally, the in vitro hydrolysis of levetiracetam to its major metabolite and the inhibition of this reaction in human blood have been studied. Levetiracetam was very rapidly absorbed in man, with the peak plasma concentration of the unchanged drug occurring at 0.25–0.50 h. The unchanged drug accounted for a very high percentage of plasma radioactivity (97–82%) at all the times measured, i.e. until 48 h after administration. The apparent volume of distribution of the compound was close (0.55–0.62 l/kg) to the volume of total body water. Total body clearance (0.80–0.97 ml/min/kg) was much lower than the nominal hepatic blood flow. The plasma elimination half-life of the unchanged drug varied between 7.4 h and 7.9 h. Plasma to blood ratio of total radioactivity concentrations was 1.1–1.3, showing that radioactivity concentrations were similar in blood cells and plasma. The balance of excretion was very high in all four volunteers. The predominant route of excretion was via urine, accounting for a mean of 95% of the administered dose after 4 days. Two major radioactive components were present in urine, the unchanged drug and the acid obtained by hydrolysis, accounting for 66% and 24% of the dose after 48 h, respectively. Hydrolysis of levetiracetam in human blood followed Michaelis-Menten kinetics with Km and Vmax values of 435 µM and 129 pmol/min/ml blood, respectively. Among the inhibitory agents investigated in this study, only paraoxon inhibited levetiracetam hydrolysis (92% inhibition at 100 µM). Oxidative metabolism occurred in man, although it accounted for no more than 2.5% of the dose. There was no evidence of chiral inversion.

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

Similar content being viewed by others

References

  1. Perucca E (1996) The new generation of antiepileptic drugs: advantages and disadvantages. Br J Clin Pharmacol 42:531–543

    CAS  PubMed  Google Scholar 

  2. Perucca E, Bialer M (1996) The clinical pharmacokinetics of the newer antiepileptic drugs. Clin Pharmacokin 31:29–46

    CAS  Google Scholar 

  3. Riva R, Albani F, Contin L, Baruzzi A (1996) Pharmacokinetic interactions between antiepileptic drugs. Clin Pharmacol 31:470–493

    CAS  Google Scholar 

  4. Roche C (1999) Qu’apportent les nouveaux antiepileptiques? Impact Médecin Hebdo 454:41–42

    Google Scholar 

  5. Strolin Benedetti M (2000) Enzyme induction and inhibition by new antiepileptic drugs: a review of human studies. Fund Clin Pharmacol 14:301–319

    Google Scholar 

  6. Gatti G, Bonami I, Jannuzzi G, Perucca E (2000) The new antiepileptic drugs: pharamacological and clinical aspects. Curr Pharm Des 6:839–860

    CAS  PubMed  Google Scholar 

  7. Lloyd P, Flesch G, Dieterle W (1994) Clinical pharmacology and pharmacokinetics of oxcarbazepine. Epilepsia 35:S10-S13

    Google Scholar 

  8. Stables J, Bialer M, Johannessen SI, Kupferberg HJ, Levy RH, Loiseau P, Perucca E (1995) Progress report on new antiepileptic drugs: a summary of the 2nd Eilat Conference. Epilepsy Res 22:235–246

    CAS  PubMed  Google Scholar 

  9. Bialer M, Johannessen SI, Kupferberg HJ, Levy RH, Loiseau P, Perucca E (1996) Progress report on new antiepileptic drugs: a summary of the 3rd Eilat Conference. Epilepsy Res 25:299–319

    CAS  PubMed  Google Scholar 

  10. Löscher W (1998) New visions in the pharmacology of anticonvulsion. Eur J Pharmacol 342:1–13

    PubMed  Google Scholar 

  11. Shorvon SD, van Rijckevorsel K (2002) A new antiepileptic drug—Levetiracetam, a pyrrolidone recently licensed as an antiepileptic drug. J Neurol Neurosurg Psychiatry 72:426–428

    CAS  PubMed  Google Scholar 

  12. Patsalos PN, Walker MC, Ratnaraj N, Sander JWAS, Shorvon SD (1995) The pharmacokinetics of levetiracetam (ucb L059) in patients with intractable epilepsy (abstract). Epilepsia 36:54

    Google Scholar 

  13. Edelbroeck PM, de Wilde-Ockeleon JM, Kastelejin-Nolste-Trenite DGA, Alpherts WCJ, Meijer JWA (1993) Evaluation of the pharmacokinetics and neuropsychometrics parameters in chronic comedicated epileptic patients of three increasing dosages of a novel antiepileptic drug, ucb LO59 250 mg capsules per each dose for one week followed by two weeks of placebo (abstract). Epilepsia 34:7

    Google Scholar 

  14. Caldwell J, Hutt AJ, Fournel-Gigleux S (1988) The metabolic chiral inversion and dispositional enantioselectivity of the 2-arylpropionic acids and their biological consequences. Biochem Pharmacol 37:105–114

    CAS  PubMed  Google Scholar 

  15. Wechter WJ, Loughead DG, Reischer RJ, Van-Giessen GJ, Kaiser DG (1974) Enzymatic inversion at saturated carbon: nature and mechanism of the inversion of (−)R-p-iso-butyl hydratropic acid. Biochem Biophys Res Commun 61:833–837

    CAS  PubMed  Google Scholar 

  16. Kaiser DG, Van-Giessen GJ, Reischer RJ, WecherWS (1976) Isomeric inversion of ibuprofen (R)-enantiomer in humans. J Pharm Sci 65:269–273

    CAS  Google Scholar 

  17. Lee EJD, Williams K, Day R, Graham G, Champion D (1985) Stereoselective disposition of ibuprofen enantiomers in man. Br J Clin Pharmacol 19:669–674

    CAS  PubMed  Google Scholar 

  18. Nakamura Y, Yamaguchi T, Hashimoto S, Ikatani S,Nakagawa Y (1981) Optical isomerization mechanism of(−)R-hydratropic acid derivatives. J Pharmacobiol Dyn 4:51

    Google Scholar 

  19. Naitoh T, Kawaguchi S, Kakiki M, Ohe H, Kajiwara A, Horie T (1998) Species differences and mechanism of the epimerization of a new MAO-A inhibitor. Xenobiotica 28:269–280

    CAS  PubMed  Google Scholar 

  20. Madhu C, Duff S, Baumgarten V, Rix P, Small D, Tang-Liu D (1997) Metabolic deesterification of tazorotene in human blood and rat and human liver microsomes. J Pharmaceut Sci 86:972–974

    CAS  Google Scholar 

  21. Nishigaki I, Itoh T, Ogasawara N (1983) Quantitative variations in polymorphic types of human red cell esterase D. Ann Hum Genet 47:187–192

    CAS  PubMed  Google Scholar 

  22. Trundle D, Marcial G, (1988) Detection of cholinesterase inhibition. The significance of cholinesterase measurements. Ann Clin Lab Sci 18:345–352

    CAS  PubMed  Google Scholar 

  23. Maghoub A, Idle JR, Dring LG, Lancaster R, Smith RL (1977) Polymorphic hydroxylation of debrisoquine in man. Lancet September 17:584–586

    Google Scholar 

  24. Nicolas J-M, Collart P, Gerin B, Mather G, Trager W, Levy R, Roba J, (1998) Levetiracetam: in vitro metabolism assays and prediction of drug-drug interaction. In: Boobis AR, Kremers P, Pelkonen O, Pithan K (eds) Proceedings, European symposium on the prediction of drug metabolism in man: progress and problems. 19–20 May 1998, Liège (Belgium), pp 193–195

  25. Wagner JG (1975) Fundamentals of clinical pharmacokinetics, 1st edn. Drug Intelligence Publications, Inc., Hamilton, Illinois, pp 57–128

  26. Rowland M, Tozer TN (1995) Estimation of elimination half-life from urine data. In: Clinical pharmacokinetics—concepts and applications, 3rd edn. Williams and Wilkins Media, Pa., pp 473–477

  27. Perucca E, Grimaldi R, Frigo GM, Sardi A, Mönig H, Ohnhaus EE (1988) Comparative effects of rifabutin and rifampicin in normal subjects. Eur J Clin Pharmacol 34:595–599

    CAS  PubMed  Google Scholar 

  28. Teunissen MWE, Bakker W, Meerburg-van der Torren JE, Breimer DD (1984) Influence of rifampicin treatment on antipyrine clearance and metabolite formation in patients with tuberculosis. Br J Clin Pharmac 18:701–706

    CAS  Google Scholar 

  29. Anderson GD (1998) A mechanistic approach to antiepileptic drug interactions. Ann Pharmacother 32:554–563

    CAS  PubMed  Google Scholar 

  30. Kerr BM, Rettie AE, Eddy AC, Loiseau P, Guyot M, Wilensky AJ, Levy RH (1989) Inhibition of human liver microsomal epoxide hydrolase by valproate and valpromide: in vitro/in vivo correlation. Clin Pharmacol Ther 46:82–93

    CAS  PubMed  Google Scholar 

  31. Wen X, Wang JS, Kivistö KT, Neuvonen PJ, Backman JT (2001) In vitro evaluation of valproic acid as an inhibitor of human cytochrome P450 isoforms: preferential inhibition of cytochrome-P450 2C9 (CYP2C9). Br J Clin Pharmacol 52:547–553

    CAS  PubMed  Google Scholar 

  32. Robbins DK, Wedlund PJ, Elsberg S, Oesch F, Thomas H (1992) Interaction of valproic acid and some analogues with microsomal epoxide hydrolase. Biochem Pharmacol 43:775–783

    CAS  PubMed  Google Scholar 

  33. Facciolà G, Avenoso A, Scordo MG, Madia AG, Ventimiglia A., Perucca E, Spina E (1999) Small effects of valproic acid on the plasma concentrations of clozapine and its major metabolites in patients with schizophrenic or affective disorders. Ther Drug Monitoring 21:341–345

    Article  Google Scholar 

  34. Lai ML, Huang JD (1993) Dual effect of valproic acid on the pharmacokinetics of phenytoin. Biopharmaceut Dispos 14:365–370

    CAS  Google Scholar 

  35. Tanaka E (1999) Clinically significant pharmacokinetic drug interactions between antiepileptic drugs. J Clin Pharm Ther 24:87–92

    Google Scholar 

  36. Minagawa T, Kohno Y, Suwa T, Tsuji A (1995) Species differences in hydrolysis of isocarbacyclin methyl ester (TEI-9090) by blood esterases. Biochem Pharmacol 49:1361–1365

    CAS  PubMed  Google Scholar 

  37. Kao YJ, Tellez J,Turner DR (1990) Dose-dependent effect of metoclopramide on cholinesterases and suxamethonium metabolism. Br J Anaesth 65:220–224

    CAS  PubMed  Google Scholar 

  38. Jensen FS, Skovgaard LT, Viby-Mogensen J (1995) Identification of human plasma cholinesterase variants in 6,688 individuals using biochemical analysis. Acta Anaesthesiol Scand 39:157–162

    CAS  PubMed  Google Scholar 

  39. Slatter JG, Su P, Sams JP, Schaff LJ, Wienkers LC (1997) Bioactivation of the anticancer agent CPT-11 to SN-38 by human hepatic microsomal carboxylesterases and the in vitro assessment of potiential drug interactions. Drug Metab Dispos 25:1157–1164

    CAS  PubMed  Google Scholar 

  40. Maren TH (1977) Use of inhibitors in physiological studies of carbonic anhydrase. Am J Physiol 232:F291–F297

    CAS  PubMed  Google Scholar 

  41. Matsumoto K, Miyazaki H, Fujii T, Hashimoto M (1989) Binding of sulfonamides to erythrocytes and their components. Chem Pharm Bull 37:1913–1915

    CAS  PubMed  Google Scholar 

  42. Masereel B, Rolin S, Abbate F, Scozzafava A, Supuran CT (2002) Carbonic anhydrase inhibitors: anticonvulsant sulfonamides incorporating valproyl and other lipophilic moieties. J Med Chem 45:312–320

    CAS  PubMed  Google Scholar 

  43. Calvo R, Carlos R, Errill S (1980) Effects of disease and acetasolamide on procaine hydrolysis by red blood cells enzymes. Clin Pharmacol Ther 27:179–183

    CAS  PubMed  Google Scholar 

  44. Williams FM, Clinical significance of esterases in man (1985) Clin Pharmacokin 10:392–403

    CAS  Google Scholar 

  45. Rowland M, Tozer TN (1995) Intravenous dose. In: Clinical pharmacokinetics—concepts and applications, 3rd edn. Williams and Wilkins Media, Pa., pp 18–33

  46. Rowland M, Tozer TN (1995) Elimination. In: Clinical pharmacokinetics—Concepts and applications, 3rd edn. Williams and Wilkins Media, Pa., pp 156–183

  47. Bourgeois BFD (1995) Important pharmacokinetic properties of antiepileptic drugs. Epilepsia 36:S1–S7

    Google Scholar 

  48. Testa B (1995) Monooxygenase catalysed N-C cleavage. In: The metabolism of drugs and other xenobiotics—biochemistry of Redox reactions. Testa B, Caldwell J (eds), Academic Press limited, London, pp 215–224

  49. Gower AJ, Noyer M, Verloes R, Gobert J., Wülfert E (1992) ucb L059, a novel anticonvulsant drug: pharmacological profile in animals. Eur J Pharmacol 222:193

    CAS  PubMed  Google Scholar 

  50. Noyer M, Gillard M, Matagne A, Hénichart JP, Wülfert E (1995) The novel antiepileptic drug levetiracetam (ucb L059) appears to act via a specific binding site in CNS membranes. Eur J Pharmacol 286:137–146

    CAS  PubMed  Google Scholar 

  51. Eichelbaum M, Gross A (1991) Implications of stereoselectivity in clinical pharmacokinetics. In: New trends in pharmacokinetics, Rescigno A Thakur AK (eds), NATO ASI Series, Series A: Life Sci 221, Plenum Press, New York, pp 271–279

  52. Beydoun A, Uthman BM, Sackellares JC (1995) Gabapentin: pharmacokinetics, efficacy and safety. Clin Neuropharmacol 18:469–481

    CAS  PubMed  Google Scholar 

  53. Elwes RDC, Binnie CD (1996) Clinical pharmacokinetics of newer antiepileptic drugs: lamotrigine, vigabatrin, gabapentin and oxcarbazepine. Clin Pharmacokin 30:403–415

    CAS  PubMed  Google Scholar 

  54. Van Wieringen A, Meijer JWA, Van Emde Boas W, Vermeij TAC (1990) Pilot study to determine the interaction of oxyracetam with antiepileptic drugs. Clin Pharmacokinet 18:332–338

    PubMed  Google Scholar 

  55. Perucca E, Gidal BE, Ledent E, Baltes E (2000) Levetiracetam does not interact with other antiepileptic drugs. Epilepsia 41:L08 (Proceedings Annual Meeting of the American Epilepsy Society, Los Angeles, USA, December 2000)

    Google Scholar 

  56. Browne TR, Szabo GK, Leppik IE, Josephs E, Paz J, Baltes E, Jensen CM (2000) Absence of pharmacokinetic drug interaction of levetiracetam with phenytoin determined by new technique. J Clin Pharmacol 40:590–595

    CAS  PubMed  Google Scholar 

  57. Klitgaard H, Matagne A, Gobert J, Wuelfert E. (1998) Evidence for a unique profile of levetiracetam in rodent models of seizures and epilepsy. Eur J Pharmacol 353:191–206

    CAS  PubMed  Google Scholar 

  58. Jamis-Dow CA, Katki AG, Collins JM, Klecker RW (1997) Rifampicin and rifabutin and their metabolism by human liver esterases. Xenobiotica 27:1015–1024

    CAS  PubMed  Google Scholar 

  59. Khanna P, Gupta MB, Gupta GP, Sanwal GG, Ali B (1991) Influence of chronic oral intake of cannabis extract on oxidative and hydrolytic metabolism of xenobiotics in rat. Biochem Pharmacol 41:109–113

    CAS  PubMed  Google Scholar 

  60. Khanna P, Kaur S, Sanwal GG and Ali B (1991) Characteristics of a cytosolic arylacylamidase metabolizing thiacetazone. J Pharmacol Exper Ther 262:1225–1231

    Google Scholar 

  61. Kaur S, Ali B (1983) The effects of phenobarbital, 3-methylcholanthrene and benzo(a)pyrene on the hydrolysis of xenobiotics in the rat. Biochem Pharmacol 32:3479–3480

    CAS  PubMed  Google Scholar 

  62. Satoh T, Moroi K (1977) Effect of pretreatment with tricresylphosphates and phenobarbital on the metabolism and toxicity of procaine in rats. Jpn J Pharmacol 27:233–237

    CAS  PubMed  Google Scholar 

  63. Schwark WS, Ecobichon DJ (1968) Subcellular localization and drug-induced changes of rat liver and kidneys esterases. Can J Physiol Pharmacol 46:207–212

    CAS  PubMed  Google Scholar 

  64. Siddiqui A, Srivastava SP, Ali B (1990) Effect of mancozeb on hydrolytic metabolism of xenobiotics. Res Commun Chem Pathol Pharmacol 70:249–252

    CAS  PubMed  Google Scholar 

  65. Nousiainen U, Hänninen O (1981) On the inducibility of cytosolic and microsomal carboxylesterase by phenobarbital in rat tissues. Acta Pharmacol Toxicol 49:77–80

    Google Scholar 

  66. Nousiainen U, Törrönen R, Hänninen O (1984) Differential induction of various carboxylesterases by certain polycyclic aromatic hydrocarbons in the rat. Toxicology 32:243–251

    CAS  PubMed  Google Scholar 

  67. Puche E, Garcia de la Serrana H, Mota C, Sazucedo R (1989) Serum aspirin-esterase activity in epileptic patients receiving treatment with phenobarbital, phenytoin, carbamazepine and valproic acid. Int J Clin Pharmacol Res 9:55–58

    CAS  PubMed  Google Scholar 

  68. Reife RA (1998) Assessing pharmacokinetic and pharmacodynamic interactions in clinical trials of antiepileptic drugs. Antiepil drug Dev Adv Neurol 76:95–103

    CAS  Google Scholar 

Download references

Acknowledgement

The authors wish to thank B. Kenda and M. Plisnier for helpful discussion on the structures of reference standards and metabolites, C. Doumergue for documentation and M. Rovei for preparation of the manuscript. The experiments comply with the current laws of the country in which the experiments were performed.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Margherita Strolin Benedetti.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Strolin Benedetti, M., Whomsley, R., Nicolas, JM. et al. Pharmacokinetics and metabolism of 14C-levetiracetam, a new antiepileptic agent, in healthy volunteers. Eur J Clin Pharmacol 59, 621–630 (2003). https://doi.org/10.1007/s00228-003-0655-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00228-003-0655-6

Keywords

Navigation