Linking Tumor Growth Dynamics to Survival in Ipilimumab-Treated Patients With Advanced Melanoma Using Mixture Tumor Growth Dynamic Modeling

Yan Feng, Xiaoning Wang, Satyendra Suryawanshi, Akintunde Bello, Amit Roy, Yan Feng, Xiaoning Wang, Satyendra Suryawanshi, Akintunde Bello, Amit Roy

Abstract

Early tumor assessments have been widely used to predict overall survival (OS), with potential application to dose selection and early go/no-go decisions. Most published tumor dynamic models assume a uniform pattern of tumor growth dynamics (TGDs). We developed a mixture TGD model to characterize different patterns of longitudinal tumor sizes. Data from 688 patients with advanced melanoma who received ipilimumab 3 or 10 mg/kg every 3 weeks in a phase III study (NCT01515189) were used in a TGD-OS analysis. The mixture model described TGD profiles using three subpopulations (no-growth, intermediate, and fast). The TGD model showed a positive exposure/dose-response (i.e., a higher proportion of patients in no/intermediate growth subpopulations and a lower tumor growth rate with ipilimumab 10 mg/kg relative to the 3 mg/kg dose). Finally, the mixture TGD model-based measures of tumor response provided better predictions of OS compared with the nonmixture model.

Conflict of interest statement

Y.F., S.S., A.B., and A.R. are employees of Bristol‐Myers Squibb. Y.F., A.B., A.R., and X.W. hold stock in Bristol‐Myers Squibb.

© 2019 Bristol-Myers Squibb CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals, Inc. on behalf of the American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
The distribution of tumor growth dynamics (TGDs) mixture model parameters by subpopulation: no‐growth, intermediate tumor growth (TG) and tumor shrinkage (TS), and fast TG. The box plot shows the median and interquartile range of TGD parameter estimates in each group. SLD, sum of the longest diameter.
Figure 2
Figure 2
Individual time course of (a) observed and (b) predicted change in tumor size from baseline stratified by dose and subpopulation. SLD, sum of the longest diameter; TG, tumor growth; TS, tumor shrinkage.
Figure 3
Figure 3
Effect of covariates on the hazard ratio of overall survival (OS‐model 1). Missing postbaseline tumor burden (TB), patients only had baseline TB assessment and without post treatment TB assessment. CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; LDH, lactate dehydrogenase; PRW8.MIXC, progressive rate at week 8 from covariate mixture model; ULN, upper limit of normal.
Figure 4
Figure 4
Model evaluation of overall survival (OS)‐model 1 analysis stratified (a) by treatment and (b) by treatment and subpopulation. PI, prediction interval; TG, tumor growth; TS, tumor shrinkage.

References

    1. Wang, Y. et al Elucidation of relationship between tumor size and survival in non‐small‐cell lung cancer patients can aid early decision making in clinical drug development. Clin. Pharmacol. Ther. 86, 167–174 (2009).
    1. Stein, W.D. et al Tumor regression and growth rates determined in five intramural NCI prostate cancer trials: the growth rate constant as an indicator of therapeutic efficacy. Clin. Cancer Res. 17, 907–917 (2011).
    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. Tham, L.S. et al A pharmacodynamic model for the time course of tumor shrinkage by gemcitabine + carboplatin in non‐small cell lung cancer patients. Clin. Cancer Res. 14, 4213–4218 (2008).
    1. Houk, B.E. , Bello, C.L. , Poland, B. , Rosen, L.S. , Demetri, G.D. & Motzer, R.J. Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer: results of a pharmacokinetic/pharmacodynamic meta‐analysis. Cancer Chemother. Pharmacol. 66, 357–371 (2010).
    1. Bonate, L. Modeling tumor growth in oncology In Pharmacokinetics in Drug Development: Advances and Applications (Bonate P.L. and Howard D.R., eds.) 1–19 (Springer, New York, NY, 2011).
    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. Feng, Y. , Agrawal, S. , Gupta, M. , Gupta, A.K. , Masson, E. & Roy, A. Association between immune‐checkpoint inhibitor induced tumor shrinkage and overall survival in advanced melanoma and NSCLC. J. Clin. Oncol. 32, abstract 3053 (2014).
    1. Suryawanshi, S. , Godfrey, C.J. , Roy, A. , Simonsen, K. , French, J. & Gupta, M. Leveraging longitudinal tumor data for prediction of overall survival from I‐O therapy: a proof of concept with quantitative models and external validation. J. Pharmacokinet. Pharmacodyn. 42, S11–S107 (2015).
    1. Claret, L. et al A model of overall survival predicts treatment outcomes with atezolizumab vs chemotherapy in non‐small cell lung cancer based on early tumor kinetics. Clin. Cancer Res. 24, 3292–3298 (2018).
    1. Zheng, Y. et al Population modeling of tumor kinetics and overall survival to identify prognostic and predictive biomarkers of efficacy for durvalumab in patients with urothelial carcinoma. Clin. Pharmacol. Ther. 103, 643–652 (2018).
    1. Chatterjee, M.S. et al Population pharmacokinetic/pharmacodynamic modeling of tumor size dynamics in pembrolizumab‐treated advanced melanoma. CPT Pharmacometrics Syst. Pharmacol. 6, 29–39 (2017).
    1. Pearson, K. Contributions to the mathematical theory of evolution. Philos. Trans. R. Soc. Lond. A 185, 71–110 (1894).
    1. Feng, Y. et al Population pharmacokinetic analysis for risperidone using highly sparse sampling measurements from the CATIE study. Br. J. Clin. Pharmacol. 66, 629–639 (2008).
    1. Choi, S. , Zhu, L. & Huang, X. Semiparametric accelerated failure time cure rate mixture models with competing risks. Stat. Med. 37, 48–59 (2018).
    1. Sondak, V.K. , Smalley, K.S. , Kudchadkar, R. , Grippon, S. & Kirkpatrick, P. Ipilimumab. Nat. Rev. Drug Discov. 10, 411–412 (2011).
    1. Hoos, A. et al Development of ipilimumab: contribution to a new paradigm for cancer immunotherapy. Semin. Oncol. 37, 533–546 (2010).
    1. Hodi, F.S. et al Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–713 (2010).
    1. Robert, C. et al Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 364, 2517–2526 (2011).
    1. Wolchok, J.D. et al Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double‐blind, multicentre, phase 2, dose‐ranging study. Lancet Oncol. 11, 155–164 (2011).
    1. Ascierto, P.A. et al Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double‐blind, multicentre, phase 3 trial. Lancet Oncol. 18, 611–622 (2017).
    1. Vezina, H.E. , Wang, S. , Statkevich, P. , Bello, A. & Roy, A. Exposure‐response analysis to support benefit‐risk assessment of ipilimumab 3 vs 10 mg/kg in patients with advanced melanoma. J. Pharmacokinet. Pharmacodyn. 45, S3–S134 (2018).

Source: PubMed

3
購読する