Stephanie van Hoppe

117 Brain accumulation of ponatinib and its active metabolite is limited by ABCB1 and ABCG2 R E F E R E N C E S 1. Center for Drug valuation and Research of the U.S., Department of Health and Human Services Food and Drug Administration, 203469Orig1s000 . 2012; Available from: http://www.accessdata.fda.gov/drugsatfda_ docs/nda/2012/203469Orig1s000ClinPharmR.pdf. 2. European Medicines Agency, Science Medicines Health, Assessment report Iclusig . 2013; Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/ human/002695/WC500145648.pdf. 3. FDA Drug Safety Communication. U.S. Food and Drug Administration, “FDA asks manufacturer of the leukemia drug Iclusig (ponatinib) to suspend marketing and sales”. . 2013; Available from: http://www.fda. gov/Drugs/DrugSafety/ucm373040.htm. 4. FDA Drug Safety Communication. U.S. Food and Drug Administration, “Iclusig (Ponatinib): Drug Safety Communication - Increased Reports Of Serious Blood Clots In Arteries And Veins” 2013; Available from: http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ ucm370971.htm. 5. O’Hare, T., et al., AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell, 2009. 16(5): p. 401-12. 6. Gozgit, J.M., et al., Ponatinib (AP24534), a multitargeted pan-FGFR inhibitor with activity in multiple FGFR- amplified or mutated cancer models. Mol Cancer Ther, 2012. 11(3): p. 690-9. 7. Mologni, L., et al., Ponatinib is a potent inhibitor of wild-type and drug-resistant gatekeeper mutant RET kinase. Mol Cell Endocrinol, 2013. 377(1-2): p. 1-6. 8. Garner, A.P., et al., Ponatinib inhibits polyclonal drug-resistant KIT oncoproteins and shows therapeutic potential in heavily pretreated gastrointestinal stromal tumor (GIST) patients. Clin Cancer Res, 2014. 20(22): p. 5745-55. 9. Ahmad, S., G.L. Johnson, and J.E. Scott, Identification of ponatinib and other known kinase inhibitors with potent MEKK2 inhibitory activity. Biochem Biophys Res Commun, 2015. 463(4): p. 888-93. 10. Gozgit, J.M., et al., Potent activity of ponatinib (AP24534) in models of FLT3-driven acute myeloid leukemia and other hematologic malignancies. Mol Cancer Ther, 2011. 10(6): p. 1028-35. 11. Fauster, A., et al., A cellular screen identifies ponatinib and pazopanib as inhibitors of necroptosis. Cell Death Dis, 2015. 6: p. e1767. 12. Novartis Pharmaceuticals Corporation, Gleevec [prescribing information] 2004; Available from: https:// www.pharma.us.novartis.com/sites/www.pharma.us.novartis.com/files/gleevec_tabs.pdf. 13. Novartis Pharmaceuticals, Tasigna [package insert] . 2014; Available from: https://www.pharma.us.novartis . com/product/pi/pdf/tasigna.pdf. 14. Li, X., et al., Characterization of dasatinib and its structural analogs as CYP3A4mechanism-based inactivators and the proposed bioactivation pathways. Drug Metab Dispos, 2009. 37(6): p. 1242-50. 15. Abbas, R., et al., Effect of ketoconazole on the pharmacokinetics of oral bosutinib in healthy subjects. J Clin Pharmacol, 2011. 51(12): p. 1721-7. 16. Narasimhan, N.I., et al., Evaluation of pharmacokinetics and safety of ponatinib in subjects with chronic hepatic impairment and matched healthy subjects. Cancer Chemother Pharmacol, 2014. 74(2): p. 341-8. 17. Adnan A. Kadi, H.W.D., Mohamed W. Attwa, Sawsan M. Amer, Detection and characterization of ponatinib reactive metabolites by liquid chromatography tandem mass spectrometry and elucidation of bioactivation pathways. RSC advances, 2016. 6(76). 18. Ye, Y.E., C.N. Woodward, and N.I. Narasimhan, Absorption, metabolism, and excretion of [14C]ponatinib after a single oral dose in humans. Cancer Chemother Pharmacol, 2017. 79(3): p. 507-518. 19. Schinkel, A.H., et al., P-glycoprotein in the blood-brain barrier of mice influences the brain penetration and pharmacological activity of many drugs. J Clin Invest, 1996. 97(11): p. 2517-24. 20. Vlaming, M.L., J.S. Lagas, and A.H. Schinkel, Physiological and pharmacological roles of ABCG2 (BCRP): recent findings in Abcg2 knockout mice. Adv Drug Deliv Rev, 2009. 61(1): p. 14-25. 21. Yabuki, N., et al., Gene amplification and expression in lung cancer cells with acquired paclitaxel resistance. Cancer Genet Cytogenet, 2007. 173(1): p. 1-9.

RkJQdWJsaXNoZXIy MTk4NDMw