A Phase 1, Open-Label Study in Healthy Subjects to Evaluate the Absolute Bioavailability of AG-221 by a Microtracer Approach

Xiaomin Wang, Jian Chen, Josephine Reyes, Simon Zhou, Maria Palmisano, Yan Li, Xiaomin Wang, Jian Chen, Josephine Reyes, Simon Zhou, Maria Palmisano, Yan Li

Abstract

Introduction: The purpose of this study was to evaluate the absolute bioavailability (BA) of AG-221 following a single oral dose of 100 mg AG-221 and an intravenous (IV) dose of ~ 100 μg AG-221 containing approximately 300 nCi of [14C]-AG-221.

Methods: This was a phase 1, open-label study. Six subjects who met all of the inclusion criteria and none of the exclusion criteria were enrolled in the study. After an overnight fast of at least 10 h, the subjects received an oral dose (coated tablet) of 100 mg of AG-221 at 0 h on dosing day. Four hours after the oral dose, the subjects received 100 μg AG-221 containing ~ 300 nCi of [14C]-AG-221 administered as an IV bolus. Blood samples were collected and analyzed for plasma concentrations of AG-221 and [14C]-AG-221 using a validated liquid chromatography with tandem mass spectrometry (LC-MS/MS) system and high-performance liquid chromatography (HPLC) fractionation followed by accelerator mass spectrometry analysis (AMS), respectively. Safety was evaluated throughout the study.

Results: The absolute BA after a 100-mg oral dose of AG-221 was measured as 57.2%. While the total clearance was 1.37 L/h, ~ 1/60 of the liver blood flow in a typical 70-kg human subject, the first-pass extraction was estimated to be less than 2%, assuming that the total clearance was entirely due to liver metabolism. Thus, the fraction of the AG-221 dose absorbed was at least 50%. AG-221 was safe and well tolerated when given under fasted conditions in a single 100-mg dose as a coated tablet with a microtracer [14C]-AG-221 solution, as few drug-related treatment-emergent adverse events (TEAEs) were reported. No clinically significant changes or findings were noted in the clinical laboratory evaluations, vital sign measurements, and electrocardiograms (ECGs) performed during this study.

Conclusions: In healthy subjects under fasting conditions, the absolute BA following oral administration of a 100-mg AG-221 tablet was 57.2%. AG-221 was safe and well tolerated in healthy male subjects when administered as a single 100-mg film-coated tablet plus 100 µg [14C]-AG-221 given intravenously.

Trial registration: ClinicalTrials.gov identifier, NCT02443168.

Funding: Celgene Corporation.

Keywords: AG-221; Absolute bioavailability; Accelerator mass spectrometry; Relapsed or refractory acute myeloid leukemia.

Figures

Fig. 1
Fig. 1
Study design
Fig. 2
Fig. 2
Structures of AG-221 and [14C]-AG-221
Fig. 3
Fig. 3
Mean (± standard deviation) dose-normalized AG-221 plasma concentration–time profiles: absolute BA assessment for oral and IV administration (red lines and symbols represent data from IV administration and blue lines and symbols represent data from oral administration)
Fig. 4
Fig. 4
Individual dose-normalized AUC0–inf for AG-221 (oral) and [14C]-AG-221 (IV). Squares represent individual values. Box plot provides median and 25%/75% quartiles with whiskers to the last point within the 1.5 × interquartile range. Squares connected by dotted lines are data from the same subject

References

    1. Dogra R, Bhatia R, Shankar R, Bansal P, Rawal RK. Enasidenib: first mutant IDH2 inhibitor for the treatment of refractory and relapsed acute myeloid leukemia. Anticancer Agents Med Chem. 2018;18(14):1936–1951. doi: 10.2174/1871520618666181025091128.
    1. Myers RA, Wirth S, Williams S, Kiel PJ. Enasidenib: an oral IDH2 inhibitor for the treatment of acute myeloid leukemia. J Adv Pract Oncol. 2018;9(4):435–440.
    1. Pollyea DA, Tallman MS, de Botton S, Kantarjian HM, Collins R, Stein AS, et al. Enasidenib, an inhibitor of mutant IDH2 proteins, induces durable remissions in older patients with newly diagnosed acute myeloid leukemia. Leukemia. 2019 doi: 10.1038/s41375-019-0472-2.
    1. Celgene Corporation. Package insert for enasidenib. Summit, NJ: Celgene Corporation; 2017.
    1. Click ZR, Seddon AN, Bae YR, Fisher JD, Ogunniyi A. New Food and Drug Administration-approved and emerging novel treatment options for acute myeloid leukemia. Pharmacotherapy. 2018;38(11):1143–54.
    1. Talati C, Sweet K. Recently approved therapies in acute myeloid leukemia: a complex treatment landscape. Leuk Res. 2018;73:58–66. doi: 10.1016/j.leukres.2018.09.001.
    1. Li Y, Connarn JN, Chen J, Tong Z, Palmisano M, Zhou S. Modeling and simulation of the endogenous CYP3A induction marker 4β-hydroxycholesterol during enasidenib treatment. Clin Pharmacol. 2019;11:39–50.
    1. Li Y, Liu L, Gomez D, Chen J, Tong Z, Palmisano M, et al. Pharmacokinetics and safety of enasidenib following single oral doses in Japanese and Caucasian subjects. Pharmacol Res Perspect. 2018;6(6):e00436.
    1. Tong Z, Atsriku C, Yerramilli U, Wang X, Li Y, Reyes J, et al. Absorption, distribution, metabolism and excretion of an isocitrate dehydrogenase-2 inhibitor enasidenib in rats and humans. Xenobiotica. 2019;49(2):200–210. doi: 10.1080/00498254.2018.1425511.
    1. Shaik N, Hee B, Liang Y, LaBadie RR. Absolute oral bioavailability of glasdegib (PF-04449913), a smoothened inhibitor, in randomized healthy volunteers. Clin Pharmacol Drug Dev. 2019 doi: 10.1002/cpdd.692.
    1. US FDA. Bioavailability studies submitted in NDAs or INDs—general considerations. Silver Spring, MD: US FDA; 2019.
    1. European Medicines Agency. Pharmacokinetic studies in man. Guideline. Amsterdam: European Medicines Agency; 1987.
    1. Boulton DW, Kasichayanula S, Keung CF, Arnold ME, Christopher LJ, Xu XS, et al. Simultaneous oral therapeutic and intravenous 14C-microdoses to determine the absolute oral bioavailability of saxagliptin and dapagliflozin. Br J Clin Pharmacol. 2013;75(3):763–8.
    1. Center for Drug Evaluation and Research. Waiver of in vivo bioavailability and bioequivalence studies for immediate-release solid oral dosage forms based on a biopharmaceutics classification system. Guidance for industry. Silver Spring, MD: US FDA; 2000.
    1. Lappin G, Noveck R, Burt T. Microdosing and drug development: past, present and future. Expert Opin Drug Metab Toxicol. 2013;9(7):817–834. doi: 10.1517/17425255.2013.786042.
    1. Burt T, Yoshida K, Lappin G, Vuong L, John C, de Wildt SN, et al. Microdosing and other phase 0 clinical trials: facilitating translation in drug development. Clin Transl Sci. 2016;9(2):74–88.
    1. Sudo K. Microdosing for reduction of the time and resources for drug development. Drug Metab Pharmacokinet. 2007;22(5):327. doi: 10.2133/dmpk.22.327.
    1. Ings RM. Microdosing: a valuable tool for accelerating drug development and the role of bioanalytical methods in meeting the challenge. Bioanalysis. 2009;1(7):1293–1305. doi: 10.4155/bio.09.107.
    1. Burt T, Vuong LT, Baker E, Young GC, McCartt AD, Bergstrom M, et al. Phase 0, including microdosing approaches: applying the three Rs and increasing the efficiency of human drug development. Altern Lab Anim. 2018;46(6):335–46.
    1. Vishwanathan K, So K, Thomas K, Bramley A, English S, Collier J. Absolute bioavailability of osimertinib in healthy adults. Clin Pharmacol Drug Dev. 2019;8(2):198–207.
    1. Raje S, Callegari E, Sahasrabudhe V, Vaz A, Shi H, Fluhler E, et al. Novel application of the two-period microtracer approach to determine absolute oral bioavailability and fraction absorbed of ertugliflozin. Clin Transl Sci. 2018;11(4):405–411. doi: 10.1111/cts.12549.
    1. Cho YS, Lim HS, Han S, Yoon SK, Kim H, Cho YL, et al. Single-dose intravenous safety, tolerability, and pharmacokinetics and absolute bioavailability of LCB01-0371. Clin Ther. 2019;41(1):92–106. doi: 10.1016/j.clinthera.2018.11.009.

Source: PubMed

3
Sottoscrivi