Bayesian dose regimen assessment in early phase oncology incorporating pharmacokinetics and pharmacodynamics

Emma Gerard, Sarah Zohar, Hoai-Thu Thai, Christelle Lorenzato, Marie-Karelle Riviere, Moreno Ursino, Emma Gerard, Sarah Zohar, Hoai-Thu Thai, Christelle Lorenzato, Marie-Karelle Riviere, Moreno Ursino

Abstract

Phase I dose-finding trials in oncology seek to find the maximum tolerated dose of a drug under a specific schedule. Evaluating drug schedules aims at improving treatment safety while maintaining efficacy. However, while we can reasonably assume that toxicity increases with the dose for cytotoxic drugs, the relationship between toxicity and multiple schedules remains elusive. We proposed a Bayesian dose regimen assessment method (DRtox) using pharmacokinetics/pharmacodynamics (PK/PD) to estimate the maximum tolerated dose regimen (MTD-regimen) at the end of the dose-escalation stage of a trial. We modeled the binary toxicity via a PD endpoint and estimated the dose regimen toxicity relationship through the integration of a dose regimen PD model and a PD toxicity model. For the first model, we considered nonlinear mixed-effects models, and for the second one, we proposed the following two Bayesian approaches: a logistic model and a hierarchical model. In an extensive simulation study, the DRtox outperformed traditional designs in terms of proportion of correctly selecting the MTD-regimen. Moreover, the inclusion of PK/PD information helped provide more precise estimates for the entire dose regimen toxicity curve; therefore the DRtox may recommend alternative untested regimens for expansion cohorts. The DRtox was developed to be applied at the end of the dose-escalation stage of an ongoing trial for patients with relapsed or refractory acute myeloid leukemia (NCT03594955) once all toxicity and PK/PD data are collected.

Keywords: Bayesian inference; dose regimen; early phase oncology; hierarchical model; pharmacokinetics/pharmacodynamics; toxicity.

© 2021 The Authors. Biometrics published by Wiley Periodicals LLC on behalf of International Biometric Society.

Figures

FIGURE 1
FIGURE 1
Trial scheme: the DRtox method is applied at the end of the dose‐escalation stage of a phase I trial
FIGURE 2
FIGURE 2
Concentration (up) and cytokine (down) profiles of two patients, one receiving a dose regimen with intrapatient escalation in solid line and the other receiving a dose regimen without intrapatient escalation in dashed line, administered on days 1, 5, 9, 13, 17, 21, and 25. Horizontal lines represent the maximum peak of cytokine observed after each dose regimen
FIGURE 3
FIGURE 3
The first three subplots represent the panel of dose regimens from S1 in spaced dashed line to S6 in solid line, for the three main scenarios, where the type of points is specific to each scenario. In the last subplot in the lower right corner, the dose regimen toxicity relationship is represented for each scenario, where the MTD‐regimen is the dose regimen having the toxicity probability the closest to the target δT, plotted in dashed line
FIGURE 4
FIGURE 4
Violin plots of the estimated toxicity probabilities in an additional scenario in which the dose regimen panel missed the true MTD‐regimen and in Scenario 3 on 1000 trials implemented with the CRM including 30 patients. The predicted toxicity probability of a new regimen Snew is framed in dotted line. Horizontal lines on the density estimates represent the median and first and third quantiles of the distributions and the plus sign represents the mean. The dashed line represents the toxicity target and the solid line represents the true toxicity probabilities

References

    1. Berry, S.M. , Carlin, B.P. , Lee, J.J. and Muller, P. (2010) Bayesian adaptive methods for clinical trials. Boca Raton, FL: CRC Press.
    1. Boissel, N. , de Botton, S. , Thomas, X.G. , Rao, E. , Bonnevaux, H. , Rubin‐Carrez, C. et al. (2018) An open‐label, first‐in‐human, dose escalation study of a novel CD3‐CD123 bispecific T‐cell engager administered as a single agent by intravenous infusion in patients with relapsed or refractory acute myeloid leukemia, B‐cell acute lymphoblastic leukemia, or high risk myelodysplastic syndrome. American Society of Clinical Oncology, 36, TPS7076‐TPS7076.
    1. Braun, T.M. , Thall, P.F. , Nguyen, H. and de Lima, M. (2007) Simultaneously optimizing dose and schedule of a new cytotoxic agent. Clinical Trials, 4, 113–124.
    1. Braun, T.M. , Yuan, Z. and Thall, P.F. (2005) Determining a maximum‐tolerated schedule of a cytotoxic agent. Biometrics, 61, 335–343.
    1. Bullock, J.M. , Lin, T. and Bilic, S. (2017) Clinical pharmacology tools and evaluations to facilitate comprehensive dose finding in oncology: a continuous risk‐benefit approach. The Journal of Clinical Pharmacology, 57, S105–S115.
    1. Chen, X. , Kamperschroer, C. , Wong, G. and Xuan, D. (2019) A modeling framework to characterize cytokine release upon T‐cell–engaging bispecific antibody treatment: methodology and opportunities. Clinical and Translational Science, 12(6), 600–608.
    1. Cheung, Y.K. (2011) Dose finding by the continual reassessment method. Boca Raton, FL: CRC Press.
    1. Chevret, S. (2006) Statistical methods for dose‐finding experiments (Vol. 24). Chichester: Wiley.
    1. Crowley, J. and Hoering, A. (2012) Handbook of statistics in clinical oncology. Boca Raton, FL: CRC Press.
    1. Gelman, A. (2006) Prior distributions for variance parameters in hierarchical models (comment on article by Browne and Draper). Bayesian Analysis, 1(3), 515–534.
    1. Günhan, B.K. , Weber, S. and Friede, T. (2020) A Bayesian time‐to‐event pharmacokinetic model for phase I dose‐escalation trials with multiple schedules. Statistics in Medicine, 39(27), 3986–4000.
    1. Guo, B. , Li, Y. and Yuan, Y. (2016) A dose–schedule finding design for phase I–II clinical trials. Journal of the Royal Statistical Society: Series C (Applied Statistics), 65, 259–272.
    1. Li, Y. , Bekele, B.N. , Ji, Y. and Cook, J.D. (2008) Dose–schedule finding in phase I/II clinical trials using a Bayesian isotonic transformation. Statistics in Medicine, 27, 4895–4913.
    1. Liu, C.A. and Braun, T.M. (2009) Parametric non‐mixture cure models for schedule finding of therapeutic agents. Journal of the Royal Statistical Society: Series C (Applied Statistics), 58, 225–236.
    1. Lixoft SAS (2019) Monolix version 2019R1. Antony, France. Available at:
    1. Lyu, J. , Curran, E. and Ji, Y. (2018) Bayesian adaptive design for finding the maximum tolerated sequence of doses in multicycle dose‐finding clinical trials. JCO Precision Oncology, 2, 1–19.
    1. Morita, S. , Thall, P.F. and Müller, P. (2008) Determining the effective sample size of a parametric prior. Biometrics, 64(2), 595–602.
    1. Musuamba, F.T. , Manolis, E. , Holford, N. , Cheung, S.Y.A. , Friberg, L.E. , Ogungbenro, K. et al. (2017) Advanced methods for dose and regimen finding during drug development: summary of the EMA/EFPIA workshop on dose finding (London 4–5 December 2014). CPT: Pharmacometrics and Systems Pharmacology, 6(7), 418–429.
    1. O'Quigley, J. , Pepe, M. and Fisher, L. (1990) Continual reassessment method: a practical design for phase 1 clinical trials in cancer. Biometrics, 33‐48.
    1. R Core Team (2018) R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing. Available at: .
    1. Schmoor, C. and Schumacher, M. (1992) Adaptive statistical procedures for the analysis of nonmonotone dose–response relationships. Biometrie und Informatik in Medizin und Biologi, 23, 113–126.
    1. Shimabukuro‐Vornhagen, A. , Gödel, P. , Subklewe, M. , Stemmler, H.J. , Schlößer, H.A. , Schlaak, M. et al. (2018) Cytokine release syndrome. Journal For Immunotherapy Of Cancer, 6, 56.
    1. Stan Development Team (2019) RStan: the R interface to Stan. R package version 2.19.2. Available at: .
    1. Storer, B.E. (1989) Design and analysis of phase I clinical trials. Biometrics, 45, 925–937.
    1. Teachey, D.T. , Lacey, S.F. , Shaw, P.A. , Melenhorst, J.J. , Maude, S.L. , Frey, N. et al. (2016) Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T‐cell therapy for acute lymphoblastic leukemia. Cancer Discovery, 6(6), 664–679.
    1. Thall, P.F. , Nguyen, H.Q. , Braun, T.M. and Qazilbash, M.H. (2013) Using joint utilities of the times to response and toxicity to adaptively optimize schedule–dose regimes. Biometrics, 69, 673–682.
    1. Ursino, M. , Zohar, S. , Lentz, F. , Alberti, C. , Friede, T. , Stallard, N. et al. (2017) Dose‐finding methods for Phase I clinical trials using pharmacokinetics in small populations. Biometrical Journal, 59, 804–825.
    1. US Department of Health and Human Services (2017) Common terminology criteria for adverse events (CTCAE) v5.0.
    1. Wages, N.A. , O'Quigley, J. and Conaway, M.R. (2014) Phase I design for completely or partially ordered treatment schedules. Statistics in Medicine, 33, 569–579.
    1. Yuan, Y. , Nguyen, H.Q. and Thall, P.F. (2017) Bayesian designs for phase I‐II clinical trials. Boca Raton, FL: CRC Press.
    1. Zhang, J. and Braun, T.M. (2013) A phase I Bayesian adaptive design to simultaneously optimize dose and schedule assignments both between and within patients. Journal of the American Statistical Association, 108, 892–901.
    1. Zhu, M. , Wu, B. , Brandl, C. , Johnson, J. , Wolf, A. , Chow, A. et al. (2016) Blinatumomab, a bispecific T‐cell engager (BiTE®) for CD‐19 targeted cancer immunotherapy: clinical pharmacology and its implications. Clinical Pharmacokinetics, 55(10), 1271–1288.

Source: PubMed

3
Abonnieren