Pharmacokinetic Profile of Gilteritinib: A Novel FLT-3 Tyrosine Kinase Inhibitor

Angela Joubert James, Catherine C Smith, Mark Litzow, Alexander E Perl, Jessica K Altman, Dale Shepard, Takeshi Kadokura, Kinya Souda, Melanie Patton, Zheng Lu, Chaofeng Liu, Selina Moy, Mark J Levis, Erkut Bahceci, Angela Joubert James, Catherine C Smith, Mark Litzow, Alexander E Perl, Jessica K Altman, Dale Shepard, Takeshi Kadokura, Kinya Souda, Melanie Patton, Zheng Lu, Chaofeng Liu, Selina Moy, Mark J Levis, Erkut Bahceci

Abstract

Background and objective: Gilteritinib is a novel, highly selective tyrosine kinase inhibitor approved in the USA, Canada, Europe, Brazil, Korea, and Japan for the treatment of FLT3 mutation-positive acute myeloid leukemia. This article describes the clinical pharmacokinetic profile of gilteritinib.

Methods: The pharmacokinetic profile of gilteritinib was assessed from five clinical studies.

Results: Dose-proportional pharmacokinetics was observed following once-daily gilteritinib administration (dose range 20-450 mg). Median maximum concentration was reached 2-6 h following single and repeat dosing of gilteritinib; mean elimination half-life was 113 h. Elimination was primarily via feces. Exposure to gilteritinib was comparable under fasted and fed conditions. Gilteritinib is primarily metabolized via cytochrome P450 (CYP) 3A4; coadministration of gilteritinib with itraconazole (a strong P-glycoprotein inhibitor and CYP3A4 inhibitor) or rifampicin (a strong P-glycoprotein inducer and CYP3A inducer) significantly affected the gilteritinib pharmacokinetic profile. No clinically relevant interactions were observed when gilteritinib was coadministered with midazolam (a CYP3A4 substrate) or cephalexin (a multidrug and toxin extrusion 1 substrate). Unbound gilteritinib exposure was similar between subjects with hepatic impairment and normal hepatic function.

Conclusions: Gilteritinib exhibits a dose-proportional pharmacokinetic profile in healthy subjects and in patients with relapsed/refractory acute myeloid leukemia. Gilteritinib exposure is not significantly affected by food. Moderate-to-strong CYP3A inhibitors demonstrated a significant effect on gilteritinib exposure. Coadministration of gilteritinib with CYP3A4 or multidrug and toxin extrusion 1 substrates did not impact substrate concentrations. Unbound gilteritinib was comparable between subjects with hepatic impairment and normal hepatic function; dose adjustment is not warranted for patients with hepatic impairment.

Clinical trial registration: NCT02014558, NCT02456883, NCT02571816.

Conflict of interest statement

Angela Joubert James, Melanie Patton, Chaofeng Liu, Selina Moy, and Erkut Bahceci are employees of Astellas Pharma, US Inc. Zheng Lu was an employee of Astellas Pharma US, Inc. during the time of the study and development of the manuscript. Takeshi Kadokura and Kinya Souda are employees of Astellas Pharma, Inc. Catherine C. Smith received research grants from Astellas Pharma US, Inc. Mark Litzow has no conflicts of interest that are directly relevant to the content of this article. Alexander E. Perl has received funding, honoraria, or travel reimbursement from Astellas, Daiichi-Sankyo, Arog, Novartis, Pfizer, Actinium Pharmaceuticals, Jazz Pharmaceuticals, Takeda, AbbVie, NewLink Genetics, Asana Biosciences, and Seattle Genetics. Jessica K. Altman has received funding, honoraria, or travel reimbursement from AbbVie, Agios, Ariad, Astellas, Bayer, BioSight, BMS, Boeringer Ingelheim, Cancer Expert Now, Celator, Celgene, Daiichi Sankyo, Epizyme, France Foundation, FujiFilm, Genentech, Glycomimetics, GSK, Incyte, Janssen Pharmaceuticals, Novartis, PeerView, Pfizer, prIME Oncology, Syros, and Theradex. Dale Shepard has received funding, honoraria, or travel reimbursement from Celgene, Sanofi, AstraZeneca, Ipsen, Amgen, Genentech, Eli Lilly, Bayer, Leap, Alkermes, Aduro, Halozyme, Ignyta, BMS, Pfizer, Kinex, and Corvus. Mark J. Levis has received funding or personal fees from Astellas, Novartis, Daiichi-Sankyo, and FujiFilm.

Figures

Fig. 1
Fig. 1
Mean plasma gilteritinib concentrations after escalating oral doses in patients with acute myeloid leukemia (AML). Single-dose concentration–time curve (a) and multiple-dose concentration–time curve (b). Single dose (Cycle 1 Day-2), 20 mg: n = 5; 40–450 mg: n = 3. Steady state (Cycle 1 Day 15), 20 mg: n = 4; 40 mg, 80 mg, 120 mg, 300 mg: n = 3; 200 mg: n = 2; 450 mg: n = 1
Fig. 2
Fig. 2
Median dose-normalized gilteritinib trough plasma concentration–time profiles by use of a cytochrome P450 (CYP) 3A inhibitor in patients with relapsed or refractory acute myeloid leukemia (AML)
Fig. 3
Fig. 3
Relationship between gilteritinib trough concentration and clinical response. Numbers inside the bars represent the number (n) of patients achieving each response, while the proportion (%) of patients achieving each response is represented on the y axis. All patients who received at least one dose of the study drug for whom sufficient plasma concentration data were available to facilitate the derivation of at least one pharmacokinetic (PK) parameter, and for whom the time of dosing on the day of sampling was known, were included in the analysis. CRc composite complete response, n number of patients, NE not evaluable, NR no response, PR partial response
Fig. 4
Fig. 4
Mean midazolam [MDZ] (a) and 1-OH midazolam (b) plasma concentration–time profiles in patients with relapsed or refractory acute myeloid leukemia (AML). Concentrations below the limit of quantification (BLQ) [lower limit of quantification (LLOQ) = 0.1 ng/mL] were set to zero. Mean was not calculated if concentration was BLQ for all subjects in a given category. Day-1, n = 15 at 24 h; Cycle 1, Day 15, n = 8 at 24 h. 1-OH 1-hydroxymidazolam
Fig. 5
Fig. 5
Effect of gilteritinib on the pharmacokinetics of cephalexin in patients with relapsed or refractory acute myeloid leukemia (AML). Linear scale (a) and Log scale (b). Day-1, n = 20 at 24 h; Cycle 1, Day 15, n = 16 at 24 h
Fig. 6
Fig. 6
Proposed metabolic pathway for gilteritinib
Fig. 7
Fig. 7
Effects of strong cytochrome P450 (CYP) 3A4 inhibitors and inducers on mean gilteritinib plasma concentrations in healthy adult subjects. Concentrations below the limit of quantification (BLQ) [lower limit of quantification (LLOQ) = 0.1 ng/mL] were set to zero. Mean was not calculated if concentration was BLQ for all subjects in a given category. N = 20 for each treatment arm

References

    1. The American Cancer Society . Cancer facts & figures 2018. Atlanta (GA): American Cancer Society; 2018.
    1. Schlenk RF, Dohner K. Impact of new prognostic markers in treatment decisions in acute myeloid leukemia. Curr Opin Hematol. 2009;16(2):98–104. doi: 10.1097/MOH.0b013e3283257adb.
    1. Ben-Batalla I, Schultze A, Wroblewski M, Erdmann R, Heuser M, Waizenegger JS, et al. Axl, a prognostic and therapeutic target in acute myeloid leukemia mediates paracrine crosstalk of leukemia cells with bone marrow stroma. Blood. 2013;122(14):2443–2452. doi: 10.1182/blood-2013-03-491431.
    1. Park IK, Mishra A, Chandler J, Whitman SP, Marcucci G, Caligiuri MA. Inhibition of the receptor tyrosine kinase Axl impedes activation of the FLT3 internal tandem duplication in human acute myeloid leukemia: implications for Axl as a potential therapeutic target. Blood. 2013;121(11):2064–2073. doi: 10.1182/blood-2012-07-444018.
    1. Gunawardane RN, Nepomuceno RR, Rooks AM, Hunt JP, Ricono JM, Belli B, et al. Transient exposure to quizartinib mediates sustained inhibition of FLT3 signaling while specifically inducing apoptosis in FLT3-activated leukemia cells. Mol Cancer Ther. 2013;12(4):438–447. doi: 10.1158/1535-7163.MCT-12-0305.
    1. Kampa-Schittenhelm KM, Heinrich MC, Akmut F, Dohner H, Dohner K, Schittenhelm MM. Quizartinib (AC220) is a potent second generation class III tyrosine kinase inhibitor that displays a distinct inhibition profile against mutant-FLT3, -PDGFRA and -KIT isoforms. Mol Cancer. 2013;12:19. doi: 10.1186/1476-4598-12-19.
    1. Cortes JE, Kantarjian H, Foran JM, Ghirdaladze D, Zodelava M, Borthakur G, et al. Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal tandem duplication status. J Clin Oncol. 2013;31(29):3681–3687. doi: 10.1200/JCO.2013.48.8783.
    1. Galanis A, Levis M. Inhibition of c-Kit by tyrosine kinase inhibitors. Haematologica. 2015;100(3):e77–e79. doi: 10.3324/haematol.2014.117028.
    1. Nybakken GE, Canaani J, Roy D, Morrissette JD, Watt CD, Shah NP, et al. Quizartinib elicits differential responses that correlate with karyotype and genotype of the leukemic clone. Leukemia. 2016;30(6):1422–1425. doi: 10.1038/leu.2015.320.
    1. Mori M, Kaneko N, Ueno Y, Yamada M, Tanaka R, Saito R, et al. Gilteritinib, a FLT3/AXL inhibitor, shows antileukemic activity in mouse models of FLT3 mutated acute myeloid leukemia. Investig New Drugs. 2017;35(5):556–565. doi: 10.1007/s10637-017-0470-z.
    1. Lee LY, Hernandez D, Rajkhowa T, Smith SC, Raman JR, Nguyen B, et al. Pre-clinical studies of gilteritinib, a next-generation FLT3 inhibitor. Blood. 2017;129(2):257–260. doi: 10.1182/blood-2016-10-745133.
    1. Perl AE, Altman JK, Cortes J, Smith C, Litzow M, Baer MR, et al. Selective inhibition of FLT3 by gilteritinib in relapsed/refractory acute myeloid leukemia: a multicenter, first-in-human, open-label, phase 1/2 study. Lancet Oncol. 2017;18(8):1061–1075. doi: 10.1016/S1470-2045(17)30416-3.
    1. Niwa T, Imagawa Y, Yamazaki H. Drug interactions between nine antifungal agents and drugs metabolized by human cytochromes P450. Curr Drug Metab. 2014;15(7):651–679. doi: 10.2174/1389200215666141125121511.
    1. Scheife RT, Hines LE, Boyce RD, Chung SP, Momper JD, Sommer CD, et al. Consensus recommendations for systematic evaluation of drug-drug interaction evidence for clinical decision support. Drug Saf. 2015;38(2):197–206. doi: 10.1007/s40264-014-0262-8.
    1. Faucette S, Wagh S, Trivedi A, Venkatakrishnan K, Gupta N. Reverse translation of US Food and Drug Administration reviews of oncology new molecular entities approved in 2011-2017: lessons learned for anticancer drug development. Clin Transl Sci. 2018;11(2):123–146. doi: 10.1111/cts.12527.
    1. Shah RR, Roberts SA, Shah DR. A fresh perspective on comparing the FDA and the CHMP/EMA: approval of antineoplastic tyrosine kinase inhibitors. Br J Clin Pharmacol. 2013;76(3):396–411. doi: 10.1111/bcp.12085.
    1. Hartmann JT, Haap M, Kopp HG, Lipp HP. Tyrosine kinase inhibitors: a review on pharmacology, metabolism and side effects. Curr Drug Metab. 2009;10(5):470–481. doi: 10.2174/138920009788897975.
    1. Herviou P, Thivat E, Richard D, Roche L, Dohou J, Pouget M, et al. Therapeutic drug monitoring and tyrosine kinase inhibitors. Oncol Lett. 2016;12(2):1223–1232. doi: 10.3892/ol.2016.4780.
    1. US FDA. Rifadin (rifampin capsules USP) and Rifadin IV (rifampin for injection USP). 2010. . Accessed 10 Oct 2019.
    1. Kapetas AJ, Sorich MJ, Rodrigues AD, Rowland A. Guidance for rifampin and midazolam dosing protocols to study intestinal and hepatic cytochrome P450 (CYP) 3A4 induction and de-induction. AAPS J. 2019;21(5):78. doi: 10.1208/s12248-019-0341-y.
    1. Almond LM, Mukadam S, Gardner I, Okialda K, Wong S, Hatley O, et al. Prediction of drug-drug interactions arising from CYP3A induction using a physiologically based dynamic model. Drug Metab Dispos. 2016;44(6):821–832. doi: 10.1124/dmd.115.066845.
    1. Smith CC, Levis MJ, Litzow MR, Perl AE, Altman JK, Gill S, et al. Pharmacokinetics and pharmacodynamics of gilteritinib in patients with relapsed or refractory acute myeloid leukemia. J Clin Oncol. 2016;34(15_Suppl.):76.

Source: PubMed

3
Se inscrever