Population Pharmacokinetic Modeling of Lucitanib in Patients with Advanced Cancer

Mingxiang Liao, Jie Zhou, Kenton Wride, Denise Lepley, Terri Cameron, Mark Sale, Jim Xiao, Mingxiang Liao, Jie Zhou, Kenton Wride, Denise Lepley, Terri Cameron, Mark Sale, Jim Xiao

Abstract

Background: Lucitanib is an oral, potent, selective inhibitor of the tyrosine kinase activity of vascular endothelial growth factor receptors 1‒3, fibroblast growth factor receptors 1‒3, and platelet-derived growth factor receptors alpha/beta.

Objective: We aimed to develop a population pharmacokinetics (PopPK) model for lucitanib in patients with advanced cancers.

Methods: PopPK analyses were based on intensive and sparse oral pharmacokinetic data from 5 phase 1/2 clinical studies of lucitanib in a total of 403 patients with advanced cancers. Lucitanib was administered at 5‒30 mg daily doses as 1 of 2 immediate-release oral formulations: a film-coated tablet or a hard gelatin capsule.

Results: Lucitanib pharmacokinetics were best described by a 2-compartment model with zero-order release into the dosing compartment, followed by first-order absorption and first-order elimination. Large between-subject pharmacokinetic variability was partially explained by body weight. No effects of demographics or tumor type on lucitanib pharmacokinetics were observed. The model suggested that the formulation impacted release duration (tablet, 0.243 h; capsule, 0.814 h), but the effect was not considered clinically meaningful. No statistically significant effects were detected for concomitant cytochrome P450 (CYP) 3A4 inhibitors or inducers, CYP2C8 or P-glycoprotein inhibitors, serum albumin, mild/moderate renal impairment, or mild hepatic impairment. Concomitant proton pump inhibitors had no clinically significant effect on lucitanib absorption.

Conclusions: The PopPK model adequately described lucitanib pharmacokinetics. High between-subject pharmacokinetic variability supports a safety-based dose-titration strategy currently being used in an ongoing clinical study of lucitanib to optimize drug exposure and clinical benefit.

Trial registration: ClinicalTrials.gov Identifier: NCT01283945, NCT02053636, ISRCTN23201971, NCT02202746, NCT02109016.

Conflict of interest statement

Mingxiang Liao, Kenton Wride, Denise Lepley, Terri Cameron, and Jim Xiao are or were employees of Clovis Oncology, Inc. Mark Sale and Jie Zhou are or were paid consultants for Clovis Oncology, Inc.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
PopPK model structure. The pharmacokinetics of lucitanib were described by a 2-compartment linear model with a zero-order release into the dosing compartment, followed by first-order absorption and first-order elimination. Volume (Vc, Vp) and clearance (Q, CL) terms were proportional to Weightexponent (a power model of weight with exponents fixed to 1 for volume terms and 0.75 for clearance terms). Duration of release from the tablet formulation (0.243 h) differed from the capsule (0.814 h). CL clearance, Ka first-order absorption rate constant, PopPK population pharmacokinetics, Q intercompartmental clearance, Vc central volume, Vp peripheral volume
Fig. 2
Fig. 2
Goodness-of-fit plots for the final lucitanib PopPK model. a Population prediction (ETA = 0) versus observed; b individual prediction (ETA ≠ 0) versus observed; c prediction versus conditional weighted residual; and d time versus conditional weighted residual. Blue lines indicate smoothed means (locally estimated scatterplot smoothing, LOESS [34]). Black trend lines indicate lines of unity (intercept = 0, slope = 1). Observations (concentrations or residuals, black dots) from the same individual are connected by black lines. ETA random effects values for pharmacokinetic parameters, PopPK population pharmacokinetics
Fig. 3
Fig. 3
Visual predictive check for the final lucitanib PopPK model: time since first dose versus lucitanib concentration. PopPK population pharmacokinetics
Fig. 4
Fig. 4
Conditional weighted residuals by formulation. Black horizontal lines represent median values, and boxes correspond to the ranges of the first and third quartiles. Upper and lower whiskers extend from the box to the largest or smallest value, respectively, within 1.5 times the interquartile range. Observations outside the whisker range are represented as dots
Fig. 5
Fig. 5
Post hoc estimates of BSV of Ka by formulation. Black horizontal lines represent median values, and boxes correspond to the ranges of the first and third quartiles. Upper and lower whiskers extend from the box to the largest or smallest value, respectively, within 1.5 times the interquartile range. Observations outside the whisker range are represented as dots. BSV between-subject variability, ETA random effects values for pharmacokinetic parameters, Ka first-order absorption rate constant
Fig. 6
Fig. 6
Post hoc estimates of BSV of CL/F by formulation. Black horizontal lines represent median values, and boxes correspond to the ranges of the first and third quartiles. Upper and lower whiskers extend from the box to the largest or smallest value, respectively, within 1.5 times the interquartile range. Observations outside the whisker range are represented as dots. BSV between-subject variability, CL/F apparent clearance, ETA random effects values for pharmacokinetic parameters

References

    1. Sherwood LM, Parris EE, Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182–1186. doi: 10.1056/NEJM197111182852108.
    1. Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci. 2020;77:1745–1770. doi: 10.1007/s00018-019-03351-7.
    1. Dusek RL, Robillard L, Harding TC, Simmons AD, Nguyen M. Abstract 1214: enhancement of anti-PD-1 antitumor efficacy in syngenic preclinical models by the angiogenesis inhibitor lucitanib. Cancer Res. 2019;79:1214. doi: 10.1158/1538-7445.AM2019-1214.
    1. Guffanti F, Chilà R, Bello E, Zucchetti M, Zangarini M, Ceriani L, et al. In vitro and in vivo activity of lucitanib in FGFR1/2 amplified or mutated cancer models. Neoplasia. 2017;19:35–42. doi: 10.1016/j.neo.2016.11.008.
    1. Soria J, De Braud F, Cereda R, Bahleda R, Delmonte A, Angevin E, et al. First-in-man study of E-3810, a novel VEGFR and FGFR inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2011;29:TPS149. doi: 10.1200/jco.2011.29.15_suppl.tps149.
    1. Soria JC, DeBraud F, Bahleda R, Adamo B, Andre F, Dientsmann R, et al. Phase I/IIa study evaluating the safety, efficacy, pharmacokinetics, and pharmacodynamics of lucitanib in advanced solid tumors. Ann Oncol. 2014;25:2244–2251. doi: 10.1093/annonc/mdu390.
    1. Hui R, Pearson A, Cortes J, Campbell C, Poirot C, Azim HA, et al. Lucitanib for the treatment of HR+/HER2− Metastatic breast cancer: results from the multicohort phase II FINESSE study. Clin Cancer Res. 2020;26:354–363. doi: 10.1158/1078-0432.CCR-19-1164.
    1. Campone M, Bachelot T, Penault-Llorca F, Pallis A, Agrapart V, Pierrat MJ, et al. A phase Ib dose allocation study of oral administration of lucitanib given in combination with fulvestrant in patients with estrogen receptor-positive and FGFR1-amplified or non-amplified metastatic breast cancer. Cancer Chemother Pharmacol. 2019;83:743–753. doi: 10.1007/s00280-018-03765-3.
    1. Abu-Khalaf M, Mayer IA, Tankersley C, Moy J, Allen A, Vogel C, et al. A phase 2, randomized, pen-label study of lucitanib in patients with FGF aberrant metastatic breast cancer. J Clin Oncol. 2015;33:TPS628. doi: 10.1200/jco.2015.33.3_suppl.628.
    1. . A study to assess the efficacy and safety of the VEGFR-FGFR inhibitor, lucitanib, given to patients with advanced/metastatic lung cancer and FGF, VEGF, or PDGF related genetic alterations. Bethesda: US National Library of Medicine. 2019. . Accessed 24 Jun 2021.
    1. Mayer I, Arteaga C, Nanda R, Miller K, Jhaveri K, Brufsky A, et al. Abstract P6–11-03: a phase 2 open-label study of lucitanib in patients (pts) with FGF aberrant metastatic breast cancer (MBC) Cancer Res. 2017;77:P6-11-03. doi: 10.1158/1538-7445.SABCS16-P6-11-03.
    1. Herbrink M, Nuijen B, Schellens JHM, Beijnen JH. Variability in bioavailability of small molecular tyrosine kinase inhibitors. Cancer Treat Rev. 2015;41:412–422. doi: 10.1016/j.ctrv.2015.03.005.
    1. Tyson RJ, Park CC, Powell JR, Patterson JH, Weiner D, Watkins PB, et al. Precision dosing priority criteria: drug, disease, and patient population variables. Front Pharmacol. 2020;11:420. doi: 10.3389/fphar.2020.00420.
    1. Schmidinger M, Danesi R, Jones R, McDermott R, Pyle L, Rini B, et al. Individualized dosing with axitinib: rationale and practical guidance. Future Oncol. 2018;14:861–875. doi: 10.2217/fon-2017-0455.
    1. Bekaii-Saab TS, Ou FS, Ahn DH, Boland PM, Ciombor KK, Heying EN, et al. Regorafenib dose-optimisation in patients with refractory metastatic colorectal cancer (ReDOS): a randomised, multicentre, open-label, phase 2 study. Lancet Oncol. 2019;20:1070–1082. doi: 10.1016/S1470-2045(19)30272-4.
    1. Keizer RJ, Gupta A, Shumaker R, Beijnen JH, Schellens JHM, Huitema ADR. Model-based treatment optimization of a novel VEGFR inhibitor. Br J Clin Pharmacol. 2012;74:315–326. doi: 10.1111/j.1365-2125.2012.04197.x.
    1. Loriot Y, Necchi A, Park SH, Garcia-Donas J, Huddart R, Burgess E, et al. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N Engl J Med. 2019;381:338–348. doi: 10.1056/NEJMoa1817323.
    1. Sala F, Bagnati R, Livi V, Cereda R, D’Incalci M, Zucchetti M. Development and validation of a high-performance liquid chromatography-tandem mass spectrometry method for the determination of the novel inhibitor of angiogenesis E-3810 in human plasma and its application in a clinical pharmacokinetic study. J Mass Spectrom. 2011;46:1039–1045. doi: 10.1002/jms.1985.
    1. Yu G, Zheng QS, Wang DX, Zhou HH, Li GF. Drug interactions between tyrosine-kinase inhibitors and acid suppressive agents: more than meets the eye. Lancet Oncol. 2014;15:e469–e470. doi: 10.1016/S1470-2045(14)70458-9.
    1. Sharma M, Holmes HM, Mehta HB, Chen H, Aparasu RR, Shih YCT, et al. The concomitant use of tyrosine kinase inhibitors and proton pump inhibitors: prevalence, predictors, and impact on survival and discontinuation of therapy in older adults with cancer. Cancer. 2019;125:1155–1162. doi: 10.1002/cncr.31917.
    1. van Leeuwen RWF, Jansman FGA, Hunfeld NG, Peric R, Reyners AKL, Imholz ALT, et al. Tyrosine kinase inhibitors and proton pump inhibitors: an evaluation of treatment options. Clin Pharmacokinet. 2017;56:683–688. doi: 10.1007/s40262-016-0503-3.
    1. Cancer Therapy Evaluation Program. Specific instructions for the use of protocol templates for organ dysfunction studies Bethesda (MD): National Cancer Institute, Division of Cancer Treatment & Diagnosis. 2021. . Accessed 24 June 2021.
    1. US Food and Drug Administration. Guidance for industry: pharmacokinetics in patients with impaired renal function: study design, data analysis, and impact on dosing. Silver Spring: US Department of Health and Human Services. 2020. . Accessed 24 June 2021.
    1. DIDB—The Drug Interaction Database—UW Drug Interaction Solutions. . Accessed 24 June 2022.
    1. Faslodex (fulvestrant) injection [prescribing information]. Cambridge: AstraZeneca, Plc; 2020. .
    1. Dumbrava E, Shapiro G, Bendell J, Yap T, Jeselsohn R, Lepley D, et al. Phase 1b/2 SEASTAR trial: safety, pharmacokinetics, and preliminary efficacy of the poly(ADP)-ribose polymerase (PARP) inhibitor rucaparib and angiogenesis inhibitor lucitanib in patients with advanced solid tumors. J Clin Oncol. 2021;39:3102. doi: 10.1200/JCO.2021.39.15_suppl.3102.
    1. Hamilton EP, Patel MR, Gunderson CC, Wride K, Lepley D, Dusek R, et al. 556P Initial clinical experience of lucitanib + nivolumab in advanced metastatic solid tumours: data from the phase Ib/II LIO-1 study (CO-3810-101; NCT04042116) Ann Oncol. 2020;31:S480–S481. doi: 10.1016/j.annonc.2020.08.670.
    1. Chen Y, Tortorici MA, Garrett M, Hee B, Klamerus KJ, Pithavala YK. Clinical pharmacology of axitinib. Clin Pharmacokinet. 2013;52:713–725. doi: 10.1007/s40262-013-0068-3.
    1. Rini BI, Melichar B, Ueda T, Grünwald V, Fishman MN, Arranz JA, et al. Axitinib with or without dose titration for first-line metastatic renal-cell carcinoma: a randomised double-blind phase 2 trial. Lancet Oncol. 2013;14:1233–1242. doi: 10.1016/S1470-2045(13)70464-9.
    1. Keytruda (pembrolizumab) injection [prescribing information]. Kenilworth, NJ: Merck & Co., Inc; 2021. . Accessed 26 July 2021.
    1. Bavencio (avelumab) injection [prescribing information]. Rockland, MA: EMD Serono Inc; 2021. . Accessed 26 July 2021.
    1. Hamilton EP, Jackson CC, Eskander R, Backes F, Makker V, Musa F, et al. LIO-1: Lucitanib + nivolumab in patients with advanced solid tumors—updated phase 1b results and initial experience in phase 2 ovarian cancer cohort. J Clin Oncol. 2021;39:5538. doi: 10.1200/JCO.2021.39.15_suppl.5538.
    1. Concin N, Gonzalez Martin A, Vergote I, Pignata S, Harter P, Patel MR, et al. 885TiP LIO-1: a phase II study of lucitanib + nivolumab in patients (pts) with gynaecological tumours (CO-3810-101; NCT04042116; ENGOT-GYN3/AGO/LIO) Ann Oncol. 2020;31:S648. doi: 10.1016/j.annonc.2020.08.2091.
    1. Cleveland WS. LOWESS: a program for smoothing scatterplots by robust locally weighted regression. Am Stat. 1981;35:54. doi: 10.2307/2683591.

Source: PubMed

3
Iratkozz fel