Population pharmacokinetics-pharmacodynamics of sunitinib in pediatric patients with solid tumors

Erjian Wang, Steven G DuBois, Cynthia Wetmore, Reza Khosravan, Erjian Wang, Steven G DuBois, Cynthia Wetmore, Reza Khosravan

Abstract

Purpose: The safety profile of sunitinib in children, including the impact of sunitinib exposure on safety endpoints, was assessed using population pharmacokinetic (PK) and pharmacokinetic-pharmacodynamic (PK-PD) models.

Methods: Data were from two clinical studies in 59 children with solid tumors (age range 2-21 years, 28 male/31 female, body weight range 16.2-100 kg, body surface are [BSA] range 0.7-2.1 m2). Analysis of covariates that affected PK and PD parameters was conducted using a nonlinear mixed-effects model. Safety and tolerability endpoints were absolute neutrophil count, hepatic transaminases, diastolic blood pressure, hemoglobin, lymphocyte count, platelet count, white blood cell count, hand-foot syndrome, fatigue, nausea, intracranial hemorrhage, and vomiting.

Results: The models well described the time courses of concentrations of sunitinib and its primary active metabolite SU012662, as well as safety and tolerability endpoints. In PK models for sunitinib and SU012662, BSA was the only covariate that statistically significantly affected apparent clearance (CL/F) and apparent central volume of distribution (Vc/F). Higher BSA was associated with greater CL/F and Vc/F. No statistically significant covariates were identified in the PK-PD models. For safety endpoints that had a sufficient number of adverse events, a higher probability of adverse events was associated with higher average plasma sunitinib concentrations.

Conclusion: In PK models, BSA was the only covariate that affected major PK parameters of sunitinib and SU012662. Based on analysis of safety and tolerability endpoints, the PK-PD relationships were mainly driven by sunitinib plasma exposures and were not affected by age, sex, respective baseline safety endpoint values, baseline Eastern Cooperative Oncology Group performance status, or body size.

Trial registration: ClinicalTrials.gov: NCT00387920 (registered October 13, 2006), NCT01462695 (registered October 31, 2011).

Keywords: Children; Pharmacodynamic; Pharmacokinetic; Safety; Solid tumor; Sunitinib.

Conflict of interest statement

Erjian Wang and Reza Khosravan are employees of Pfizer and hold stock or stock options with Pfizer. Steven G. DuBois has received prior travel expenses from Loxo Oncology, Roche and Salarius, and prior consulting fees from Loxo Oncology. Cynthia Wetmore has received research support and prior travel expenses from Eli Lilly & Co.

Figures

Fig. 1
Fig. 1
Final model prediction and variance-corrected visual predictive check plots for sunitinib and SU012662 plasma concentrations, and safety and tolerability endpoints, up to 3000 h post-dose. Visual predictive check plots for a sunitinib, b SU012662, c ANC, d ALT, e AST, f diastolic BP, g hemoglobin, h lymphocyte count, i platelet count, and j WBC count. Blue circles represent the observed data. Red lines represent the median (solid line) and 2.5th and 97.5th percentiles (dashed lines) of the observed data. Black lines represent the median (solid line) and 2.5th and 97.5th percentiles (dashed lines) of the simulated data. The 95% CIs for simulated median and each percentile are shown by pink and blue shaded areas, respectively. ALT alanine transaminase, ANC absolute neutrophil count, AST aspartate transaminase, BP blood pressure, WBC white blood cell
Fig. 2
Fig. 2
Observed and predicted probabilities of worst adverse event grades for the safety and tolerability endpoints. Probabilities of worst grade adverse events for a neutropenia (n = 23 patients), b increased ALT (n = 15), c increased AST (n = 13), d hypertension (n = 10), (E) lymphopenia (n = 16), f thrombocytopenia (n = 13), g leukopenia (n = 20), h fatigue (n = 24), i nausea (n = 7), and j vomiting (n = 7). ALT alanine transaminase, AST aspartate transaminase

References

    1. Chow LQ, Eckhardt SG. Sunitinib: from rational design to clinical efficacy. J Clin Oncol. 2007;25:884–896. doi: 10.1200/JCO.2006.06.3602.
    1. Faivre S, Demetri G, Sargent W, Raymond E. Molecular basis for sunitinib efficacy and future clinical development. Nat Rev Drug Discov. 2007;6:734–745. doi: 10.1038/nrd2380.
    1. Mena AC, Pulido EG, Guillén-Ponce C. Understanding the molecular-based mechanism of action of the tyrosine kinase inhibitor: sunitinib. Anticancer Drugs. 2010;21(Suppl 1):S3–11. doi: 10.1097/01.cad.0000361534.44052.c5.
    1. Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, Schreck RE, Abrams TJ, Ngai TJ, Lee LB, et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res. 2003;9:327–337.
    1. Pfizer Inc (2019) Sutent® (sunitinib) prescribing information. . Accessed 17 May 2019
    1. European Medicines Agency (2016) Sutent® (sunitinib) summary of product characteristics. . Accessed 17 May 2019
    1. Janeway KA, Albritton KH, Van Den Abbeele AD, D'Amato GZ, Pedrazzoli P, Siena S, Picus J, Butrynski JE, Schlemmer M, Heinrich MC, et al. Sunitinib treatment in pediatric patients with advanced GIST following failure of imatinib. Pediatr Blood Cancer. 2009;52:767–771. doi: 10.1002/pbc.21909.
    1. Dubois SG, Shusterman S, Ingle AM, Ahern CH, Reid JM, Wu B, Baruchel S, Glade-Bender J, Ivy P, Grier HE, et al. Phase I and pharmacokinetic study of sunitinib in pediatric patients with refractory solid tumors: a children’s oncology group study. Clin Cancer Res. 2011;17:5113–5122. doi: 10.1158/1078-0432.CCR-11-0237.
    1. DuBois SG, Shusterman S, Reid JM, Ingle AM, Ahern CH, Baruchel S, Glade-Bender J, Ivy P, Adamson PC, Blaney SM. Tolerability and pharmacokinetic profile of a sunitinib powder formulation in pediatric patients with refractory solid tumors: a children’s oncology group study. Cancer Chemother Pharmacol. 2012;69:1021–1027. doi: 10.1007/s00280-011-1798-2.
    1. Wetmore C, Daryani VM, Billups CA, Boyett JM, Leary S, Tanos R, Goldsmith KC, Stewart CF, Blaney SM, Gajjar A. Phase II evaluation of sunitinib in the treatment of recurrent or refractory high-grade glioma or ependymoma in children: a children’s oncology group study ACNS1021. Cancer Med. 2016;5:1416–1424. doi: 10.1002/cam4.713.
    1. Rutkowski P, Magnan H, Chou AJ, Benson C. Treatment of gastrointestinal stromal tumours in paediatric and young adult patients with sunitinib: a multicentre case series. BMC Cancer. 2017;17:717. doi: 10.1186/s12885-017-3727-1.
    1. Verschuur AC, Bajčiová V, Mascarenhas L, Khosravan R, Lin X, Ingrosso A, Janeway KA. Sunitinib in pediatric patients with advanced gastrointestinal stromal tumor: results from a phase I/II trial. Cancer Chemother Pharmacol. 2019;84:41–50. doi: 10.1007/s00280-019-03814-5.
    1. Houk BE, Bello CL, Kang D, Amantea M. A population pharmacokinetic meta-analysis of sunitinib malate (SU11248) and its primary metabolite (SU12662) in healthy volunteers and oncology patients. Clin Cancer Res. 2009;15:2497–2506. doi: 10.1158/1078-0432.CCR-08-1893.
    1. Houk BE, Bello CL, Poland B, Rosen LS, Demetri GD, Motzer RJ. Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta-analysis. Cancer Chemother Pharmacol. 2010;66:357–371. doi: 10.1007/s00280-009-1170-y.
    1. Lindauer A, Di Gion P, Kanefendt F, Tomalik-Scharte D, Kinzig M, Rodamer M, Dodos F, Sörgel F, Fuhr U, Jaehde U. Pharmacokinetic/pharmacodynamic modeling of biomarker response to sunitinib in healthy volunteers. Clin Pharmacol Ther. 2010;87:601–608. doi: 10.1038/clpt.2010.20.
    1. Khosravan R, Motzer RJ, Fumagalli E, Rini BI. Population pharmacokinetic/pharmacodynamic modeling of sunitinib by dosing schedule in patients with advanced renal cell carcinoma or gastrointestinal stromal tumor. Clin Pharmacokinet. 2016;55:1251–1269. doi: 10.1007/s40262-016-0404-5.
    1. European Medicines Agency Committee For Medicinal Products For Human Use (2007) Guideline on the role of pharmacokinetics in the development of medicinal products in the paediatric population. . Accessed 18 October 2019
    1. U.S. Department of Health and Human Servics Food and Drug Administration, Center for Drug Evaluation and Research (CDER) (2014) General clinical pharmacology consideration for pediatric studies for drugs and biological products. Guidance for industry. Accessed 18 October 2019

Source: PubMed

Подписаться