Abstract
Background: The potential of tyrosine kinase inhibitors (TKIs) interacting with other therapeutics through hepatic uptake transporter inhibition has not been fully delineated in drug-drug interactions (DDIs). This study was designed to estimate the half-maximal inhibitory concentration (IC50) values of five small-molecule TKIs (pazopanib, nilotinib, vandetanib, canertinib and erlotinib) interacting with organic anion-transporting polypeptides (OATPs): OATP-1B1 and -1B3.
Methods: The IC50 values of TKIs and rifampicin (positive control) were determined by concentration-dependent inhibition of TKIs on cellular accumulation of radiolabeled probe substrates [3H]estrone sulfate and [3H]cholecystokinin octapeptide. Chinese hamster ovary cells transfected with humanized OATP-1B1 and OATP-1B3 transporter proteins, respectively, were utilized to carry out these studies.
Results: Pazopanib and nilotinib show inhibitory activity on OATP-1B1 transporter protein. IC50 values for rifampicin, pazopanib and nilotinib were 10.46±1.15, 3.89±1.21 and 2.78±1.13 μM, respectively, for OATP-1B1 transporter. Vandetanib, canertinib and erlotinib did not exhibit any inhibitory potency toward OATP-1B1 transporter protein. Only vandetanib expressed inhibitory potential toward OATP-1B3 transporter protein out of the five selected TKIs. IC50 values for rifampicin and vandetanib for OATP-1B3 transporter inhibition were 3.67±1.20 and 18.13±1.21 μM, respectively. No significant inhibition in the presence of increasing concentrations of pazopanib, nilotinib, canertinib and erlotinib were observed for OATP-1B3 transporter.
Conclusions: Because selected TKIs are inhibitors of OATP-1B1 and -1B3 expressed in hepatic tissue, these compounds can be regarded as molecular targets for transporter-mediated DDIs. These findings provide the basis for further preclinical and clinical studies investigating the transporter-based DDI potential of TKIs.
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 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 funding: None declared.
Employment or leadership: None declared.
Honorarium: None declared.
References
1. Minematsu T, Giacomini KM. Interactions of tyrosine kinase inhibitors with organic cation transporters and multidrug and toxic compound extrusion proteins. Mol Cancer Ther 2011;10:531–9.10.1158/1535-7163.MCT-10-0731Suche in Google Scholar PubMed PubMed Central
2. Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, et al. Membrane transporters in drug development. Nat Rev Drug Discov 2010;9:215–36.10.1038/nrd3028Suche in Google Scholar PubMed PubMed Central
3. Hartmann JT, Haap M, Kopp HG, Lipp HP. Tyrosine kinase inhibitors – a review on pharmacology, metabolism and side effects. Curr Drug Metab 2009;10:470–81.10.2174/138920009788897975Suche in Google Scholar PubMed
4. Undevia SD, Gomez-Abuin G, Ratain MJ. Pharmacokinetic variability of anticancer agents. Nat Rev Cancer 2005;5:447–58.10.1038/nrc1629Suche 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-12-3306Suche in Google Scholar PubMed PubMed Central
6. van Erp NP, Gelderblom H, Guchelaar HJ. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev 2009;35:692–706.10.1016/j.ctrv.2009.08.004Suche in Google Scholar PubMed
7. Teo YL, Ho HK, Chan A. Risk of tyrosine kinase inhibitors-induced hepatotoxicity in cancer patients: a meta-analysis. Cancer Treat Rev 2013;39:199–206.10.1016/j.ctrv.2012.09.004Suche in Google Scholar PubMed
8. Xu CF, Reck BH, Xue Z, Huang L, Baker KL, Chen M, et al. Pazopanib-induced hyperbilirubinemia is associated with Gilbert’s syndrome UGT1A1 polymorphism. Br J Cancer 2010;102: 1371–7.10.1038/sj.bjc.6605653Suche in Google Scholar PubMed PubMed Central
9. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 2010;28:1061–8.10.1200/JCO.2009.23.9764Suche in Google Scholar PubMed
10. Swift B, Nebot N, Lee JK, Han T, Proctor WR, Thakker DR, et al. Sorafenib hepatobiliary disposition: mechanisms of hepatic uptake and disposition of generated metabolites. Drug Metab Dispos 2013;41:1179–86.10.1124/dmd.112.048181Suche in Google Scholar PubMed PubMed Central
11. 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-07-4913Suche in Google Scholar PubMed
12. 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.2012.05.038Suche in Google Scholar PubMed PubMed Central
13. 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.2012.05.028Suche in Google Scholar PubMed PubMed Central
14. Khurana V, Minocha M, Pal D, Mitra AK. Role of OATP-1B1 and/or OATP-1B3 in hepatic disposition of tyrosine kinase inhibitors. Drug Metab Drug Interact 2014. Epub ahead of print. doi: 10.1515/dmdi-2013-0062.10.1515/dmdi-2013-0062Suche in Google Scholar PubMed PubMed Central
15. Karlgren M, Vildhede A, Norinder U, Wisniewski JR, Kimoto E, Lai Y, et al. Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions. J Med Chem 2012;55:4740–63.10.1021/jm300212sSuche in Google Scholar PubMed PubMed Central
16. Shitara Y. Clinical importance of OATP1B1 and OATP1B3 in drug-drug interactions. Drug Metab Pharmacokinet 2011;26:220–7.10.2133/dmpk.DMPK-10-RV-094Suche in Google Scholar PubMed
17. Kumar R, Knick VB, Rudolph SK, Johnson JH, Crosby RM, Crouthamel MC, et al. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol Cancer Ther 2007;6:2012–21.10.1158/1535-7163.MCT-07-0193Suche in Google Scholar PubMed
18. Hu S, Niu H, Inaba H, Orwick S, Rose C, Panetta JC, et al. Activity of the multikinase inhibitor sorafenib in combination with cytarabine in acute myeloid leukemia. J Natl Cancer Inst 2011;103:893–905.10.1093/jnci/djr107Suche in Google Scholar PubMed PubMed Central
19. Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. Br J Pharmacol 2009;158:693–705.10.1111/j.1476-5381.2009.00430.xSuche in Google Scholar PubMed PubMed Central
20. 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.M001448200Suche in Google Scholar PubMed
21. 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.112.084152Suche in Google Scholar
22. 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;4:1–8.10.2174/1875397301004010001Suche in Google Scholar
23. 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.104.001909Suche in Google Scholar
24. Hu S, Mathijssen RH, de Bruijn P, Baker SD, Sparreboom A. Inhibition of OATP1B1 by tyrosine kinase inhibitors: in vitro-in vivo correlations. Br J Cancer 2014;110:894–8.10.1038/bjc.2013.811Suche in Google Scholar
25. Hagenbuch B, Meier PJ. The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta 2003;1609:1–18.10.1016/S0005-2736(02)00633-8Suche in Google Scholar
26. 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-ySuche in Google Scholar PubMed
27. Smith NF, Figg WD, Sparreboom A. Role of the liver-specific transporters OATP1B1 and OATP1B3 in governing drug elimination. Expert Opin Drug Metab Toxicol 2005;1:429–45.10.1517/17425255.1.3.429Suche in Google Scholar PubMed
28. Abe T, Unno M, Onogawa T, Tokui T, Kondo TN, Nakagomi R, et al. LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers. Gastroenterology 2001;120:1689–99.10.1053/gast.2001.24804Suche in Google Scholar PubMed
29. Muto M, Onogawa T, Suzuki T, Ishida T, Rikiyama T, Katayose Y, et al. Human liver-specific organic anion transporter-2 is a potent prognostic factor for human breast carcinoma. Cancer Sci 2007;98:1570–6.10.1111/j.1349-7006.2007.00570.xSuche in Google Scholar PubMed
30. Smith NF, Acharya MR, Desai N, Figg WD, Sparreboom A. Identification of OATP1B3 as a high-affinity hepatocellular transporter of paclitaxel. Cancer Biol Ther 2005;4:815–8.10.4161/cbt.4.8.1867Suche in Google Scholar PubMed
31. de Graan AJ, Lancaster CS, Obaidat A, Hagenbuch B, Elens L, Friberg LE, et al. Influence of polymorphic OATP1B-type carriers on the disposition of docetaxel. Clin Cancer Res 2012;18: 4433–40.10.1158/1078-0432.CCR-12-0761Suche in Google Scholar
32. Yamazaki M, Suzuki H, Sugiyama Y. Recent advances in carrier-mediated hepatic uptake and biliary excretion of xenobiotics. Pharm Res 1996;13:497–513.10.1023/A:1016077517241Suche in Google Scholar
33. Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci 2006;27:425–46.10.1016/j.ejps.2005.12.003Suche in Google Scholar
34. Asberg A, Hartmann A, Fjeldsa E, Bergan S, Holdaas H. Bilateral pharmacokinetic interaction between cyclosporine A and atorvastatin in renal transplant recipients. Am J Transplant 2001;1:382–6.10.1034/j.1600-6143.2001.10415.xSuche in Google Scholar
35. Hedman M, Neuvonen PJ, Neuvonen M, Holmberg C, Antikainen M. Pharmacokinetics and pharmacodynamics of pravastatin in pediatric and adolescent cardiac transplant recipients on a regimen of triple immunosuppression. Clin Pharmacol Ther 2004;75:101–9.10.1016/j.clpt.2003.09.011Suche in Google Scholar
36. Nakagomi-Hagihara R, Nakai D, Tokui T, Abe T, Ikeda T. Gemfibrozil and its glucuronide inhibit the hepatic uptake of pravastatin mediated by OATP1B1. Xenobiotica 2007;37:474–86.10.1080/00498250701278442Suche in Google Scholar
37. Kyrklund C, Backman JT, Neuvonen M, Neuvonen PJ. Gemfibrozil increases plasma pravastatin concentrations and reduces pravastatin renal clearance. Clin Pharmacol Ther 2003;73: 538–44.10.1016/S0009-9236(03)00052-3Suche in Google Scholar
38. Whitfield LR, Porcari AR, Alvey C, Abel R, Bullen W, Hartman D. Effect of gemfibrozil and fenofibrate on the pharmacokinetics of atorvastatin. J Clin Pharmacol 2011;51:378–88.10.1177/0091270010366446Suche in Google Scholar PubMed
39. 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.1823Suche in Google Scholar PubMed
40. Kim RB. Organic anion-transporting polypeptide (OATP) transporter family and drug disposition. Eur J Clin Invest 2003;33:Suppl 2:1–5.10.1046/j.1365-2362.33.s2.5.xSuche in Google Scholar PubMed
41. Tan AR, Dowlati A, Jones SF, Infante JR, Nishioka J, Fang L, et al. Phase I study of pazopanib in combination with weekly paclitaxel in patients with advanced solid tumors. Oncologist 2010;15:1253–61.10.1634/theoncologist.2010-0095Suche in Google Scholar PubMed PubMed Central
42. Jang SH, Wientjes MG, Au JL. Kinetics of P-glycoprotein-mediated efflux of paclitaxel. J Pharmacol Exp Ther 2001;298:1236–42.Suche in Google Scholar
43. Kemper EM, van Zandbergen AE, Cleypool C, Mos HA, Boogerd W, Beijnen JH, et al. Increased penetration of paclitaxel into the brain by inhibition of P-Glycoprotein. Clin Cancer Res 2003;9:2849–55.Suche in Google Scholar
44. 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;42:380–401.10.3109/03602530903491683Suche in Google Scholar PubMed
45. Singer JB, Shou Y, Giles F, Kantarjian HM, Hsu Y, Robeva AS, et al. UGT1A1 promoter polymorphism increases risk of nilotinib-induced hyperbilirubinemia. Leukemia 2007;21:2311–5.10.1038/sj.leu.2404827Suche in Google Scholar PubMed
©2014 by De Gruyter
Artikel in diesem Heft
- Frontmatter
- Editorial
- CYP 2C19 and UDP-glucuronosyltransferases not only for drugs but also for endobiotics
- Review
- Molecular functionality of CYP2C9 polymorphisms and their influence on drug therapy
- Reviews in Population Pharmacogenomics
- Pharmacogenetics in Jewish populations
- Frequency of CYP450 enzyme gene polymorphisms in the Greek population: review of the literature, original findings and clinical significance
- Original Articles
- Inhibition of OATP-1B1 and OATP-1B3 by tyrosine kinase inhibitors
- Effect of silymarin pretreatment on the bioavailability of domperidone in healthy human volunteers
- In vitro inhibitory effects of herbal supplements on tamoxifen and irinotecan metabolism
- Acknowledgment
- Acknowledgment
Artikel in diesem Heft
- Frontmatter
- Editorial
- CYP 2C19 and UDP-glucuronosyltransferases not only for drugs but also for endobiotics
- Review
- Molecular functionality of CYP2C9 polymorphisms and their influence on drug therapy
- Reviews in Population Pharmacogenomics
- Pharmacogenetics in Jewish populations
- Frequency of CYP450 enzyme gene polymorphisms in the Greek population: review of the literature, original findings and clinical significance
- Original Articles
- Inhibition of OATP-1B1 and OATP-1B3 by tyrosine kinase inhibitors
- Effect of silymarin pretreatment on the bioavailability of domperidone in healthy human volunteers
- In vitro inhibitory effects of herbal supplements on tamoxifen and irinotecan metabolism
- Acknowledgment
- Acknowledgment