Population Pharmacokinetics of Sunitinib and its Active Metabolite SU012662 in Pediatric Patients with Gastrointestinal Stromal Tumors or Other Solid Tumors

Erjian Wang, Steven G DuBois, Cynthia Wetmore, Arnauld C Verschuur, Reza Khosravan, Erjian Wang, Steven G DuBois, Cynthia Wetmore, Arnauld C Verschuur, Reza Khosravan

Abstract

Background and objective: Population pharmacokinetic analysis explored the pharmacokinetics of sunitinib and its primary active metabolite, SU012662, in children and evaluated the sunitinib dose(s) that produce comparable plasma exposures to adults receiving the approved daily dose.

Methods: Data were from 65 children with gastrointestinal stromal tumors (GIST) or solid tumors. Pharmacokinetic models of sunitinib and SU012662 were developed using a systematic multi-step approach employing nonlinear mixed-effects modeling. The effect of predefined covariates on pharmacokinetic parameters was assessed. Final models were validated using visual predictive check and statistical techniques.

Results: The final dataset comprised 439 sunitinib and 417 SU012662 post-baseline plasma observations. Base models were characterized by two-compartment models with first-order absorption and lag time. Body surface area (BSA) was the only covariate that affected (P < 0.001) pharmacokinetic parameters for sunitinib and SU012662 and was incorporated into the final models. Bootstrap results indicated that the final models represented the final dataset adequately. Based on the final models, a sunitinib dose of ~ 20mg/m2/day in children with GIST aged 6-17 years would be expected to lead to similar total plasma exposures of sunitinib and SU012661 as a dose of 50 mg/day in an adult with GIST on schedule 4/2.

Conclusions: In children with GIST or solid tumors receiving sunitinib, population pharmacokinetic analysis identified BSA as the only covariate that affected pharmacokinetic parameters and predicted a dose of ~ 20 mg/m2/day as achieving equivalent exposure to 50 mg/day in adults with GIST on schedule 4/2.

Trial registration: ClinicalTrials.gov identifiers (date registered): NCT01396148 (July 2011); NCT01462695 (October 2011); NCT00387920 (October 2006).

Conflict of interest statement

EW and RK are employees of Pfizer and hold stock or stock options with Pfizer. SGD has received travel expenses from Roche, Salarius, and Loxo Oncology, and consulting fees from Bayer and Loxo Oncology. CW has received research support and prior travel expenses from Eli Lilly & Co, and is an employee Exelixis Inc and holds stock or stock options with Exelixis Inc. ACV is a consultant for Pfizer.

Figures

Fig. 1
Fig. 1
Goodness-of-fit diagnostic plots for plasma sunitinib (a) and SU012662 (b) concentrations from the final model.
Fig. 2
Fig. 2
Visual predictive check plots for sunitinib (a) and SU012662 (b) in the first 12 h post-dose and for sunitinib (c) and SU012662 (d) up to 3000 h post-dose. Blue circles represent the observed data, and the red lines represent the median (solid line) and 2.5th and 97.5th percentiles (dashed lines) of the observed data. The solid black line and dashed black lines represent the median and 2.5th and 97.5th percentiles of the simulated data. The 95% confidence intervals for the simulated median and each percentile are shown by the pink and blue shaded areas, respectively. Sunitinib was administered at a dose of 15 mg/m2 once daily on a schedule of 4 weeks on treatment followed by 2 weeks off treatment (schedule 4/2)
Fig. 3
Fig. 3
Effect of BSA on sunitinib (a) and SU012662 (b) AUC following multiple dosing of sunitinib 20 mg/m2 for different age groups, based on the final model. Effects are presented as probability density plots on a relative scale to indicate proportional size and precision relative to the adult reference patient. The plots represent distributions of 1000 nonparametric bootstrap estimates, with the heights of the plots representing probability. The reference is the geometric mean steady-state 24 h sunitinib and SU012662 AUCs of 1233 and 551 ng • h/ml, respectively, in an adult patient with GIST receiving a sunitinib dose of 50 mg. The green shaded areas represent the 0.75–1.25 ratio range. AUC area under the curve, BSA body surface area, GIST gastrointestinal stromal tumor

References

    1. Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G, 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. 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, Guillen-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. Pfizer Inc. Sutent® (sunitinib malate) prescribing information. Pfizer, New York, NY. 2006. . Accessed 5 Nov 2019.
    1. European Medicines Agency. Sutent® (sunitinib) summary of product characteristics. European Medicines Agency, Sandwich, UK. 2009. . Accessed 5 Nov 2019.
    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. 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. Lindauer A, Di Gion P, Kanefendt F, Tomalik-Scharte D, Kinzig M, Rodamer M, et al. 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. Wang E, DuBois SG, Wetmore C, Khosravan R. Population pharmacokinetics-pharmacodynamics of sunitinib in pediatric patients with solid tumors. Cancer Chemother Pharmacol. 2020;86:181–192. doi: 10.1007/s00280-020-04106-z.
    1. Dubois SG, Shusterman S, Ingle AM, Ahern CH, Reid JM, Wu B, 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, et al. 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, et al. 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. Verschuur AC, Bajciova V, Mascarenhas L, Khosravan R, Lin X, Ingrosso A, et al. 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. European Medicines Agency. Guideline on the role of pharmacokinetics in the development of medicinal products in the paediatric population. Committee For Medicinal Products For Human Use, London. 2007. . Accessed 5 Nov 2019.
    1. U.S. Food and Drug Administration. General clinical pharmacology consideration for pediatric studies for drugs and biological products. Guidcance for industry. U.S. Department of Health and Human Services, Center for Drug Evaluation and Research (CDER), Bethesda, MD. 2014. . Accessed 5 Nov 2019.

Source: PubMed

3
Abonner