Skip to main content

Advertisement

Log in

Characterization and Validation of Canine P-Glycoprotein-Deficient MDCK II Cell Lines for Efflux Substrate Screening

  • RESEARCH PAPER
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose

We characterized three canine P-gp (cP-gp) deficient MDCKII cell lines. Their relevance for identifying efflux transporter substrates and predicting limitation of brain penetration were evaluated. In addition, we discuss how compound selection can be done in drug discovery by using these cell systems.

Method

hMDR1, hBCRP-transfected, and non-transfected MDCKII ZFN cells (all with knock-down of endogenous cP-gp) were used for measuring permeability and efflux ratios for substrates. The compounds were also tested in MDR1_Caco-2 and BCRP_Caco-2, each with a double knock-out of BCRP/MRP2 or MDR1/MRP2 transporters respectively. Efflux results were compared between the MDCK and Caco-2 models. Furthermore, in vitro MDR1_ZFN efflux data were correlated with in vivo unbound drug brain-to-plasma partition coefficient (Kp,uu).

Results

MDR1 and BCRP substrates are correctly classified and robust transporter affinities with control substrates are shown. Cell passage mildly influenced mRNA levels of transfected transporters, but the transporter activity was proven stable for several years. The MDCK and Caco-2 models were in high consensus classifying same efflux substrates. Approx. 80% of enlisted substances were correctly predicted with the MDR1_ZFN model for brain penetration.

Conclusion

cP-gp deficient MDCKII ZFN models are reliable tools to identify MDR1 and BCRP substrates and useful for predicting efflux liability for brain penetration.

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

Abbreviations

ABC:

ATP-binding cassette

API:

Active pharmaceutical ingredient

BCRP:

Breast cancer resistance protein

CNS:

Central nervous system

cP-gp:

Canine P-glycoprotein

Cpm:

Count per minute

CT :

Cycle threshold

DMEM:

Dulbecco’s modified eagle medium

DMSO:

Dimethyl sulfoxide

DPBS:

Dulbecco’s phosphate buffered saline

EDTA:

Ethylenediaminetetraacetic acid

EMA:

European medicines agency

ER:

Efflux ratio

FBS:

Fetal bovine serum

FDA:

U.S. Food and drug administration

GAPDH:

Glyceraldehyde 3-phosphate dehydrogenase

HBSS:

Hank’s balanced salt solution

MDCK:

Madin-Darby canine kidney

MDR:

Multidrug resistance

mRNA:

Messenger RNA

MRP:

Multidrug resistance associated protein

NEA:

Non-essential amino acids

NER:

Net efflux ratio

OATP:

Organic anion-transporting polypeptide

OCT:

Organic cation transporter

Papp :

Apparent permeability coefficient

RT-PCR:

Reverse transcription-polymerase chain reaction

P-gp:

P-glycoprotein

WT:

Wild type

ZFN:

Zinc finger nuclease

References

  1. Guidance for Industry: In Vitro Metabolism and Transporter Mediated Drug-Drug Interaction Studies. 2017. https://wwwfdagov/media/108130/download.

  2. Hellinger É, Veszelka S, Tóth AE, Walter F, Kittel Á, Bakk ML, et al. Comparison of brain capillary endothelial cell-based and epithelial (MDCK-MDR1, Caco-2, and VB-Caco-2) cell-based surrogate blood–brain barrier penetration models. Eur J Pharm Biopharm. 2012;82(2):340–51.

    CAS  PubMed  Google Scholar 

  3. Garberg P, Ball M, Borg N, Cecchelli R, Fenart L, Hurst R, et al. In vitro models for the blood–brain barrier. Toxicol in Vitro. 2005;19(3):299–334.

    CAS  PubMed  Google Scholar 

  4. Sai Y, Tsuji A. Transporter-mediated drug delivery: recent progress and experimental approaches. Drug Discov Today. 2004;9(16):712–20.

    CAS  PubMed  Google Scholar 

  5. Sampson KE, Brinker A, Pratt J, Venkatraman N, Xiao Y, Blasberg J, et al. Zinc finger nuclease–mediated gene knockout results in loss of transport activity for P-glycoprotein, BCRP, and MRP2 in Caco-2 cells. Drug Metab Dispos. 2015;43(2):199–207.

    PubMed  Google Scholar 

  6. Dukes JD, Whitley P, Chalmers AD. The MDCK variety pack: choosing the right strain. BMC Cell Biol. 2011;12:43.

    CAS  PubMed  Google Scholar 

  7. Kikuchi R, de Morais SM, Kalvass JC. In vitro P-glycoprotein efflux ratio can predict the in vivo brain penetration regardless of biopharmaceutics drug disposition classification system class. Drug Metab Dispos. 2013;41(12):2012–7.

    CAS  PubMed  Google Scholar 

  8. Feng B, West M, Patel NC, Wager T, Hou X, Johnson J, et al. Validation of human MDR1-MDCK and BCRP-MDCK cell lines to improve the prediction of brain penetration. J Pharm Sci. 2019;108:2476–83.

    CAS  PubMed  Google Scholar 

  9. Feng B, Doran AC, Di L, West MA, Osgood SM, Mancuso JY, et al. Prediction of human brain penetration of P-glycoprotein and breast cancer resistance protein substrates using in vitro transporter studies and animal models. J Pharm Sci. 2018;107(8):2225–35.

    CAS  PubMed  Google Scholar 

  10. Sato S, Tohyama K, Kosugi Y. Investigation of MDR1-overexpressing cell lines to derive a quantitative prediction approach for brain disposition using in vitro efflux activities. Eur J Pharm Sci. 2019;105119

  11. Gartzke D, Delzer J, Laplanche L, Uchida Y, Hoshi Y, Tachikawa M, et al. Genomic knockout of endogenous canine P-glycoprotein in wild-type, human P-glycoprotein and human BCRP transfected MDCKII cell lines by zinc finger nucleases. Pharm Res. 2015;32(6):2060–71.

    CAS  PubMed  Google Scholar 

  12. Simoff I, Karlgren M, Backlund M, Lindström A-C, Gaugaz FZ, Matsson P, et al. Complete knockout of endogenous Mdr1 (Abcb1) in MDCK cells by CRISPR-Cas9. J Pharm Sci. 2016;105(2):1017–21.

    CAS  PubMed  Google Scholar 

  13. Karlgren M, Simoff I, Backlund M, Wegler C, Keiser M, Handin N, et al. A CRISPR-Cas9 generated MDCK cell line expressing human MDR1 without endogenous canine MDR1 (cABCB1): an improved tool for drug efflux studies. J Pharm Sci. 2017;106(9):2909–13.

    CAS  PubMed  Google Scholar 

  14. Chen EC, Broccatelli F, Plise E, Chen B, Liu L, Cheong J, et al. Evaluating the utility of canine Mdr1 knockout Madin-Darby canine kidney I cells in permeability screening and efflux substrate determination. Mol Pharm. 2018;15(11):5103–13.

    CAS  PubMed  Google Scholar 

  15. Saeki T, Ueda K, Tanigawara Y, Hori R, Komano T. P-glycoprotein-mediated transcellular transport of MDR-reversing agents. FEBS Lett. 1993;324(1):99–102.

    CAS  PubMed  Google Scholar 

  16. Polli JW, Wring SA, Humphreys JE, Huang L, Morgan JB, Webster LO, et al. Rational use of in vitro P-glycoprotein assays in drug discovery. J Pharmacol Exp Ther. 2001;299(2):620–8.

    CAS  PubMed  Google Scholar 

  17. Haslam IS, Jones K, Coleman T, Simmons N. Induction of P-glycoprotein expression and function in human intestinal epithelial cells (T84). Biochem Pharmacol. 2008;76(7):850–61.

    CAS  PubMed  Google Scholar 

  18. Wang Q, Strab R, Kardos P, Ferguson C, Li J, Owen A, et al. Application and limitation of inhibitors in drug–transporter interactions studies. Int J Pharm. 2008;356(1–2):12–8.

    CAS  PubMed  Google Scholar 

  19. Sane R, Agarwal S, Mittapalli RK, Elmquist WF. Saturable active efflux by p-glycoprotein and breast cancer resistance protein at the blood-brain barrier leads to nonlinear distribution of elacridar to the central nervous system. J Pharmacol Exp Ther. 2013;345(1):111–24.

    CAS  PubMed  Google Scholar 

  20. Wang Z, Pal D, Patel A, Kwatra D, Mitra AK. Influence of overexpression of efflux proteins on the function and gene expression of endogenous peptide transporters in MDR-transfected MDCKII cell lines. Int J Pharm. 2013;441(1–2):40–9.

    CAS  PubMed  Google Scholar 

  21. Volpe DA. Variability in Caco-2 and MDCK cell-based intestinal permeability assays. J Pharm Sci. 2008;97(2):712–25.

    CAS  PubMed  Google Scholar 

  22. Heikkinen AT, Korjamo T, Lepikkö V, Mönkkönen J. Effects of experimental setup on the apparent concentration dependency of active efflux transport in in vitro cell permeation experiments. Mol Pharm. 2010;7(2):605–17.

    CAS  PubMed  Google Scholar 

  23. Wright JA, Haslam IS, Coleman T, Simmons NL. Breast cancer resistance protein BCRP (ABCG2)-mediated transepithelial nitrofurantoin secretion and its regulation in human intestinal epithelial (Caco-2) layers. Eur J Pharmacol. 2011;672(1–3):70–6.

    CAS  PubMed  Google Scholar 

  24. Cassio D. Long term culture of MDCK strains alters chromosome content. BMC Res Notes. 2013;6(1):162.

    CAS  PubMed  Google Scholar 

  25. Lewin B. Genes IV. Am J Phys Anthropol. New York: Oxford University Press; 1990. 

  26. Artursson P, Karlsson J. Correlation between oral drug absorption in humans and apparent drug permeability coefficients in human intestinal epithelial (Caco-2) cells. Biochem Biophys Res Commun. 1991;175(3):880–5.

    CAS  PubMed  Google Scholar 

  27. Lennernäs H, Palm K, Fagerholm U, Artursson P. Comparison between active and passive drug transport in human intestinal epithelial (Caco-2) cells in vitro and human jejunum in vivo. Int J Pharm. 1996;127(1):103–7.

    Google Scholar 

  28. Larregieu CA, Benet LZ. Drug discovery and regulatory considerations for improving in silico and in vitro predictions that use Caco-2 as a surrogate for human intestinal permeability measurements. AAPS J. 2013;15(2):483–97.

    CAS  PubMed  Google Scholar 

  29. Li J, Volpe DA, Wang Y, Zhang W, Bode C, Owen A, et al. Use of transporter knockdown Caco-2 cells to investigate the in vitro efflux of statin drugs. Drug Metab Dispos. 2011;39(7):1196–202.

    CAS  PubMed  Google Scholar 

  30. Huang L, Wang Y, Grimm S. ATP-dependent transport of rosuvastatin in membrane vesicles expressing breast cancer resistance protein. Drug Metab Dispos. 2006;34(5):738–42.

    CAS  PubMed  Google Scholar 

  31. Li J, Wang Y, Zhang W, Huang Y, Hein K, Hidalgo IJ. The role of a basolateral transporter in rosuvastatin transport and its interplay with apical breast cancer resistance protein in polarized cell monolayer systems. Drug Metab Dispos. 2012;40(11):2102–8.

    CAS  PubMed  Google Scholar 

  32. Hirohashi T, Suzuki H, Chu X-Y, Tamai I, Tsuji A, Sugiyama Y. Function and expression of multidrug resistance-associated protein family in human colon adenocarcinoma cells (Caco-2). J Pharmacol Exp Ther. 2000;292(1):265–70.

    CAS  PubMed  Google Scholar 

  33. Gartzke D, Fricker G. Establishment of optimized MDCK cell lines for reliable efflux transport studies. J Pharm Sci. 2014;103(4):1298–304.

    CAS  PubMed  Google Scholar 

  34. Schinkel AH, Wagenaar E, van Deemter L, Mol C, Borst P. Absence of the mdr1a P-Glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A. J Clin Invest. 1995;96(4):1698–705.

    CAS  PubMed  Google Scholar 

  35. Van Asperen J, Schinkel AH, Beijnen JH, Nooijen WJ, Borst P, van Tellingen O. Altered pharmacokinetics of vinblastine in Mdr1a P-glycoprotein-deficient mice. JNCI: J Natl Cancer Inst. 1996;88(14):994–9.

    PubMed  Google Scholar 

  36. Evans DC, O’Connor D, Lake BG, Evers R, Allen C, Hargreaves R. Eletriptan metabolism by human hepatic CYP450 enzymes and transport by human P-glycoprotein. Drug Metab Dispos. 2003;31(7):861–9.

    CAS  PubMed  Google Scholar 

  37. Schinkel A, Smit J, Van Tellingen M, Beijnen J, Wagenaar E, Van Deemter L, et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell. 1994;77(4):491–502.

    CAS  PubMed  Google Scholar 

  38. Kemper EM, Cleypool C, Boogerd W, Beijnen JH, van Tellingen O. The influence of the P-glycoprotein inhibitor zosuquidar trihydrochloride (LY335979) on the brain penetration of paclitaxel in mice. Cancer Chemother Pharmacol. 2004;53(2):173–8.

    CAS  PubMed  Google Scholar 

  39. Kusuhara H, Suzuki H, Terasaki T, Kakee A, Lemaire M, Sugiyama Y. P-glycoprotein mediates the efflux of quinidine across the blood-brain barrier. J Pharmacol Exp Ther. 1997;283(2):574–80.

    CAS  PubMed  Google Scholar 

  40. Hendrikse N, Schinkel A, De Vries E, Fluks E, Van der Graaf W, Willemsen A, et al. Complete in vivo reversal of P-glycoprotein pump function in the blood-brain barrier visualized with positron emission tomography. Br J Pharmacol. 1998;124(7):1413–8.

    CAS  PubMed  Google Scholar 

  41. Pereira JNDS, Tadjerpisheh S, Abed MA, Saadatmand AR, Weksler B, Romero IA, et al. The poorly membrane permeable antipsychotic drugs amisulpride and sulpiride are substrates of the organic cation transporters from the SLC22 family. AAPS J. 2014;16(6):1247–58.

    Google Scholar 

  42. Wang J-S, Ruan Y, Taylor RM, Donovan JL, Markowitz JS, DeVane CL. The brain entry of risperidone and 9-hydroxyrisperidone is greatly limited by P-glycoprotein. Int J Neuropsychopharmacol. 2004;7(4):415–9.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgement

The authors would like to thank Juliane Hoeckels-Messemer, Sylvia Hellwig and Patricia Muschong of AbbVie for their technical support.

Disclosures

Dong Ye, Manuel Weinheimer, Loic Laplanche, Mario Mezler are employees of AbbVie and may own AbbVie stock. Zhizhou Fang was an employee of AbbVie at the time when he contributed to the work being discussed, and he currently works at Merck Healthcare KGaA, Germany. Anna Harder was a master student of AbbVie for this work at the time and currently works at Braun Melsungen AG, Germany. The design, study conduct, and financial support for this research was provided by AbbVie. AbbVie participated in the interpretation of data, review, writing and approval of the publication. All authors declare no competing financial interest in this work.

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by Dong Ye, Anna Harder, Zhizhou Fang. The first draft of the manuscript was written by Dong Ye and all authors commented on previous versions of the manuscript. All authors actively contributed to the final text, read and approved the final manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mario Mezler.

Additional information

Publisher’s Note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ye, D., Harder, A., Fang, Z. et al. Characterization and Validation of Canine P-Glycoprotein-Deficient MDCK II Cell Lines for Efflux Substrate Screening. Pharm Res 37, 194 (2020). https://doi.org/10.1007/s11095-020-02895-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11095-020-02895-9

Key Words

Navigation