Population Pharmacokinetics and Exposure-Safety Relationships of Alisertib in Children and Adolescents With Advanced Malignancies

Xiaofei Zhou, Diane R Mould, Ying Yuan, Elizabeth Fox, Emily Greengard, Douglas V Faller, Karthik Venkatakrishnan, Xiaofei Zhou, Diane R Mould, Ying Yuan, Elizabeth Fox, Emily Greengard, Douglas V Faller, Karthik Venkatakrishnan

Abstract

Population pharmacokinetic (PK) and exposure-safety analyses of alisertib were performed in children enrolled in 2 clinical trials: NCT02444884 and NCT01154816. NCT02444884 was a dose-finding study in children with relapsed/refractory solid malignancies (phase 1) or neuroblastomas (phase 2). Patients received oral alisertib 45 to 100 mg/m2 as powder-in-capsule once daily or twice daily for 7 days in 21-day cycles. Serial blood samples were collected up to 24 hours after dosing on cycle 1, day 1. NCT01154816 was a phase 2 single-arm study evaluating efficacy in children with relapsed/refractory solid malignancies or acute leukemias. Patients received alisertib 80 mg/m2 as enteric-coated tablets once daily for 7 days in 21-day cycles. Sparse PK samples were collected up to 8 hours after dosing on cycle 1, day 1. Sources of alisertib PK variability were characterized and quantified using nonlinear mixed-effects modeling to support dosing recommendations in children and adolescents. A 2-compartment model with oral absorption described by 3 transit compartments was developed using data from 146 patients. Apparent oral clearance and central distribution volume were correlated with body surface area across the age range of 2 to 21 years, supporting the use of body surface area-based alisertib dosing in the pediatric population. The recommended dose of 80 mg/m2 once daily enteric-coated tablets provided similar alisertib exposures across pediatric age groups and comparable exposure to that in adults receiving 50 mg twice daily (recommended adult dose). Statistically significant relationships (P < .01) were observed between alisertib exposures and incidence of grade ≥2 stomatitis and febrile neutropenia, consistent with antiproliferative mechanism-related toxicities.

Keywords: Aurora A kinase; alisertib; exposure-safety; pediatric; population pharmacokinetics.

Conflict of interest statement

X.Z., Y.Y., and D.V.F. disclose employment by Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. D.R.M. is a paid consultant for Millennium Pharmaceuticals, Inc, Cambridge, MA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. K. V. discloses previous employment by Millennium Pharmaceuticals, Inc. E.F. and E.G. have no disclosures to report.

© 2021 Millennium Pharmaceuticals Inc. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
BSA covariate effect in the final model. BSA, body surface area; BSV, between subject variability; eta, empirical Bayes prediction of the interindividual random effect.
Figure 2
Figure 2
Alisertib CL/F (A) and BSA‐normalized CL/F (B) by age group for pediatric patients. Estimates were obtained under the final model. Data for age groups 2 to 5, 6 to 11, and 12 to 16 are pooled for the group “2 through 16” box. Note that CL/F is presented as reference CL/F (ie, CL/F associated with capsule formulation). BSA, body surface area; CL/F, apparent clearance.
Figure 3
Figure 3
Formulation covariate effect in the final model. BASENOBIO refers to the base model without a bioavailability term for the enteric‐coated tablet rather than the base model, which includes this term. The FINAL model includes the effects of formulation on F and of BSA on CL/F and V1/F. BSA, body surface area; BSV, between subject variability; CL/F, apparent clearance; eta, empirical Bayes estimate of the interindividual random effect; F, bioavailability; V1/F, apparent central volume.
Figure 4
Figure 4
Visual predictive check for base and final models for (A) powder‐in‐capsule and (B) enteric‐coated tablet formulations for day 1 of once daily regimen. The observed analysis data (symbols) were compared to 200 data sets simulated using the base and final models. Black circles are observed data, red lines are observed data 5th, 50th, and 95th percentiles. Pink shaded regions are 95% prediction intervals for the 5th, 50th, and 95th percentiles of simulated data.
Figure 5
Figure 5
Alisertib AUCss by age group—pediatric simulated 80 mg/m2. Post hoc clearance used to calculate AUCss, and enteric‐coated tablet doses were adjusted for relative bioavailability. Results pooled across analyses. Results are presented for final pediatric model using 80 mg/m2 (recommended pediatric dose) using the enteric‐coated tablet formulation (F = 0.671). Results for adults are derived from the final adult model using 50 mg twice daily (100 mg total daily dose, recommended clinical dose for adults). Only adult data from Western region are presented. AUCss, area under the concentration time curve at a steady state.
Figure 6
Figure 6
Logistic regression fitted model and 95% confidence intervals for (A) grade ≥2 stomatitis and (B) febrile neutropenia. Circles represent observed incidence in each tertile of alisertib average concentration. Logistic regression model was used to estimate adverse event incidence of clinical interest with alisertib average concentration as a predictor (base model). The covariate effects that may potentially modulate the exposure–safety relationship were examined in a full model via a stepwise procedure, where a covariate was entered into the model if it was significant at the 10% level. A covariate remained in the model at the backward elimination step when the covariate was significant at the 5% level. The covariates considered in the full model include age (2 to ≤12 years, >12 to ≤16 years, >16 years), sex (male, female), Eastern Cooperative Oncology Group performance score (0, >0), number of prior therapy regimens (≤1, ≥2), cancer type (hematologic, nonhematologic) and number of treatment cycles while on study (1, ≥2). For the adverse event end point “grade ≥2 stomatitis” (A) the full model is reduced to the base model after the stepwise selection procedure. For the adverse event end point “febrile neutropenia” (B), the full model with stepwise selection identified that the covariate of “cancer type” is significant. As the sample size of patients with hematologic malignancies was small (n = 19; 13% of the evaluable data set), the relationship between exposure and probability of febrile neutropenia for the base model is shown here.

References

    1. Bolanos‐Garcia VM. Aurora kinases. Int J Biochem Cell Biol. 2005;37(8):1572‐1577.
    1. Venkatakrishnan K, Zhou X, Ecsedy J, et al. Dose selection for the investigational anticancer agent alisertib (MLN8237): pharmacokinetics, pharmacodynamics, and exposure‐safety relationships. J Clin Pharmacol. 2015;55(3):336‐347.
    1. Zullo KM, Guo Y, Cooke L, et al. Aurora A kinase inhibition selectively synergizes with histone deacetylase inhibitor through cytokinesis failure in T‐cell lymphoma. Clin Cancer Res. 2015;21(18):4097‐4109.
    1. Nikonova AS, Astsaturov I, Serebriiskii IG, Dunbrack RL Jr, Golemis EA. Aurora A kinase (AURKA) in normal and pathological cell division. Cell Mol Life Sci. 2013;70(4):661‐687.
    1. Ikezoe T, Yang J, Nishioka C, et al. A novel treatment strategy targeting Aurora kinases in acute myelogenous leukemia. Mol Cancer Ther. 2007;6(6):1851‐1857.
    1. Dar AA, Zaika A, Piazuelo MB, et al. Frequent overexpression of Aurora kinase A in upper gastrointestinal adenocarcinomas correlates with potent antiapoptotic functions. Cancer. 2008;112(8):1688‐1698.
    1. Mazumdar A, Henderson YC, El‐Naggar AK, Sen S, Clayman GL. Aurora kinase A inhibition and paclitaxel as targeted combination therapy for head and neck squamous cell carcinoma. Head Neck. 2009;31(5):625‐634.
    1. Manfredi MG, Ecsedy JA, Chakravarty A, et al. Characterization of alisertib (MLN8237), an investigational small‐molecule inhibitor of Aurora A kinase using novel in vivo pharmacodynamic assays. Clin Cancer Res. 2011;17(24):7614‐7624.
    1. Ding YH, Zhou ZW, Ha CF, et al. Alisertib, an Aurora kinase A inhibitor, induces apoptosis and autophagy but inhibits epithelial to mesenchymal transition in human epithelial ovarian cancer cells. Drug Des Devel Ther. 2015;9:425‐464.
    1. Wang F, Li H, Yan XG, et al. Alisertib induces cell cycle arrest and autophagy and suppresses epithelial‐to‐mesenchymal transition involving PI3K/Akt/mTOR and sirtuin 1‐mediated signaling pathways in human pancreatic cancer cells. Drug Des Devel Ther. 2015;9:575‐601.
    1. Goldberg SL, Fenaux P, Craig MD, et al. An exploratory phase 2 study of investigational Aurora A kinase inhibitor alisertib (MLN8237) in acute myelogenous leukemia and myelodysplastic syndromes. Leuk Res Rep. 2014;3(2):58‐61.
    1. Melichar B, Adenis A, Lockhart AC, et al. Safety and activity of alisertib, an investigational Aurora kinase A inhibitor, in patients with breast cancer, small‐cell lung cancer, non‐small‐cell lung cancer, head and neck squamous‐cell carcinoma, and gastro‐oesophageal adenocarcinoma: a five‐arm phase 2 study. Lancet Oncol. 2015;16(4):395‐405.
    1. Zhou X, Mould DR, Takubo T, et al. Global population pharmacokinetics of the investigational Aurora A kinase inhibitor alisertib in cancer patients: rationale for lower dosage in Asia. Br J Clin Pharmacol. 2018;84(1):35‐51.
    1. Carol H, Boehm I, Reynolds CP, et al. Efficacy and pharmacokinetic/pharmacodynamic evaluation of the Aurora kinase A inhibitor MLN8237 against preclinical models of pediatric cancer. Cancer Chemother Pharmacol. 2011;68(5):1291‐1304.
    1. Maris JM, Morton CL, Gorlick R, et al. Initial testing of the Aurora kinase A inhibitor MLN8237 by the Pediatric Preclinical Testing Program (PPTP). Pediatr Blood Cancer. 2010;55(1):26‐34.
    1. Muscal JA, Scorsone KA, Zhang L, Ecsedy JA, Berg SL. Additive effects of vorinostat and MLN8237 in pediatric leukemia, medulloblastoma, and neuroblastoma cell lines. Invest New Drugs. 2013;31(1):39‐45.
    1. Lipsitz EG, Nguyen V, Zhao H, et al. Modeling MLN8237, an Aurora kinase A inhibitor, with irinotecan (IRN) and temozolomide (TMZ) in neuroblastoma (NB). J Clin Oncol. 2010;28(15_suppl):10593‐10593.
    1. Zhou X, Pant S, Nemunaitis J, et al. Effects of rifampin, itraconazole and esomeprazole on the pharmacokinetics of alisertib, an investigational Aurora A kinase inhibitor in patients with advanced malignancies. Invest New Drugs. 2018;36(2):248‐258.
    1. Zhou X, Pusalkar S, Chowdhury SK, et al. Mass balance, routes of excretion, and pharmacokinetics of investigational oral [14C]‐alisertib (MLN8237), an Aurora A kinase inhibitor in patients with advanced solid tumors. Invest New Drugs. 2019;37(4):666‐673.
    1. Pusalkar S, Zhou X, Li Y, et al. Biotransformation pathways and metabolite profiles of oral [14C]alisertib (MLN8237), an investigational Aurora A kinase inhibitor, in patients with advanced solid tumors. Drug Metab Dispos. 2020;48(3):217‐229.
    1. Zhou X, Lockhart AC, Fu S, et al. Pharmacokinetics of the investigational Aurora A kinase inhibitor alisertib in adult patients with advanced solid tumors or relapsed/refractory lymphoma with varying degrees of hepatic dysfunction. J Clin Pharmacol. 2019;59(9):1204‐1215.
    1. Mosse YP, Lipsitz E, Fox E, et al. Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective small‐molecule inhibitor of Aurora kinase A: a children's oncology group phase I consortium study. Clin Cancer Res. 2012;18(21):6058‐6064.
    1. Mosse YP, Fox E, Teachey DT, et al. A phase II study of alisertib in children with recurrent/refractory solid tumors or leukemia: Children's Oncology Group phase I and pilot consortium (ADVL0921). Clin Cancer Res. 2019;25(11):3229‐3238.
    1. Skolnik JM, Barrett JS, Jayaraman B, Patel D, Adamson PC. Shortening the timeline of pediatric phase I trials: the rolling six design. J Clin Oncol. 2008;26(2):190‐195.
    1. Lipsitz E, Moorthy G, Mosse Y, Fox E, Adamson PC. A sensitive and selective liquid chromatography/tandem mass spectrometry method for determination of MLN8237 in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2010;878(25):2369‐2373.
    1. Dees EC, Cohen RB, von Mehren M, et al. Phase I study of Aurora A kinase inhibitor MLN8237 in advanced solid tumors: safety, pharmacokinetics, pharmacodynamics, and bioavailability of two oral formulations. Clin Cancer Res. 2012;18(17):4775‐4784.
    1. Cervantes A, Elez E, Roda D, et al. Phase I pharmacokinetic/pharmacodynamic study of MLN8237, an investigational, oral, selective Aurora A kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res. 2012;18(17):4764‐4774.
    1. Kelly KR, Shea TC, Goy A, et al. Phase I study of MLN8237–investigational Aurora A kinase inhibitor–in relapsed/refractory multiple myeloma, non‐Hodgkin lymphoma and chronic lymphocytic leukemia. Invest New Drugs. 2014;32(3):489‐499.
    1. Falchook G, Kurzrock R, Gouw L, et al. Investigational Aurora A kinase inhibitor alisertib (MLN8237) as an enteric‐coated tablet formulation in non‐hematologic malignancies: phase 1 dose‐escalation study. Invest New Drugs. 2014;32(6):1181‐1187.
    1. Fernandez E, Perez R, Hernandez A, Tejada P, Arteta M, Ramos JT. Factors and mechanisms for pharmacokinetic differences between pediatric population and adults. Pharmaceutics. 2011;3(1):53‐72.
    1. O'Connor OA, Özcan M, Jacobsen ED, et al. Randomized phase III study of alisertib or investigator's choice (selected single agent) in patients with relapsed or refractory peripheral T‐cell lymphoma. J Clin Oncol. 2019;37(8):613‐623.
    1. Battisti WP, Wager E, Baltzer L, et al. Good publication practice for communicating company‐sponsored medical research: GPP3. Ann Intern Med. 2015;163:461‐464.

Source: PubMed

3
Tilaa