Skip to main content
Log in

Sex Specificity in Methadone Analgesia in the Rat: A Population Pharmacokinetic and Pharmacodynamic Approach

  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose. To quantify the extent to which a sex-specific dichotomy in the temporal evolution of the analgesic effect, after intravenous (i.v.) methadone injection in the rat, relates to the pharmacokinetics (PK) and pharmacodynamics (PD) that mediate the dose-to-effect pathway.

Methods. Tail-flick analgesia was measured after i.v. methadone injection (0.35 mg/kg) in female (n = 16) and male (n = 16) Sprague-Dawley rats. The PK were evaluated in separate female (n = 56) and male (n = 56) rats after they had received the same dose of methadone i.v. (0.35 mg/kg). A bicompartmental model described the kinetics and a sigmoid Emax model-related drug effect vs. simulated concentrations (pharmacodynamics) at the times of effect measurement. All model parameters as well as interanimal and assay variabilities were estimated with a mixed-effects population method using the program NONMEM.

Results. The area under the effect-time curve (AUCE0-120) was (mean ± interanimal SD) 1859 ± 346 min in the females, which was significantly lower than the 4871 ± 393 min in the males (P < 0.0001). On the contrary, the profiles of concentration vs. time were higher in females and, therefore, corresponded inversely to the effect vs. time-relative magnitudes. The central volume of distribution, V1, was 1.94 ± 0.37 l/kg for female rats and 3.01 ± 0.33 l/kg for male rats. Also, the central clearance was 0.077 ± 0.006 l/min/kg and 0.102 ± 0.005 l/min/kg, respectively, for female and male rats. Both parameters differed significantly between sexes (P < 0.0001). The pharmacodynamic maximum observed effect parameter (Emax) was 37% ± 29% in female rats and 85% ± 16% in male rats, and these values were significantly different (P < 0.0001). The parameter for the concentration eliciting half of Emax (EC50) was 24.1 ± 7.5 μg/l in female rats and 20.3 ± 2.9 μg/l in male rats, and the Hill-related exponent, γ, was 6.3 ± 3.9 in female rats and 5.5 ± 4.1 in male rats. These parameters did not differ significantly (at the P < 0.05 level).

Conclusions. A sex-specific dichotomy in the methadone antinociceptive effect, in the rat, was not proportionally related to plasma concentrations. Each sex corresponded to a distinct subpopulation of the PK parameters and one of the pharmacodynamic parameters (Emax). When the course of a drug involves PK or PD subpopulations, PK/PD modeling can afford the safest prediction of the effect-time evolution for a particular dose.

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.

Similar content being viewed by others

REFERENCES

  1. A. I. Baamonde, A. Hidelgo, and F. Andres-Trelles. Sex related differences in the effects of morphine and stress on visceral pain. Neuropharmacology 28:967–970 (1989).

    Google Scholar 

  2. T. J. Cicero, B. Nock, and E. R. Meyer. Gender-related differences in the antinociceptive properties of morphine. J. Pharmacol. Exp. Ther. 279:767–773 (1996).

    Google Scholar 

  3. T. J. Cicero, B. Nock, and E. R. Meyer. Sex-related differences in morphine´ s antinociceptive activity: Relationship to serum and brain morphine concentration. J. Pharmacol. Exp. Ther. 282:939–944 (1997).

    Google Scholar 

  4. B. Kest, E. Sarton, and A. Dahan. Gender differences in opioidmediated analgesia. Anesthesiology 93:539–547 (2000).

    Google Scholar 

  5. R. Z. Harris, L. Z. Benet, and J. B. Schwartz. Gender effects in pharmacokinetics and pharmacodynamics. Drugs 50:222–239 (1995).

    Google Scholar 

  6. P. A. Thurmann and B. C. Hompesch. Influence of gender on the pharmacokinetics and pharmacodynamics of drugs. Int. J. Clin. Pharmacol. Ther. 36:586–590 (1998).

    Google Scholar 

  7. B. Beierle. Meibohm, and H. Derendorf. Gender differences in pharmacokinetics and pharmacodynamics. Int. J. Clin. Pharmacol. Ther. 37:529–547 (1999).

    Google Scholar 

  8. P. L. Bonate. Gender-related differences in xenobiotic metabolism. J. Clin. Pharmacol. 31:684–690 (1991).

    Google Scholar 

  9. R. Kato and Y. Yamazoe. Sex-specific cytochrome P-450 as a cause of sex-and species-related differences in drug toxicity. Toxicol. Lett. 65:661–667 (1992).

    Google Scholar 

  10. E. Tanaka. Gender-related differences in pharmacokinetics and their clinical significance. J. Clin. Pharmacol. Ther. 24:339–346 (1999).

    Google Scholar 

  11. P. A. Routledge, W. W. Stargel, B. B. Kitchell, A. Barchowsky, and D. G. Shand. Sex-related differences in the plasma protein binding of lignocaine and diazepam. Br. J. Clin. Pharmacol. 11:245–250 (1981).

    Google Scholar 

  12. M. Farrell, J. Ward, R. Mattick, W. Hall, G. V. Stimson, D. des Jarlais, M. Gossop, and J. Strang. Methadone maintenance treatment in opiate dependence: A review. Br. Med. J. 309:997–1001 (1994).

    Google Scholar 

  13. R. Fainsinger, T. Schoeller, and E. Bruera. Methadone in the management of cancer pain: a review. Pain 52:137–147 (1993).

    Google Scholar 

  14. C. Ripamonti, E. Zecca, and E. Bruera. An update on the clinical use of methadone for cancer pain. Pain 70:109–115 (1997).

    Google Scholar 

  15. K. R. Dyer, D. J. Foster, J. M. White, A. A. Somogyi, A. Menelaou, and F. Bochner. Steady-state pharmacokinetics and pharmacodynamics in methadone maintenance patients: comparison of those who do and do not experience withdrawal and concentration-effect relationships. Clin. Pharmacol. Ther. 65:685–694 (1999).

    Google Scholar 

  16. C. E. Inturrisi, W. A. Colburn, R. F. Kaiko, R. W. Houden, and K. M. Foley. Pharmacokinetics and pharmacodynamics of methadone in patients with chronic pain. Clin. Pharmacol. Ther. 41:392–401 (1987).

    Google Scholar 

  17. K. Wolff, A. Rostami-Hodjegan, S. Shires, A. W. M. Hay, M. Feely, R. Calvert, D. Raistrick, and G. T. Tucker. The pharmacokinetics of methadone in healthy subjects and opiate users. Br. J. Clin. Pharmacol. 44:325–334 (1997).

    Google Scholar 

  18. C. B. Eap, C. Cuendet, and P. Baumann. Binding of dmethadone, l-methadone and d-l-methadone to protein in plasma of healthy volunteers: Role of the variants of a1-acid glycoprotein. Clin. Pharmacol. Ther. 47:338–346 (1990).

    Google Scholar 

  19. E. Gomez, R. Martinez-Jorda, E. Suarez, M. J. Garrido, and R. Calvo. Altered methadone analgesia due to changes in plasma protein binding: Role of the route of administration. Gen. Pharmacol. 26:1273–1276 (1995).

    Google Scholar 

  20. D. Rostami-Hodjegan, W. Wolff, A. W. M. Hay, D. Raistrick, R. Calvert, and G. T. Tucker. Population pharmacokinetics of methadone in opiate users: Characterization of time-dependent changes. Br. J. Clin. Pharmacol. 48:43–52 (1999).

    Google Scholar 

  21. D. J. Foster, A. A. Somogyi, and F. Bochner. Methadone Ndemethylaction in human liver microsomes: Lack of stereoselectivity and involvement of CYP3A4. Br. J. Clin. Pharmacol. 47:403–412 (1999).

    Google Scholar 

  22. R. K. Verbeeck. J-A Cardinal, and S. M. Wallace. Effect of age and gender on the plasma binding of acidic and basic drugs. Eur. J. Clin. Pharmacol. 27:91–97 (1984).

    Google Scholar 

  23. R. E. Bartok and R. M. Craft. Sex differences in opioid antinociception. J. Pharmacol. Exp. Ther. 282:769–778 (1997).

    Google Scholar 

  24. J. Candido, K. Lutfy, B. Billings, V. Sierra, A. Duttaroy, C. E. Inturrisi, and B. C. Yoburn. Effect of adrenal and sex hormones on opioid analgesia and opioid receptor regulation. Pharmacol. Biochem. Behav. 42:685–692 (1992).

    Google Scholar 

  25. F. E. D'Amour and D. L. Smith. A method for determining loss of pain sensation. J. Pharmacol. Exp. Ther. 72:74–79 (1941).

    Google Scholar 

  26. K. Wolff, M. Sanderson, A. W. M. Hay, and D. Raistrick. Methadone concentrations in plasma and their relationship to drug dosage. Clin. Chem. 37:205–209 (1991).

    Google Scholar 

  27. M. J. Garrido, M. Valle, R. Calvo, and I. F. Troconiz. Altered plasma and brain disposition and pharmacodynamics of methadone in abstinent rats. J. Pharmacol. Exp. Ther. 288:179–187 (1999).

    Google Scholar 

  28. E. I. Ette, A. W. Kelman, C. A. Howie, and B. Whiting. Analysis of animal pharmacokinetic data: performance of the one point per animal design. J. Pharmacokinet. Biopharm. 23:551–566 (1995).

    Google Scholar 

  29. L. B. Sheiner, D. R. Stanski, S. Vozeh, R. D. Miller, and J. Ham. Simultaneous modeling of pharmacokinetics and pharmacodynamics: Application to d-tubocurarine. Clin. Pharmacol. Ther. 25:358–371 (1979).

    Google Scholar 

  30. A. M. Davis and C. E. Inturrisi. d-Methadone blocks morphine tolerance and N-methyl-d-aspartate induced hyperalgesia. J. Pharmacol Exp. Ther. 289:1048–1053 (1999).

    Google Scholar 

  31. K. L. Kepler, K. M. Standifer, D. Paul, G. W. Pasternak, B. Kest, and R. J. Bodnar. Gender effects upon central opioid analgesia. Pain 45:87–94 (1991).

    Google Scholar 

  32. R. M. Craft, J. A. Stratmann, R. E. Bartok, T. I. Walpole, and S. J. King. Sex differences in development of morphine tolerance and dependence in the rat. Psychopharmacology 143:1–7 (1999).

    Google Scholar 

  33. K. Wolff, A. Rostami-Hodjegan, A. W. M. Hay, D. Raistrick, and G. Tucker. Population based pharmacokinetic approach for methadone monitoring of opiate addicts: Potential clinical utility. Addiction 95:1771–1783 (2000).

    Google Scholar 

  34. G. S. F. Ling, J. C. Umans, and C. E. Inturrisi. Methadone: Radioimmunoassay and pharmacokinetics in the rat. J. Pharmacol. Exp. Ther. 217:147–151 (1981).

    Google Scholar 

  35. M.-I. Nilsson, E. Anggard, and J. Holmstrand. and L-M Gunne. Pharmacokinetics of methadone during maintenance treatment: Adaptive changes during the induction phase. Eur. J. Clin. Pharmacol. 22:343–349 (1982).

    Google Scholar 

  36. K. Wilson, C. N. Reynolds, and D. Burnett. Inter and intraindividual variation in the metabolism of methaqualone in man after a single oral dose. Eur. J. Clin. Pharmacol. 13:291–297 (1978).

    Google Scholar 

  37. J. D. Lane, J. F. Steege, S. L. Rupp, and C. M. Kuhn. Menstrual cycle effects on caffeine elimination in the human female. Eur. J. Clin. Pharmacol. 43:543–546 (1992).

    Google Scholar 

  38. M. Rowland and T. N. Tozer. Clinical Pharmacokinetics: Concepts and Applications 3rd Ed. William & Wilkins, Media, PA, 1995 pp. 137–155.

    Google Scholar 

  39. D. J. Greenblat, H. Friedman, E. S. Burstein, J. M. Scavone, G. T. Blyden, H. R. Ochs, L. G. Miller, J. S. Harmatz, and R. I. Shader. Trazodone kinetics: Effect of age, gender, and obesity. Clin. Pharmacol. Ther. 42:193–200 (1987).

    Google Scholar 

  40. E. Schwartz, R. S. Matteo, E. Ornstein, W. L. Young, and K. L. Myers. Pharmacokinetics of sufentanil in obese patients. Anesth. Analg. 73:790–793 (1991).

    Google Scholar 

  41. C. M. Hunt, W. R. Westerkam, and G. M. Stave. Effect of age and gender on the activity of human hepatic CYP3A. Biochem. Pharmacol. 44:275–283 (1992).

    Google Scholar 

  42. E. K. Krzanowska and R. J. Bodner. Morphine antinociception elicited from the ventrolateral periaductal gray is sensitive to sex and gonadectomy differences in rats. Brain Res. 821:224–230 (1999).

    Google Scholar 

  43. J. S. Boyer, M. M. Morgan, and R. M. Craft. Microinjection of morphine into rostral ventromedial medulla produces greater antinociception in male compared to female rats. Brain Res. 796:315–318 (1998).

    Google Scholar 

  44. S. J. Liu, D. L. Roerig, and R. I. H. Wang. Brain and plasma levels of methadone and their relationship to analgesic activity of methadone in rats. Drug Metab. Dispos. 11:335–338 (1983).

    Google Scholar 

  45. K. Kristensen, C. B. Christensen, and L. L. Christrup. The mu1, mu2, delta, kappa opioid receptor binding profiles of methadone stereoisomers and morphine. Life Sci. 56:45–50 (1995).

    Google Scholar 

  46. K. Kristensen, T. Blemmer, H. R. Angelo, L. L. Christrup, N. E. Drenck, S. M. Rasmussen, and P. Sjogren. Stereoselective pharmacokinetics of methadone in chronic pain patients. Ther. Drug Monitor. 18:221–227 (1996).

    Google Scholar 

  47. M. A. Carlos, P. du Souich, R. Carlos, E. Suarez, J. C. Lukas, and R. Calvo. Effect of omeprazole on oral and intravenous RS-methadone pharmacokinetics and pharmacodynamics in the rat. J. Pharm. Sci. 91:1627–1638 (2002).

    Google Scholar 

  48. C. Cordon-Cardo, J. P. O'Brien, D. Casals, L. Rittman-Grauer, J. L. Bierdler, M. R. Melamed, and J. R. Bertino. Multidrug resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc. Natl. Acad. Sci. USA 86:695–698 (1989).

    Google Scholar 

  49. R. Bouer, L. Barthe, C. Philibert, C. Tourmaire, J. Woodley, and G. Houin. The roles of P-glycoprotein and intracellular metabolism in the intestinal absorption of methadone: in vitro studies using the rat-inverted intestinal sack. Fundam. Clin. Pharmacol. 13:494–500 (1999).

    Google Scholar 

  50. S. J. Thopmson, D. V. M. Kari Koszdin, and C. M. Bernards. Opiate-induced analgesia is increased and prolonged in mice lacking P-glycoprotein. Anesthesiology 92:1392–1399 (2000).

    Google Scholar 

  51. C. J. R. Parker, J. M. Hunter, and S. L. Snowdon. Effect of age, gender, and anesthetic technique on the pharmacodynamics of atracurium. Br. J. Anaesth. 70:38–41 (1993).

    Google Scholar 

  52. E. Sarton, E. Olofsen, R. Romberg, J. Hartigh, B. Kest, D. Nieuwenhuijs, A. Burm, L. Teppema, and A. Dahan. Sex differences in morphine analgesia. Anesthesiology 93:1245–1254 (2000).

    Google Scholar 

  53. R. P. Hammer. Mu-opiate receptor binding in the medial preoptic area is cyclical and sexually dimorphic. Brain Res. 515:187–192 (1990).

    Google Scholar 

  54. R. B. Simerly, L. Swanson, and R. A. Gorski. Demonstration of a sexual dimorphism in the distribution of serotonin inmunoreactive fevers in the medial preoptic nucleus of the rat. J. Comp. Neurol. 225:151–176 (1984).

    Google Scholar 

  55. K. L. Kepler, B. Kest, J. M. Kiefel, M. L. Cooper, and R. J. Bodnar. Roles of gender and gonadectomy and estrous phase in the analgesic effects of intracerebroventricular morphine in rats. Pharmacol. Biochem. Behav. 34:119–127 (1989).

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rosario Calvo.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rodriguez, M., Carlos, M.A., Ortega, I. et al. Sex Specificity in Methadone Analgesia in the Rat: A Population Pharmacokinetic and Pharmacodynamic Approach. Pharm Res 19, 858–867 (2002). https://doi.org/10.1023/A:1016117218760

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1016117218760

Navigation