Skip to main content
Log in

Evaluation of an Innovative Population Pharmacokinetic-Based Design for Behavioral Pharmacodynamic Endpoints

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

Pre-clinical behavioral pharmacology studies supporting indications like analgesia typically consist of at least three different studies; dose-finding, duration of effect, and tolerance-development studies. Pharmacokinetic (PK) plasma samples are generally taken from a parallel group of animals to avoid disruption of the behavioral pharmacodynamic (PD) endpoint. Our objective was to investigate if pre-clinical behavioral pharmacology studies in rats could be performed effectively by combining three studies into a single experimental design and using sparse PK sampling in the same animals as for PD. A refined dosing strategy was applied for a muscarinic agonist, AZD6088, using the rat spinal nerve ligation heat hyperalgesia model. PD measurements were performed on day 1, 3, 5 and 8. Two PK samples per day were taken day 2 and 4. In a separate control group, PD measurements were performed on rats without PK sampling. Data was analyzed using a population approach in NONMEM. The animals produced a consistent and reproducible response irrespective of day of testing suggesting that blood sampling on alternate days did not interfere with the PD responses. A direct concentration–effect relationship with good precision was established and no tolerance development was observed. The new design combining three studies into one and eliminating a satellite PK group realized substantial savings compared to the old design; animal use was reduced by 58% and time required to generate results was reduced by 55%. The design described here delivers substantial savings in animal lives, time, and money whilst still delivering a good quality and precise description of the PKPD relationship.

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
Fig. 6

Similar content being viewed by others

References

  1. Abatan OI, Welch KB, Nemzek JA. Evaluation of saphenous venipuncture and modified tail-clip blood collection in mice. J Am Assoc Lab Anim Sci. 2008;47(3):8–15.

    PubMed  CAS  Google Scholar 

  2. Bardin L, Malfetes N, Newman-Tancredi A, Depoortere R. Chronic restraint stress induces mechanical and cold allodynia, and enhances inflammatory pain in rat: relevance to human stress-associated painful pathologies. Behav Brain Res. 2009;205:360–6.

    Article  PubMed  CAS  Google Scholar 

  3. Howard BR. Experimental design and statistics in biomedical research. ILAR J. 2002;43(4):194–201.

    PubMed  CAS  Google Scholar 

  4. Huang F, Zhang M, Chen Y-J, Li Q, Wu A-Z. Psychological stress induces temporary mastictory muscle mechanical sensitivity in rats. J Biomed Biotechnol. 2010;2011:1–8.

    Google Scholar 

  5. Torsten PV, Ulrich-Lai YM, Ostrander MM, Dolgas CM, Elfers EE, Seeley RJ, D’Alessio DA, Herman JP. Comparative analysis of ACTH and corticosterone sampling methods in rat. Am J Physiol Endocrinol Metab. 2005;289:E823–8.

    Article  Google Scholar 

  6. Russell WMS, Burch RL. The principles of humane experimental technique. London: Methuen; 1959.

    Google Scholar 

  7. Bartolini A, Ghelardini C, Fanetti L, Malcangio M, Malmberg-Aiello P, Giotti A. Role of muscarinic receptor subtypes in central antinociception. Br J Pharmacol. 1992;105:77–82.

    Article  PubMed  CAS  Google Scholar 

  8. Bystander FP, McKinzie DL, Felder CC, Wess J. Use of M1–M5 muscarinic receptor knockout mice as novel tools to delineate the physiological roles of the muscarinic cholinergic system. Neurochem Res. 2003;28:437–42.

    Article  Google Scholar 

  9. Duttaroy A, Gomeza J, Gan J-W, Siddiqui N, Basile AS, Harman D, Smith PL, Felder CC, Levey AI, Wess J. Evaluation of muscarinic agonist-induced analgesia in muscarinic acetylcholine receptor knockout mice. Mol Pharmacol. 2002;62:1084–93.

    Article  PubMed  CAS  Google Scholar 

  10. Ghelardini C, Galeotti N, Bartolini A. Loss of muscarinic antinociception by antisense inhibition of M1 receptors. Br J Pharmacol. 2000;129:1633–40.

    Article  PubMed  CAS  Google Scholar 

  11. Ghelardini C, Galeotti N, Lelli C, Bartolini A. M1 receptors activation is a requirement for arecoline analgesia. Il Farmaco. 2001;56:383–5.

    Article  PubMed  CAS  Google Scholar 

  12. Guimaraes AP, Guimaraes FS, Prado WA. Modulation of carbachol-induced antinociception from the rat periaqueductal gray. Brain Res. 2000;51:471–8.

    CAS  Google Scholar 

  13. Heinrich JN, Butera JA, Carrick T, Kramer A, Kowal D, Lock T, Marquis K, Pausch MH, Popiolek M, Sun R, Tseng E, Uveges A, Mayer SC. Pharmacological comparison of muscarinic ligands: historical versus more recent muscarinic M1-preferring receptor agonists. Eur J Pharmacol. 2009;605(1–3):53–6.

    Article  PubMed  CAS  Google Scholar 

  14. Kiesewetter DO, Jagoda EM, Shimoji K, Ma Y, Eckelman WC. Evaluation of [18F]fluoroxanomeline 5-{4-[(6-[18F]fluorohexyl)oxy]-1,2,5-thiadiazol-3-yl}-1-methyl-1,2,3,6-tetrahydropyridine in muscarinic knockout mice. Nucl Med Biol. 2007;34:141–52.

    Article  PubMed  CAS  Google Scholar 

  15. Naguib M, Yaksh TL. Characterization of muscarinic receptor subtypes that mediate antinociception in the rat spinal cord. Anesth Analg. 1997;85:847–53.

    PubMed  CAS  Google Scholar 

  16. Sullivan NR, Leventhal L, Harrison J, Smith VA, Cummons TA, Spangler TB, Sun S-C, Lu P, Uveges AJ, Strassle BW, Piesla MJ, Ramdass R, Barry A, Schantz J, Adams W, Whiteside GT, Adedoyin A, Jones PG. Pharmacological characterization of the muscarinic agonist (3R,4R)-3-(3-Hexylsulfanyl-pyrazin-2-yloxy)-1-aza-bicyclo[2.2.1]heptane (WAY-132983) in in vitro and in vivo models of chronic pain. J Pharmacol Exp Ther. 2007;322:1294–304.

    Article  PubMed  CAS  Google Scholar 

  17. Wess J, Duttaroy A, Gomeza J, Zhang W, Yamada M, Felder CC, Bernardini N, Reeh PW. Muscarinic receptor subtypes mediating central and peripheral antinociception studied with muscarninc receptor knockout mice: a review. Life Sci. 2003;72:2047–54.

    Article  PubMed  CAS  Google Scholar 

  18. Wess J, Eglen RM, Gautam D. Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development. Nat Rev. 2007;6:721–33.

    Article  CAS  Google Scholar 

  19. Wess J. Muscarinic acetylcholine receptor knockout mice: novel phenotypes and clinical implications. Annu Rev Pharmacol Toxicol. 2004;44:423–50.

    Article  PubMed  CAS  Google Scholar 

  20. Wess J. Novel insights into muscarinic acetylcholine receptor function using gene targeting technology. Trends Pharmacol Sci. 2003;24:414–9.

    Article  PubMed  CAS  Google Scholar 

  21. Kim SH, Chung JM. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain. 1992;50(3):355–63.

    Article  PubMed  CAS  Google Scholar 

  22. Wählby U, Jonsson EN, Karlsson MO. Assessment of actual significance levels for covariate effects in NONMEM. J Pharmacokinet Pharmacodyn. 2001;28(3):231–52.

    Article  PubMed  Google Scholar 

  23. Jonsson EN, Karlsson MO. Xpose—an S-PLUS based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput Methods Programs Biomed. 1999;58(1):51–64.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We are grateful to Denis Projean and Louis Matthyssen for their excellent contribution and technical assistance.

Conflict of Interest

The authors state that there are no conflicts of interest in respect to the work reported in this paper.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anders Viberg.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Viberg, A., Martino, G., Lessard, E. et al. Evaluation of an Innovative Population Pharmacokinetic-Based Design for Behavioral Pharmacodynamic Endpoints. AAPS J 14, 657–663 (2012). https://doi.org/10.1208/s12248-012-9380-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12248-012-9380-3

Key words

Navigation