Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 51, 2021 - Issue 4
496
Views
3
CrossRef citations to date
0
Altmetric
Xenobiotic Transporters

Differential in vitro interactions of the Janus kinase inhibitor ruxolitinib with human SLC drug transporters

, , , , &
Pages 467-478 | Received 18 Dec 2020, Accepted 10 Jan 2021, Published online: 02 Feb 2021

References

  • Agarwal, S., Chinn, L., and Zhang, L., 2013. An overview of transporter information in package inserts of recently approved new molecular entities. Pharmaceutical research, 30 (3), 899–910.
  • Aitken, A.E., Richardson, T.A., and Morgan, E.T., 2006. Regulation of drug-metabolizing enzymes and transporters in inflammation. Annual review of pharmacology and toxicology, 46 (1), 123–149.
  • Alim, K., et al., 2020. Interactions of Janus kinase inhibitors with drug transporters and consequences for pharmacokinetics and toxicity. Expert opinion on drug metabolism & toxicology, Epub ahead of print. doi:10.1080/17425255.2021.1862084
  • Belzer, M., et al., 2013. Substrate-dependent ligand inhibition of the human organic cation transporter OCT2. Journal of pharmacology and experimental therapeutics, 346 (2), 300–310.
  • Bentz, J., et al., 2013. Variability in P-glycoprotein inhibitory potency (IC50) using various in vitro experimental systems: implications for universal digoxin drug-drug interaction risk assessment decision criteria. Drug metabolism and disposition, 41 (7), 1347–1366.
  • Berg, T., et al., 2018. Expression of MATE1, P-gp, OCTN1 and OCTN2, in epithelial and immune cells in the lung of COPD and healthy individuals. Respiratory research, 19 (1), 68.
  • Bradford, M.M., 1976. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Analytical biochemistry, 72 (1-2), 248–254.
  • Burckhardt, G., 2012. Drug transport by organic anion transporters (OATs). Pharmacology & therapeutics, 136 (1), 106–130.
  • Center for Drug Evaluation and Research, 2011. Data FDA ruxolitinib transporters [online]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/202192Orig1s000ClinPharmR.pdf [Accessed 27 April 2020].
  • Chedik, L., et al., 2017. Inhibition of human drug transporter activities by the pyrethroid pesticides allethrin and tetramethrin. PLoS One, 12 (1), e0169480.
  • Ebert, C., et al., 2016. Expression and function of ABC-transporter protein ABCB1 correlates with inhibitory capacity of ruxolitinib in vitro and in vivo. Haematologica, 101 (3), e81–e85.
  • Ekaratanawong, S., et al., 2004. Human organic anion transporter 4 is a renal apical organic anion/dicarboxylate exchanger in the proximal tubules. Journal of pharmacological sciences, 94 (3), 297–304.
  • Fardel, O., et al., 2015. Nature and uses of fluorescent dyes for drug transporter studies. Expert opinion on drug metabolism & toxicology, 11 (8), 1233–1251.
  • Febvre-James, M., et al., 2018. The JAK1/2 inhibitor ruxolitinib reverses interleukin-6-mediated suppression of drug detoxifying proteins in cultured human hepatocytes. Drug metabolism and disposition, 46 (2), 131–140.
  • Febvre-James, M., Lecureur, V., and Fardel, O., 2020. Potent repression of C-reactive protein (CRP) expression by the JAK1/2 inhibitor ruxolitinib in inflammatory human hepatocytes. Inflammation research, 69 (1), 51–62.
  • Febvre-James, M., et al., 2018. Repression of interferon β-regulated cytokines by the JAK1/2 inhibitor ruxolitinib in inflammatory human macrophages. International immunopharmacology, 54, 354–365.
  • Gay, C., Toulet, D., and Le Corre, P., 2017. Pharmacokinetic drug-drug interactions of tyrosine kinase inhibitors: A focus on cytochrome P450, transporters, and acid suppression therapy: Pharmacokinetic interactions of TKIs. Hematological oncology, 35 (3), 259–280.
  • Gehringer, M., et al., 2014. Novel hinge-binding motifs for Janus kinase 3 inhibitors: a comprehensive structure-activity relationship study on tofacitinib bioisosteres. Chemmedchem, 9 (11), 2516–2527.
  • Gessner, A., König, J., and Fromm, M.F., 2019. Clinical aspects of transporter-mediated drug-drug interactions. Clinical pharmacology and therapeutics, 105 (6), 1386–1394.
  • Harrach, S., et al., 2016. MATE1 regulates cellular uptake and sensitivity to imatinib in CML patients. Blood cancer journal, 6 (9), e470.
  • Hu, S., et al., 2014. Inhibition of OATP1B1 by tyrosine kinase inhibitors: in vitro-in vivo correlations. British journal of cancer, 110 (4), 894–898.
  • Izumi, S., et al., 2013. Substrate-dependent inhibition of organic anion transporting polypeptide 1b1: comparative analysis with prototypical probe substrates estradiol-17β-glucuronide, estrone-3-sulfate, and sulfobromophthalein. Drug metabolism disposition, 41 (10), 1859–1866.
  • Jouan, E., et al., 2014. The mitochondrial fluorescent dye rhodamine 123 is a high-affinity substrate for organic cation transporters (OCTs) 1 and 2. Fundamental & clinical pharmacology, 28 (1), 65–77.
  • Koepsell, H., 2020. Organic cation transporters in health and disease. Pharmacological reviews, 72 (1), 253–319.
  • Kusakizako, T., et al., 2020. Structural biology of the multidrug and toxic compound extrusion superfamily transporters. Biochimica et biophysica Acta (BBA) - biomembranes, 1862 (12), 183154.
  • Le Vée, M., et al., 2019. Neonicotinoid pesticides poorly interact with human drug transporters. Journal of biochemical and molecular toxicology, 33 (10), e22379.
  • Le Vée, M., et al., 2020. Janus kinase-dependent regulation of drug detoxifying protein expression by interleukin-22 in human hepatic cells. International immunopharmacology, 83, 106439.
  • Le Vée, M., et al., 2015., Analysis of sinusoidal drug uptake transporter activities in primary human hepatocytes. In: M. Vinken and V. Rogiers, eds. Protocols in in vitro hepatocyte research. New York, NY: Springer, 287–302.
  • Malavolta, M., et al., 2020. Exploring the relevance of senotherapeutics for the current SARS-CoV-2 emergency and similar future global health threats. Cells, 9 (4), 909.
  • Martínez-Guerrero, L.J., and Wright, S.H., 2013. Substrate-dependent inhibition of human MATE1 by cationic ionic liquids. Journal of pharmacology and experimental therapeutics, 346 (3), 495–503.
  • Morrissey, K.M., et al., 2013. Renal transporters in drug development. Annual review of pharmacology and toxicology, 53 (1), 503–529.
  • Müller, F., et al., 2013. Role of organic cation transporter OCT2 and multidrug and toxin extrusion proteins MATE1 and MATE2-K for transport and drug interactions of the antiviral lamivudine. Biochemical pharmacology, 86 (6), 808–815.
  • Nies, A.T., et al., 2016. Structure and function of multidrug and toxin extrusion proteins (MATEs) and their relevance to drug therapy and personalized medicine. Archives of toxicology, 90 (7), 1555–1584.
  • Nies, A.T., et al., 2011. Proton pump inhibitors inhibit metformin uptake by organic cation transporters (OCTs). PLoS one, 6 (7), e22163.
  • Nies, A.T., et al., 2009. Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver. Hepatology, 50 (4), 1227–1240.
  • Ogama, Y., et al., 2013. A randomized dose-escalation study to assess the safety, tolerability, and pharmacokinetics of ruxolitinib (INC424) in healthy Japanese volunteers. International journal of hematology, 97 (3), 351–359.
  • Pedersen, J.M., et al., 2017. Substrate and method dependent inhibition of three ABC-transporters (MDR1, BCRP, and MRP2). European journal of pharmaceutical sciences, 103, 70–76.
  • Przepiorka, D., et al., 2020. FDA approval summary: ruxolitinib for treatment of steroid‐refractory acute graft‐versus‐host disease. The oncologist, 25 (2), 2019–0627.
  • Quintás-Cardama, A., et al., 2011. Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond. Nature reviews drug discovery, 10 (2), 127–140.
  • Roskoski, R., 2016. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacological research, 111, 784–803.
  • Sandoval, P.J., et al., 2018. Assessment of substrate-dependent ligand interactions at the organic cation transporter oct2 using six model substrates. Molecular pharmacology, 94 (3), 1057–1068.
  • Sayyed, K., et al., 2019. Inhibition of organic cation transporter (OCT) activities by carcinogenic heterocyclic aromatic amines. Toxicology in vitro, 54, 10–22.
  • Schmitt, C., et al., 2011. Disease-drug-drug interaction involving tocilizumab and simvastatin in patients with rheumatoid arthritis. Clinical pharmacology and therapeutics, 89 (5), 735–740.
  • Schwartz, D.M., et al., 2017. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nature reviews drug discovery, 16 (12), 843–862.
  • Shi, J., et al., 2015. Predicting drug-drug interactions involving multiple mechanisms using physiologically based pharmacokinetic modeling: a case study with ruxolitinib. Clinical pharmacology and therapeutics, 97 (2), 177–185.
  • Shi, J.G., et al., 2012. The effect of CYP3A4 inhibition or induction on the pharmacokinetics and pharmacodynamics of orally administered ruxolitinib (INCB018424 Phosphate) in healthy volunteers. Journal of clinical pharmacology, 52 (6), 809–818.
  • Shi, J.G., et al., 2011. The pharmacokinetics, pharmacodynamics, and safety of orally dosed INCB018424 phosphate in healthy volunteers. Journal of clinical pharmacology, 51 (12), 1644–1654.
  • Shilling, A.D., et al., 2010. Metabolism, excretion, and pharmacokinetics of [14C]INCB018424, a selective Janus tyrosine kinase 1/2 inhibitor, in humans. Drug metabolism and disposition, 38 (11), 2023–2031.
  • Sprowl, J.A., et al., 2016. A phosphotyrosine switch regulates organic cation transporters. Nature communications, 7 (1), 10880.
  • The International Transporter Consortium, 2010. Membrane transporters in drug development. Nature reviews drug discovery, 9 (3), 215–236.
  • U.S. Department of Health and Human Services FDA, 2017. In vitro metabolism- and transporter- mediated drug-drug interaction studies guidance for industry [online]. Available from: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/vitro-drug-interaction-studies-cytochrome-p450-enzyme-and-transporter-mediated-drug-interactions [Accessed 17 December 2020].
  • U.S. Department of Health and Human Services FDA, 2020. Guidance for Industry [online]. Available from: https://www.fda.gov/media/134582/download [Accessed 17 December 2020].
  • Umehara, K., et al., 2019. Drug-drug interaction (DDI) assessments of ruxolitinib, a dual substrate of CYP3A4 and CYP2C9, using a verified physiologically based pharmacokinetic (PBPK) model to support regulatory submissions. Drug metabolism and personalized therapy, 34 (2).doi:10.1515/dmpt-2018-0042
  • Vainchenker, W., et al., 2018. JAK inhibitors for the treatment of myeloproliferative neoplasms and other disorders. F1000research, 7, 82.
  • Wang, Z.-J., et al., 2008. OCT2 polymorphisms and in-vivo renal functional consequence: studies with metformin and cimetidine. Pharmacogenetics and genomics, 18 (7), 637–645.
  • Zamek-Gliszczynski, M.J., et al., 2018. Transporters in drug development: 2018 ITC recommendations for transporters of emerging clinical importance. Clinical pharmacology and therapeutics, 104 (5), 890–899.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.