A Pharmacometric Framework for Axitinib Exposure, Efficacy, and Safety in Metastatic Renal Cell Carcinoma Patients

E Schindler, M A Amantea, M O Karlsson, L E Friberg, E Schindler, M A Amantea, M O Karlsson, L E Friberg

Abstract

The relationships between exposure, biomarkers (vascular endothelial growth factor (VEGF), soluble VEGF receptors (sVEGFR)-1, -2, -3, and soluble stem cell factor receptor (sKIT)), tumor sum of longest diameters (SLD), diastolic blood pressure (dBP), and overall survival (OS) were investigated in a modeling framework. The dataset included 64 metastatic renal cell carcinoma patients (mRCC) treated with oral axitinib. Biomarker timecourses were described by indirect response (IDR) models where axitinib inhibits sVEGFR-1, -2, and -3 production, and VEGF degradation. No effect was identified on sKIT. A tumor model using sVEGFR-3 dynamics as driver predicted SLD data well. An IDR model, with axitinib exposure stimulating the response, characterized dBP increase. In a time-to-event model the SLD timecourse predicted OS better than exposure, biomarker- or dBP-related metrics. This type of framework can be used to relate pharmacokinetics, efficacy, and safety to long-term clinical outcome in mRCC patients treated with VEGFR inhibitors. (ClinicalTrial.gov identifier NCT00569946.).

© 2017 The Authors CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Schematic representation of the modeling framework for axitinib in metastatic renal cell carcinoma (mRCC). Axitinib daily area under the curve (AUCdaily) was used as a driver of the timecourses of biomarkers (the vascular endothelial growth factor VEGF and its soluble receptors sVEGFR‐1, ‐2, and ‐3) and diastolic blood pressure (dBP). Biomarker timecourses were described by indirect response models where axitinib inhibits the loss of VEGF response and the production of sVEGFR‐1, ‐2, and ‐3 responses. sKIT was not affected by axitinib. The model describing tumor size (sum of longest diameters, SLD) included an exponential growth and an effect of the relative change in sVEGFR‐3 from baseline over time (sVEGFR‐3rel(t)) that induces tumor size reduction and washes out over time. The SLD timecourse (SLD(t)) was predictive of overall survival. KG, first‐order growth rate constant; kout, first‐order rate constant for the degradation or loss of response; ksVEGFR‐3, tumor size reduction rate constant related to sVEGFR‐3 response; λ, tumor resistance/regrowth appearance rate constant; Rin, zero‐order rate constant for the production of response. Dashed arrows represent relationships identified as significant.
Figure 2
Figure 2
Prediction‐corrected visual predictive checks of the final biomarker models based on 500 simulations. Median (solid line), 5th, and 95th percentiles (dashed lines) of the observed data (solid circles) are compared to the 95% confidence intervals (shaded areas) for the median, 5th, and 95th percentiles of the simulated data. VEGF, vascular endothelial growth factor; sVEGFR‐1, ‐2, ‐3, soluble VEGF receptor 1, 2, 3.
Figure 3
Figure 3
Visual predictive checks of the final sum of longest diameters (SLD, left) and diastolic blood pressure (dBP, right) models based on 500 simulations. Median (solid line), 5th, and 95th percentiles (dashed lines) of the observed data (solid circles) are compared to the 95% CIs (shaded areas) for the median, 5th, and 95th percentiles of the simulated data. Prediction‐correction was used for dBP. For the SLD model, dropout was taken into account in the simulations.
Figure 4
Figure 4
Kaplan–Meier visual predictive checks for the final overall survival model driven by the sum of longest diameters timecourse. The observed Kaplan–Meier curve (black line) is compared to the 95% CI (shaded area) derived from model simulations (200 samples). Vertical black lines represent censored observations.

References

    1. Ljungberg, B. et al EAU guidelines on renal cell carcinoma: 2014 update. Eur. Urol. 67, 913–924 (2015).
    1. Rothermundt, C. et al Second‐line treatment for metastatic clear cell renal cell cancer: experts' consensus algorithms. World J. Urol. 2016. [Epub ahead of print].
    1. Pfizer Laboratories Div Pfizer Inc. INLYTA — axitinib tablet, film coated: Full prescribing information. [cited 10/18/2016] Available from: <> 2012.
    1. Rini, B.I. et al Axitinib with or without dose titration for first‐line metastatic renal‐cell carcinoma: a randomised double‐blind phase 2 trial. Lancet Oncol. 14, 1233–1242 (2013).
    1. Hutson, T.E. et al Axitinib versus sorafenib in first‐line metastatic renal cell carcinoma: overall survival from a randomized phase III trial. Clin. Genitourin. Cancer 15, 72–76 (2017).
    1. Rini, B.I. et al Axitinib dose titration: analyses of exposure, blood pressure and clinical response from a randomized phase II study in metastatic renal cell carcinoma. Ann. Oncol. 26, 1372–1377 (2015).
    1. Eisenhauer, E.A. et al New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).
    1. Ammari, S. et al Radiological evaluation of response to treatment: application to metastatic renal cancers receiving anti‐angiogenic treatment. Diagn. Interv. Imaging 95, 527–539 (2014).
    1. Escalante, C.P. & Zalpour, A. Vascular endothelial growth factor inhibitor‐induced hypertension: basics for primary care providers. Cardiol. Res. Pract. 2011, 816897 (2011).
    1. Rini, B.I. et al Axitinib in metastatic renal cell carcinoma: results of a pharmacokinetic and pharmacodynamic analysis. J. Clin. Pharmacol. 53, 491–504 (2013).
    1. Rini, B.I. et al Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Clin. Cancer Res. 17, 3841–3849 (2011).
    1. Hansson, E.K. et al PKPD modeling of predictors for adverse effects and overall survival in sunitinib‐treated patients with GIST. CPT Pharmacometrics Syst. Pharmacol. 2, e85 (2013).
    1. George, S. et al Hypertension as a potential biomarker of efficacy in patients with gastrointestinal stromal tumor treated with sunitinib. Ann. Oncol. 23, 3180–3187 (2012).
    1. Zurita, A.J. , Jonasch, E. , Wu, H.K. , Tran, H.T. & Heymach, J.V. Circulating biomarkers for vascular endothelial growth factor inhibitors in renal cell carcinoma. Cancer 115(10 suppl.), 2346–2354 (2009).
    1. Fujiwara, Y. et al Management of axitinib (AG‐013736)‐induced fatigue and thyroid dysfunction, and predictive biomarkers of axitinib exposure: results from phase I studies in Japanese patients. Investig. New Drugs 30, 1055–1064 (2012).
    1. Eto, M. et al Overall survival and final efficacy and safety results from a Japanese phase II study of axitinib in cytokine‐refractory metastatic renal cell carcinoma. Cancer Sci. 105, 1576–1583 (2014).
    1. Bender B.C., Schindler, E. & Friberg, L.E. Population pharmacokinetic pharmacodynamic modeling in oncology: a tool for predicting clinical response. Br. J. Clin. Pharmacol. 79, 56–71 (2015).
    1. Mould, D.R. , Walz, A.C. , Lave, T. , Gibbs, J.P. & Frame, B. Developing exposure/response models for anticancer drug treatment: special considerations. CPT Pharmacometrics Syst. Pharmacol. 4, e00016 (2015).
    1. Venkatakrishnan, K. et al Optimizing oncology therapeutics through quantitative translational and clinical pharmacology: challenges and opportunities. Clin. Pharmacol. Ther. 97, 37–54 (2015).
    1. Ribba, B. et al A review of mixed‐effects models of tumor growth and effects of anticancer drug treatment used in population analysis. CPT Pharmacometrics Syst. Pharmacol. 3, e113 (2014).
    1. Hansson, E.K. et al PKPD modeling of VEGF, sVEGFR‐2, sVEGFR‐3, and sKIT as predictors of tumor dynamics and overall survival following sunitinib treatment in GIST. CPT Pharmacometrics Syst. Pharmacol. 2, e84 (2013).
    1. Tomita, Y. et al Key predictive factors of axitinib (AG‐013736)‐induced proteinuria and efficacy: a phase II study in Japanese patients with cytokine‐refractory metastatic renal cell Carcinoma. Eur. J. Cancer 47, 2592–2602 (2011).
    1. Beal, S. , Sheiner, L.B. , Boeckmann, A. & Bauer, R.J. NONMEM User's Guides. Icon Development Solutions, Ellicott City, MD; 2009. 1989–2009.
    1. Keizer, R.J. , Karlsson, M.O. & Hooker, A. Modeling and simulation workbench for NONMEM: Tutorial on Pirana, PsN, and Xpose. CPT Pharmacometrics Syst. Pharmacol. 2, e50 (2013).
    1. Bergstrand, M. , Hooker, A.C. , Wallin, J.E. & Karlsson, M.O. Prediction‐corrected visual predictive checks for diagnosing nonlinear mixed‐effects models. AAPS J. 13, 143–151 (2011).
    1. Petersson, K.J. , Hanze, E. , Savic, R.M. & Karlsson, MO. Semiparametric distributions with estimated shape parameters. Pharm. Res. 26, 2174–2185 (2009).
    1. Sharma, A. & Jusko, W.J. Characteristics of indirect pharmacodynamic models and applications to clinical drug responses. Br. J. Clin. Pharmacol. 45, 229–239 (1998).
    1. Zhang, L. , Beal, S.L. & Sheiner, L.B. Simultaneous vs. sequential analysis for population PK/PD data I: best‐case performance. J. Pharmacokinet. Pharmacodyn. 30, 387–404 (2003).
    1. Wade, J.R. & Karlsson, M.O. Combining PK and PD data during population PK/PD analysis []. PAGE 8; 1999.
    1. Lacroix, B.D. , Friberg, L.E. & Karlsson, M.O. Evaluation of IPPSE, an alternative method for sequential population PKPD analysis. J. Pharmacokinet. Pharmacodyn. 39, 177–193 (2012).
    1. Dosne, A.G. , Bergstrand, M. , Harling, K. & Karlsson, M.O. Improving the estimation of parameter uncertainty distributions in nonlinear mixed effects models using sampling importance resampling. J. Pharmacokinet. Pharmacodyn. 43, 583–596 (2016).
    1. Claret, L. et al Model‐based prediction of phase III overall survival in colorectal cancer on the basis of phase II tumor dynamics. J. Clin. Oncol. 27, 4103–4108 (2009).
    1. Dansirikul, C. , Silber, H.E. & Karlsson, M.O. Approaches to handling pharmacodynamic baseline responses. J. Pharmacokinet. Pharmacodyn. 35, 269–283 (2008).
    1. Claret, L. , Mercier, F. , Houk, B.E. , Milligan, P.A. & Bruno, R. Modeling and simulations relating overall survival to tumor growth inhibition in renal cell carcinoma patients. Cancer Chemother. Pharmacol. 76, 567–573 (2015).
    1. Schoemaker, R.C. , van Gerven, J.M. & Cohen, A.F. Estimating potency for the Emax‐model without attaining maximal effects. J. Pharmacokinet. Biopharm. 26, 581–593 (1998).
    1. Lindauer, A. et al Pharmacokinetic/pharmacodynamic modeling of biomarker response to sunitinib in healthy volunteers. Clin. Pharmacol. Ther. 87, 601–608 (2010).
    1. Ait‐Oudhia, S. et al Bridging sunitinib exposure to time‐to‐tumor progression in hepatocellular carcinoma patients with mathematical modeling of an angiogenic biomarker. CPT Pharmacometrics Syst. Pharmacol. 5, 297–304 (2016).
    1. Kanefendt, F. et al Modeling Sunitinib and Biomarker Response as potential Predictors of Time to Progression in Patients with Metastatic Colorectal Cancer. Annual Meeting of the Population Approach Group in Europe (PAGE) 2012.
    1. Bocci, G. et al Increased plasma vascular endothelial growth factor (VEGF) as a surrogate marker for optimal therapeutic dosing of VEGF receptor‐2 monoclonal antibodies. Cancer Res. 64, 6616–6625 (2004).
    1. Ebos, J.M. et al Vascular endothelial growth factor‐mediated decrease in plasma soluble vascular endothelial growth factor receptor‐2 levels as a surrogate biomarker for tumor growth. Cancer Res. 68, 521–529 (2008).
    1. Cohen, E.E. et al A phase II trial of axitinib in patients with various histologic subtypes of advanced thyroid cancer: long‐term outcomes and pharmacokinetic/pharmacodynamic analyses. Cancer Chemother. Pharmacol. 74, 1261–1270 (2014).
    1. Hu‐Lowe, D.D. et al Nonclinical antiangiogenesis and antitumor activities of axitinib (AG‐013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3. Clin. Cancer Res. 14, 7272–7283 (2008).
    1. Gross‐Goupil, M. , Francois, L. , Quivy, A. & Ravaud, A. Axitinib: a review of its safety and efficacy in the treatment of adults with advanced renal cell carcinoma. Clin. Med. Insights Oncol. 7, 269–277 (2013).
    1. Wasserstrum, Y. et al Hypertension in cancer patients treated with anti‐angiogenic based regimens. Cardio‐Oncology 1, 1–10 (2015).
    1. Chen, Y. , Rini, B.I. , Bair, A.H. , Mugundu, G.M. & Pithavala, Y.K. Population pharmacokinetic‐pharmacodynamic modeling of 24‐h diastolic ambulatory blood pressure changes mediated by axitinib in patients with metastatic renal cell carcinoma. Clin. Pharmacokinet. 54, 397–407 (2015).
    1. Rini, B.I. et al Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma: a randomised phase 3 trial. Lancet 378, 1931–1939 (2011).
    1. Mistry, H.B. Time‐dependent bias of tumour growth rate and time to tumour re‐growth. CPT Pharmacometrics Syst. Pharmacol. 5, 587 (2016).
    1. Bender, B.C. et al A population pharmacokinetic/pharmacodynamic model of thrombocytopenia characterizing the effect of trastuzumab emtansine (T‐DM1) on platelet counts in patients with HER2‐positive metastatic breast cancer. Cancer Chemother. Pharmacol. 70, 591–601 (2012).
    1. Holford, N. A time to event tutorial for pharmacometricians. CPT Pharmacometrics Syst. Pharmacol. 2, e43 (2013).
    1. Ribba, B. , Holford, N. & Mentre, F. The use of model‐based tumor‐size metrics to predict survival. Clin. Pharmacol. Ther. 96, 133–135 (2014).
    1. Chen, Y. et al Axitinib plasma pharmacokinetics and ethnic differences. Investig. New Drugs 33, 521–532 (2015).

Source: PubMed

3
購読する