Home Role of OATP-1B1 and/or OATP-1B3 in hepatic disposition of tyrosine kinase inhibitors
Article
Licensed
Unlicensed Requires Authentication

Role of OATP-1B1 and/or OATP-1B3 in hepatic disposition of tyrosine kinase inhibitors

  • Varun Khurana , Mukul Minocha , Dhananjay Pal and Ashim K. Mitra EMAIL logo
Published/Copyright: March 18, 2014
Become an author with De Gruyter Brill

Abstract

Background: The metabolism of tyrosine kinase inhibitors (TKIs) is mainly mediated via hepatic route, but the mechanism responsible for their hepatocellular accumulation is still unknown. This study was designed to understand the contribution of organic anion transporting polypeptides (OATPs) in the hepatic uptake of selected TKIs – pazopanib, canertinib, erlotinib, vandetanib and nilotinib.

Methods: Michaelis-Menten (MM) kinetic parameters for TKIs were determined by concentration-dependent cellular accumulation of selected TKIs using Chinese hamster ovary cells – wild type as well as transfected with humanized OATP-1B1 and OATP-1B3 transporter proteins.

Results: The MM constant (Km) values of OATP-1B1 for nilotinib and vandetanib are 10.14±1.91 and 2.72±0.25 μM, respectively, and Vmax values of OATP-1B1 for nilotinib and vandetanib were 6.95±0.47 and 75.95±1.99 nmol/mg protein per minute, respectively. Likewise, Km values of OATP-1B3 for canertinib, nilotinib and vandetanib were 12.18±3.32, 7.84±1.43 and 4.37±0.79 μM, respectively, and Vmax values of OATP-1B3 for canertinib, nilotinib and vandetanib were 15.34±1.59, 6.75±0.42 and 194.64±10.58 nmol/mg protein per minute, respectively. Canertinib did not exhibit any substrate specificity toward OATP-1B1. Also, erlotinib and pazopanib did not exhibit any substrate specificity toward OATP-1B1 and -1B3.

Conclusions: Because selected TKIs are the substrates of OATP-1B1 and -1B3 expressed in hepatic tissue, these compounds can be regarded as molecular targets for transporter-mediated drug-drug interactions (DDIs). Any alteration in the function of these hepatic OATPs might account for the pharmacokinetic variability of TKIs.


Corresponding author: Ashim K. Mitra, PhD, Curators’ Professor and Chairman, Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA, Phone: +1-816-235-1615, Fax: +1-816-235-5190, E-mail:

Acknowledgments

This work was supported by National Institutes of Health grant 1R01 AI071199. The authors highly appreciate Dr. Bruno Stieger for the generous gift of CHO-WT and OATP-1B type transporter protein transfected cell lines.

Conflict of interest statement

Authors’ conflict of interest disclosure: The authors stated that there are no conflicts of interest regarding the publication of this article. Research support played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

Research funding: None declared.

Employment or leadership: None declared.

Honorarium: None declared.

References

1. Shimizu M, Fuse K, Okudaira K, Nishigaki R, Maeda K, Kusuhara H, et al. Contribution of OATP (organic anion-transporting polypeptide) family transporters to the hepatic uptake of fexofenadine in humans. Drug Metab Dispos 2005;33:1477–81.10.1124/dmd.105.004622Search in Google Scholar PubMed

2. Hagenbuch B, Meier PJ. Organic anion transporting polypeptides of the OATP/SLC21 family: phylogenetic classification as OATP/SLCO superfamily, new nomenclature and molecular/functional properties. Pflugers Arch 2004;447:653–65.10.1007/s00424-003-1168-ySearch in Google Scholar PubMed

3. Barton HA, Lai Y, Goosen TC, Jones HM, El-Kattan AF, Gosset JR, et al. Model-based approaches to predict drug-drug interactions associated with hepatic uptake transporters: preclinical, clinical and beyond. Expert Opin Drug Metab Toxicol 2013;9:459–72.10.1517/17425255.2013.759210Search in Google Scholar PubMed

4. Shitara Y, Maeda K, Ikejiri K, Yoshida K, Horie T, Sugiyama Y. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos 2013;34:45–78.10.1002/bdd.1823Search in Google Scholar PubMed

5. Zimmerman EI, Hu S, Roberts JL, Gibson AA, Orwick SJ, Li L, et al. Contribution of OATP1B1 and OATP1B3 to the disposition of sorafenib and sorafenib-glucuronide. Clin Cancer Res 2013;19:1458–66.10.1158/1078-0432.CCR-12-3306Search in Google Scholar PubMed PubMed Central

6. Pearson MA, Fabbro D. Targeting protein kinases in cancer therapy: a success? Expert Rev Anticancer Ther 2004;4:1113–24.10.1586/14737140.4.6.1113Search in Google Scholar PubMed

7. Undevia SD, Gomez-Abuin G, Ratain MJ. Pharmacokinetic variability of anticancer agents. Nat Rev Cancer 2005;5: 447–58.10.1038/nrc1629Search in Google Scholar PubMed

8. van Erp NP, Gelderblom H, Guchelaar HJ. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev 2009;35:692–706.10.1016/j.ctrv.2009.08.004Search in Google Scholar PubMed

9. Minocha M, Khurana V, Mitra AK. Determination of pazopanib (GW-786034) in mouse plasma and brain tissue by liquid chromatography-tandem mass spectrometry (LC/MS-MS). J Chromatogr B Analyt Technol Biomed Life Sci 2012;901:85–92.10.1016/j.jchromb.2012.06.004Search in Google Scholar PubMed PubMed Central

10. Minocha M, Khurana V, Qin B, Pal D, Mitra AK. Enhanced brain accumulation of pazopanib by modulating P-gp and Bcrp1 mediated efflux with canertinib or erlotinib. Int J Pharm 2012;436:127–34.10.1016/j.ijpharm.2012.05.038Search in Google Scholar PubMed PubMed Central

11. Minocha M, Khurana V, Qin B, Pal D, Mitra AK. Co-administration strategy to enhance brain accumulation of vandetanib by modulating P-glycoprotein (P-gp/Abcb1) and breast cancer resistance protein (Bcrp1/Abcg2) mediated efflux with m-TOR inhibitors. Int J Pharm 2012;434:306–14.10.1016/j.ijpharm.2012.05.028Search in Google Scholar PubMed PubMed Central

12. Gotze L, Hegele A, Metzelder SK, Renz H, Nockher WA. Development and clinical application of a LC-MS/MS method for simultaneous determination of various tyrosine kinase inhibitors in human plasma. Clin Chim Acta 2012;413:143–9.10.1016/j.cca.2011.09.012Search in Google Scholar PubMed

13. Lankheet NA, Hillebrand MJ, Rosing H, Schellens JH, Beijnen JH, Huitema AD. Method development and validation for the quantification of dasatinib, erlotinib, gefitinib, imatinib, lapatinib, nilotinib, sorafenib and sunitinib in human plasma by liquid chromatography coupled with tandem mass spectrometry. Biomed Chromatogr 2013;27:466–76.10.1002/bmc.2814Search in Google Scholar PubMed

14. Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. Br J Pharmacol 2009;158:693–705.10.1111/j.1476-5381.2009.00430.xSearch in Google Scholar PubMed PubMed Central

15. Konig J, Cui Y, Nies AT, Keppler D. Localization and genomic organization of a new hepatocellular organic anion transporting polypeptide. J Biol Chem 2000;275:23161–8.10.1074/jbc.M001448200Search in Google Scholar PubMed

16. De Bruyn T, van Westen GJ, Ijzerman AP, Stieger B, de Witte P, Augustijns PF, et al. Structure-based identification of OATP1B1/3 inhibitors. Mol Pharmacol 2013;83:1257–67.10.1124/mol.112.084152Search in Google Scholar PubMed

17. Gui C, Obaidat A, Chaguturu R, Hagenbuch B. Development of a cell-based high-throughput assay to screen for inhibitors of organic anion transporting polypeptides 1B1 and 1B3. Curr Chem Genomics 2010;4:1–8.10.2174/1875397301004010001Search in Google Scholar PubMed PubMed Central

18. Nozawa T, Minami H, Sugiura S, Tsuji A, Tamai I. Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7-ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms. Drug Metab Dispos 2005;33:434–9.10.1124/dmd.104.001909Search in Google Scholar PubMed

19. Agarwal S, Mittapalli RK, Zellmer DM, Gallardo JL, Donelson R, Seiler C, et al. Active efflux of Dasatinib from the brain limits efficacy against murine glioblastoma: broad implications for the clinical use of molecularly targeted agents. Mol Cancer Ther 2012;11:2183–92.10.1158/1535-7163.MCT-12-0552Search in Google Scholar PubMed PubMed Central

20. Agarwal S, Elmquist WF. Insight into the cooperation of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) at the blood-brain barrier: a case study examining sorafenib efflux clearance. Mol Pharm 2012;9:678–84.10.1021/mp200465cSearch in Google Scholar PubMed PubMed Central

21. Wang T, Agarwal S, Elmquist WF. Brain distribution of cediranib is limited by active efflux at the blood-brain barrier. J Pharmacol Exp Ther 2012;341:386–95.10.1124/jpet.111.190488Search in Google Scholar PubMed PubMed Central

22. Agarwal S, Sane R, Gallardo JL, Ohlfest JR, Elmquist WF. Distribution of gefitinib to the brain is limited by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2)-mediated active efflux. J Pharmacol Exp Ther 2010;334:147–55.10.1124/jpet.110.167601Search in Google Scholar PubMed PubMed Central

23. Seithel A, Eberl S, Singer K, Auge D, Heinkele G, Wolf NB, et al. The influence of macrolide antibiotics on the uptake of organic anions and drugs mediated by OATP1B1 and OATP1B3. Drug Metab Dispos 2007;35:779–86.10.1124/dmd.106.014407Search in Google Scholar PubMed

24. Backman JT, Kyrklund C, Neuvonen M, Neuvonen PJ. Gemfibrozil greatly increases plasma concentrations of cerivastatin. Clin Pharmacol Ther 2002;72:685–91.10.1067/mcp.2002.128469Search in Google Scholar PubMed

25. Backman JT, Kyrklund C, Kivisto KT, Wang JS, Neuvonen PJ. Plasma concentrations of active simvastatin acid are increased by gemfibrozil. Clin Pharmacol Ther 2000;68:122–9.10.1067/mcp.2000.108507Search in Google Scholar PubMed

26. Fahrmayr C, Fromm MF, Konig J. Hepatic OATP and OCT uptake transporters: their role for drug-drug interactions and pharmacogenetic aspects. Drug Metab Rev 2010;42:380–401.10.3109/03602530903491683Search in Google Scholar PubMed

27. Romaine SP, Bailey KM, Hall AS, Balmforth AJ. The influence of SLCO1B1 (OATP1B1) gene polymorphisms on response to statin therapy. Pharmacogenomics J 2010;10:1–11.10.1038/tpj.2009.54Search in Google Scholar PubMed

28. Hu S, Franke RM, Filipski KK, Hu C, Orwick SJ, de Bruijn EA, et al. Interaction of imatinib with human organic ion carriers. Clin Cancer Res 2008;14:3141–8.10.1158/1078-0432.CCR-07-4913Search in Google Scholar PubMed

29. Burger H, van Tol H, Brok M, Wiemer EA, de Bruijn EA, Guetens G, et al. Chronic imatinib mesylate exposure leads to reduced intracellular drug accumulation by induction of the ABCG2 (BCRP) and ABCB1 (MDR1) drug transport pumps. Cancer Biol Ther 2005;4:747–52.10.4161/cbt.4.7.1826Search in Google Scholar PubMed

30. Burger H, van Tol H, Boersma AW, Brok M, Wiemer EA, Stoter G, et al. Imatinib mesylate (STI571) is a substrate for the breast cancer resistance protein (BCRP)/ABCG2 drug pump. Blood 2004;104:2940–2.10.1182/blood-2004-04-1398Search in Google Scholar PubMed

31. Hamada A, Miyano H, Watanabe H, Saito H. Interaction of imatinib mesilate with human P-glycoprotein. J Pharmacol Exp Ther 2003;307:824–8.10.1124/jpet.103.055574Search in Google Scholar PubMed

32. Kim RB. Organic anion-transporting polypeptide (OATP) transporter family and drug disposition. Eur J Clin Invest 2003;33:Suppl:21–5.10.1046/j.1365-2362.33.s2.5.xSearch in Google Scholar PubMed

33. Duckett DR, Cameron MD. Metabolism considerations for kinase inhibitors in cancer treatment. Expert Opin Drug Metab Toxicol 2010;6:1175–93.10.1517/17425255.2010.506873Search in Google Scholar PubMed PubMed Central

34. Choi MK, Jin QR, Choi YL, Ahn SH, Bae MA, Song IS. Inhibitory effects of ketoconazole and rifampin on OAT1 and OATP1B1 transport activities: considerations on drug-drug interactions. Biopharm Drug Dispos 2011;32:175–84.10.1002/bdd.749Search in Google Scholar PubMed

35. Koenen A, Kroemer HK, Grube M, Meyer zu Schwabedissen HE. Current understanding of hepatic and intestinal OATP-mediated drug-drug interactions. Expert Rev Clin Pharmacol 2011; 2–11;4:729–42.10.1586/ecp.11.58Search in Google Scholar PubMed

36. Muller F, Fromm MF. Transporter-mediated drug-drug interactions. Pharmacogenomics 2011;12:1017–37.10.2217/pgs.11.44Search in Google Scholar PubMed

37. Niemi M. Role of OATP transporters in the disposition of drugs. Pharmacogenomics 2007;8:787–802.10.2217/14622416.8.7.787Search in Google Scholar PubMed

38. Keitel V, Burdelski M, Warskulat U, Kuhlkamp T, Keppler D, Haussinger D, et al. Expression and localization of hepatobiliary transport proteins in progressive familial intrahepatic cholestasis. Hepatology 2005;41:1160–72.10.1002/hep.20682Search in Google Scholar PubMed

39. Oswald M, Kullak-Ublick GA, Paumgartner G, Beuers U. Expression of hepatic transporters OATP-C and MRP2 in primary sclerosing cholangitis. Liver 2001;21:247–53.10.1034/j.1600-0676.2001.021004247.xSearch in Google Scholar PubMed

40. Wlcek K, Svoboda M, Riha J, Zakaria S, Olszewski U, Dvorak Z, et al. The analysis of organic anion transporting polypeptide (OATP) mRNA and protein patterns in primary and metastatic liver cancer. Cancer Biol Ther 2011;11:801–11.10.4161/cbt.11.9.15176Search in Google Scholar PubMed

41. Vavricka SR, Jung D, Fried M, Grutzner U, Meier PJ, Kullak-Ublick GA. The human organic anion transporting polypeptide 8 (SLCO1B3) gene is transcriptionally repressed by hepatocyte nuclear factor 3beta in hepatocellular carcinoma. J Hepatol 2004;40:212–8.10.1016/j.jhep.2003.10.008Search in Google Scholar PubMed

42. Cui Y, Konig J, Nies AT, Pfannschmidt M, Hergt M, Franke WW, et al. Detection of the human organic anion transporters SLC21A6 (OATP2) and SLC21A8 (OATP8) in liver and hepatocellular carcinoma. Lab Invest 2003;83:527–38.10.1097/01.LAB.0000065015.02412.48Search in Google Scholar PubMed

43. Di Gion P, Kanefendt F, Lindauer A, Scheffler M, Doroshyenko O, Fuhr U, et al. Clinical pharmacokinetics of tyrosine kinase inhibitors: focus on pyrimidines, pyridines and pyrroles. Clin Pharmacokinet 2011;50:551–603.10.2165/11593320-000000000-00000Search in Google Scholar PubMed

44. Feng B, Xu JJ, Bi YA, Mireles R, Davidson R, Duignan DB, et al. Role of hepatic transporters in the disposition and hepatotoxicity of a HER2 tyrosine kinase inhibitor CP-724,714. Toxicol Sci 2009;108:492–500.10.1093/toxsci/kfp033Search in Google Scholar PubMed

Received: 2013-11-21
Accepted: 2014-2-12
Published Online: 2014-3-18
Published in Print: 2014-9-1

© 2014 by De Gruyter

Downloaded on 24.5.2025 from https://www.degruyterbrill.com/document/doi/10.1515/dmdi-2013-0062/html
Scroll to top button