Skip to main content
Log in

Incorporating Pharmacological Target-Mediated Drug Disposition (TMDD) in a Whole-Body Physiologically Based Pharmacokinetic (PBPK) Model of Linagliptin in Rat and Scale-up to Human

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

Abstract

Linagliptin demonstrates substantial nonlinear pharmacokinetics due to its saturable binding to its pharmacological target dipeptidyl peptide 4 (DPP-4), a phenomenon known as target-mediated drug disposition (TMDD). In the current study, we established a novel whole-body physiologically-based pharmacokinetic (PBPK)-TMDD model for linagliptin. This comprehensive model contains plasma and 14 tissue compartments, among which TMDD binding process was incorporated in 9 of them, namely the plasma, kidney, liver, spleen, lung, skin, salivary gland, thymus, and reproductive organs. Our final model adequately captured the concentration-time profiles of linagliptin in both plasma and various tissues in both wildtype rats and DPP4-deficient rats following different doses. The association rate constant (kon) in plasma and tissues were estimated to be 0.943 and 0.00680 nM−1 h−1, respectively, and dissociation rate constant (koff), in plasma and tissues were estimated to be 0.0698 and 0.00880 h−1, respectively. The binding affinity of linagliptin to DPP-4 (Kd) was predicted to be higher in plasma (0.0740 nM) than that in tissue (1.29 nM). When scaled up to a human, this model captured the substantial and complex nonlinear pharmacokinetic behavior of linagliptin in human adults that is characterized by less-than dose-proportional increase in plasma exposure, dose-dependent clearance and volume of distribution, as well as long terminal half-life with minimal accumulation after repeated doses. Our modeling work is not only novel but also of high significance as the whole-body PBPK-TMDD model platform developed using linagliptin as the model compound could be applied to other small-molecule compounds exhibiting TMDD to facilitate their optimal dose selection.

Graphical abstract

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

Similar content being viewed by others

References

  1. Graefe-Mody U, Retlich S, Friedrich C. Clinical pharmacokinetics and pharmacodynamics of linagliptin. Clin Pharmacokinet. 2012;51(7):411–27.

    Article  CAS  PubMed  Google Scholar 

  2. Thomas L, Eckhardt M, Langkopf E, Tadayyon M, Himmelsbach F, Mark M. (R)-8-(3-amino-piperidin-1-yl)-7-but-2-ynyl-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3, 7-dihydro-purine-2, 6-dione (BI 1356), a novel xanthine-based dipeptidyl peptidase 4 inhibitor, has a superior potency and longer duration of action compared with other dipeptidyl peptidase-4 inhibitors. J Pharmacol Exp Ther. 2008;325(1):175–82.

    Article  CAS  PubMed  Google Scholar 

  3. Hartel S, Gossrau R, Hanski C, Reutter W. Dipeptidyl peptidase (DPP) IV in rat organs. Histochemistry. 1988;89(2):151–61.

    Article  CAS  PubMed  Google Scholar 

  4. Lankas GR, Leiting B, Roy RS, Eiermann GJ, Beconi MG, Biftu T, et al. Dipeptidyl peptidase IV inhibition for the treatment of type 2 diabetes: potential importance of selectivity over dipeptidyl peptidases 8 and 9. Diabetes. 2005;54(10):2988–94.

    Article  CAS  PubMed  Google Scholar 

  5. Elliott R, Morgan L, Tredger J, Deacon S, Wright J, Marks V. Glucagon-like peptide-1 (7–36) amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J Endocrinol. 1993;138(1):159–66.

    Article  CAS  PubMed  Google Scholar 

  6. Nauck M, Bartels E, Orskov C, Ebert R, Creutzfeldt W. Additive insulinotropic effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like peptide-1-(7-36) amide infused at near-physiological insulinotropic hormone and glucose concentrations. J Clin Endocrinol Metab. 1993;76(4):912–7.

    CAS  PubMed  Google Scholar 

  7. Schwartz SL. Treatment of elderly patients with type 2 diabetes mellitus: a systematic review of the benefits and risks of dipeptidyl peptidase-4 inhibitors. Am J Geriatr Pharmacother. 2010;8(5):405–18.

    Article  CAS  PubMed  Google Scholar 

  8. Retlich S, Duval V, Ring A, Staab A, Hüttner S, Jungnik A, et al. Pharmacokinetics and pharmacodynamics of single rising intravenous doses (0.5 mg–10mg) and determination of absolute bioavailability of the dipeptidyl peptidase-4 inhibitor linagliptin (BI 1356) in healthy male subjects. Clin Pharmacokinet. 2010;49(12):829–40.

    Article  CAS  PubMed  Google Scholar 

  9. Heise T, Graefe-Mody E, Hüttner S, Ring A, Trommeshauser D, Dugi K. Pharmacokinetics, pharmacodynamics and tolerability of multiple oral doses of linagliptin, a dipeptidyl peptidase-4 inhibitor in male type 2 diabetes patients. Diabetes Obes Metab. 2009;11(8):786–94.

    Article  CAS  PubMed  Google Scholar 

  10. Horie Y, Kanada S, Watada H, Sarashina A, Taniguchi A, Hayashi N, et al. Pharmacokinetic, pharmacodynamic, and tolerability profiles of the dipeptidyl peptidase-4 inhibitor linagliptin: a 4-week multicenter, randomized, double-blind, placebo-controlled phase IIa study in Japanese type 2 diabetes patients. Clin Ther. 2011;33(7):973–89.

    Article  CAS  PubMed  Google Scholar 

  11. Hüttner S, Graefe-Mody E, Withopf B, Ring A, Dugi K. Safety, tolerability, pharmacokinetics, and pharmacodynamics of single oral doses of BI 1356, an inhibitor of dipeptidyl peptidase 4, in healthy male volunteers. J Clin Pharmacol. 2008;48(10):1171–8.

    Article  PubMed  CAS  Google Scholar 

  12. Retlich S, Withopf B, Greischel A, Staab A, Jaehde U, Fuchs H. Binding to dipeptidyl peptidase-4 determines the disposition of linagliptin (BI 1356)–investigations in DPP-4 deficient and wildtype rats. Biopharm Drug Dispos. 2009;30(8):422–36.

    Article  CAS  PubMed  Google Scholar 

  13. An G. Concept of pharmacologic target-mediated drug disposition in large-molecule and small-molecule compounds. J Clin Pharmacol. 2020;60(2):149–63. https://doi.org/10.1002/jcph.1545.

    Article  CAS  PubMed  Google Scholar 

  14. Levy G. Pharmacologic target-mediated drug disposition. Clin Pharmacol Ther. 1994;56(3):248–52.

    Article  CAS  PubMed  Google Scholar 

  15. Fuchs H, Tillement JP, Urien S, Greischel A, Roth W. Concentration-dependent plasma protein binding of the novel dipeptidyl peptidase 4 inhibitor BI 1356 due to saturable binding to its target in plasma of mice, rats and humans. J Pharm Pharmacol. 2009;61(1):55–62.

    Article  CAS  PubMed  Google Scholar 

  16. Fuchs H, Binder R, Greischel A. Tissue distribution of the novel DPP-4 inhibitor BI 1356 is dominated by saturable binding to its target in rats. Biopharm Drug Dispos. 2009;30(5):229–40.

    Article  CAS  PubMed  Google Scholar 

  17. Levy G, Mager DE, Cheung WK, Jusko WJ. Comparative pharmacokinetics of coumarin anticoagulants L: physiologic modeling of S-warfarin in rats and pharmacologic target-mediated warfarin disposition in man. J Pharm Sci. 2003;92(5):985–94.

    Article  CAS  PubMed  Google Scholar 

  18. Mager DE, Jusko WJ. General pharmacokinetic model for drugs exhibiting target-mediated drug disposition. J Pharmacokinet Pharmacodyn. 2001;28(6):507–32.

    Article  CAS  PubMed  Google Scholar 

  19. Volz A-K, Krause A, Haefeli WE, Dingemanse J, Lehr T. Target-mediated drug disposition pharmacokinetic–pharmacodynamic model of bosentan and endothelin-1. Clin Pharmacokinet. 2017;56(12):1499–511.

    Article  CAS  PubMed  Google Scholar 

  20. An G, Liu W, Katz DA, Marek GJ, Awni W, Dutta S. Population pharmacokinetics of the 11β-hydroxysteroid dehydrogenase type 1 inhibitor ABT-384 in healthy volunteers following single and multiple dose regimens. Biopharm Drug Dispos. 2014;35(7):417–29.

    Article  CAS  PubMed  Google Scholar 

  21. Wu N, Hammock B, Lee KSS, An G. Simultaneous target-mediated drug disposition (TMDD) model for two small-molecule compounds competing for their pharmacological target: soluble epoxide hydrolase. J Pharmacol Exp Ther. 2020;374:223–32. https://doi.org/10.1124/jpet.120.265330.

    Article  CAS  PubMed  Google Scholar 

  22. Tadayasu Y, Sarashina A, Tsuda Y, Tatami S, Friedrich C, Retlich S, et al. Population pharmacokinetic/pharmacodynamic analysis of the DPP-4 inhibitor linagliptin in Japanese patients with type 2 diabetes mellitus. J Pharm Pharm Sci. 2013;16(5):708–21.

    Article  PubMed  Google Scholar 

  23. Gerlowski LE, Jain RK. Physiologically based pharmacokinetic modeling: principles and applications. J Pharm Sci. 1983;72(10):1103–27. https://doi.org/10.1002/jps.2600721003.

    Article  CAS  PubMed  Google Scholar 

  24. Bi Y, Deng J, Murry DJ, An G. A whole-body physiologically based pharmacokinetic model of gefitinib in mice and scale-up to humans. AAPS J. 2016;18(1):228–38. https://doi.org/10.1208/s12248-015-9836-3.

    Article  CAS  PubMed  Google Scholar 

  25. Fuchs H, Runge F, Held H-D. Excretion of the dipeptidyl peptidase-4 inhibitor linagliptin in rats is primarily by biliary excretion and P-gp-mediated efflux. Eur J Pharm Sci. 2012;45(5):533–8.

    Article  CAS  PubMed  Google Scholar 

  26. Sarashina A, Sesoko S, Nakashima M, Hayashi N, Taniguchi A, Horie Y, et al. Linagliptin, a dipeptidyl peptidase-4 inhibitor in development for the treatment of type 2 diabetes mellitus: a phase I, randomized, double-blind, placebo-controlled trial of single and multiple escalating doses in healthy adult male Japanese subjects. Clin Ther. 2010;32(6):1188–204.

    Article  CAS  PubMed  Google Scholar 

  27. Blech S, Ludwig-Schwellinger E, Gräfe-Mody EU, Withopf B, Wagner K. The metabolism and disposition of the oral dipeptidyl peptidase-4 inhibitor, linagliptin, in humans. Drug Metab Dispos. 2010;38(4):667–78.

    Article  CAS  PubMed  Google Scholar 

  28. Delp MD, Evans MV, Duan C. Effects of aging on cardiac output, regional blood flow, and body composition in Fischer-344 rats. J Appl Physiol. 1998;85(5):1813–22.

    Article  CAS  PubMed  Google Scholar 

  29. Nagler RM, Baum BJ, Fox PC. Effects of X irradiation on the function of rat salivary glands at 3 and 40 days. Radiat Res. 1993;136(3):392–6.

    Article  CAS  PubMed  Google Scholar 

  30. Mobarak N, Alice Y. Lead distribution in the saliva and blood fractions of rats after intraperitoneal injections. Toxicology. 1984;32(1):67–74.

    Article  CAS  PubMed  Google Scholar 

  31. Davies B, Morris T. Physiological parameters in laboratory animals and humans. Pharm Res. 1993;10(7):1093–5.

    Article  CAS  PubMed  Google Scholar 

  32. Jansky L, Hart J. Cardiac output and organ blood flow in warm-and cold-acclimated rats exposed to cold. Can J Physiol Pharmacol. 1968;46(4):653–9.

    Article  CAS  PubMed  Google Scholar 

  33. Lee H, Blaufox M. Blood volume in the rat. J Nucl Med. 1985;26(1):72–6.

    CAS  PubMed  Google Scholar 

  34. Valentin J. Basic anatomical and physiological data for use in radiological protection: reference values: ICRP publication 89. Ann ICRP. 2002;32(3–4):1–277.

    Article  Google Scholar 

  35. Feher JJ. Quantitative human physiology: an introduction: academic press; 2017.

  36. Tarhan S, Gümüs B, Gündüz İ, Ayyildiz V, Göktan C. Effect of varicocele on testicular artery blood flow in men color doppler investigation. Scand J Urol Nephrol. 2003;37(1):38–42.

    Article  PubMed  Google Scholar 

  37. Hurley PJ. Red cell and plasma volumes in normal adults. J Nucl Med. 1975;16(1):46–52.

    CAS  PubMed  Google Scholar 

  38. Choi G-W, Lee Y-B, Cho H-Y. Interpretation of non-clinical data for prediction of human pharmacokinetic parameters: in vitro-in vivo extrapolation and allometric scaling. Pharmaceutics. 2019;11(4):168.

    Article  CAS  PubMed Central  Google Scholar 

  39. Kenyon EM. Interspecies extrapolation. Computational Toxicology: Springer; 2012. 501–20.

  40. Yu Y, Fuscoe JC, Zhao C, Guo C, Jia M, Qing T, et al. A rat RNA-Seq transcriptomic body map across 11 organs and 4 developmental stages. Nat Commun. 2014;5(1):1–11.

    CAS  Google Scholar 

  41. van Waterschoot RAB, Parrott NJ, Olivares-Morales A, Lave T, Rowland M, Smith DA. Impact of target interactions on small-molecule drug disposition: an overlooked area. Nat Rev Drug Discov. 2018;17(4):299. https://doi.org/10.1038/nrd.2018.26.

    Article  CAS  PubMed  Google Scholar 

  42. An G. Small-molecule compounds exhibiting target-mediated drug disposition (TMDD): a minireview. J Clin Pharmacol. 2017;57(2):137–50.

    Article  CAS  PubMed  Google Scholar 

  43. An G, Liu W, Dutta S. Small-molecule compounds exhibiting target-mediated drug disposition—a case example of ABT-384. J Clin Pharmacol. 2015;55(10):1079–85.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work is partially supported via University of Iowa Pharmaceutics Development Consortium.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Guohua An.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Appendix

Appendix

Appendix Fig. 1
figure 6

Goodness fit plots for the final PBPK-TMDD modeling for linagliptin observed versus model-predicted in both wildtype and DPP4-deficient rat different tissues after 0.01, 0.1, 0.3, 1, 3, 10, and 50 mg/kg intravenous dose at 72 h. Yellow bars represent predicted linagliptin tissue concentration. Gray bars represent observed linagliptin tissue concentration

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, N., An, G. Incorporating Pharmacological Target-Mediated Drug Disposition (TMDD) in a Whole-Body Physiologically Based Pharmacokinetic (PBPK) Model of Linagliptin in Rat and Scale-up to Human. AAPS J 22, 125 (2020). https://doi.org/10.1208/s12248-020-00481-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-020-00481-w

KEY WORDS

Navigation