Abstract

Background. Absent adaptive, individualized dose-finding in early-phase oncology trials, subsequent ‘confirmatory’ Phase III trials risk suboptimal dosing, with resulting loss of statistical power and reduced probability of technical success for the investigational therapy. While progress has been made toward explicitly adaptive dose-finding and quantitative modeling of dose-response relationships, most such work continues to be organized around a concept of ‘the’ maximum tolerated dose (MTD). The purpose of this paper is to demonstrate concretely how the aim of early-phase trials might be conceived, not as ‘dose-finding’, but as dose titration algorithm (DTA)-finding. Methods. A Phase I dosing study is simulated, for a notional cytotoxic chemotherapy drug, with neutropenia constituting the critical dose-limiting toxicity. The drug’s population pharmacokinetics and myelosuppression dynamics are simulated using published parameter estimates for docetaxel. The amenability of this model to linearization is explored empirically. The properties of a simple DTA targeting neutrophil nadir of 500 cells/mm 3 using a Newton-Raphson heuristic are explored through simulation in 25 simulated study subjects. Results. Individual-level myelosuppression dynamics in the simulation model approximately linearize under simple transformations of neutrophil concentration and drug dose. The simulated dose titration exhibits largely satisfactory convergence, with great variance in individualized optimal dosing. Some titration courses exhibit overshooting. Conclusions. The large inter-individual variability in simulated optimal dosing underscores the need to replace ‘the’ MTD with an individualized concept of MTDi . To illustrate this principle, the simplest possible DTA capable of realizing such a concept is demonstrated. Qualitative phenomena observed in this demonstration support discussion of the notion of tuning such algorithms. Although here illustrated specifically in relation to cytotoxic chemotherapy, the DTAT principle appears similarly applicable to Phase I studies of cancer immunotherapy and molecularly targeted agents.

Highlights

  • Despite advances in Bayesian adaptive designs1,2 and model-based dose-finding3, oncology dose-finding studies remain conceptually in the thrall of ‘the’ maximum tolerated dose (MTD)

  • This paper demonstrates individualized dose-finding in a simulated Phase I study of a cytotoxic chemotherapy drug for which neutropenia constitutes the critical dose-limiting toxicity

  • I have advanced a concept of dose titration algorithm tuning (DTAT), drawing illustrative and orienting connections with recursive filtering and optimal control

Read more

Summary

Introduction

Despite advances in Bayesian adaptive designs and model-based dose-finding, oncology dose-finding studies remain conceptually in the thrall of ‘the’ maximum tolerated dose (MTD). This fallacious concept stands opposed to the long-recognized heterogeneity of cancer patients’ pharmacokinetics and pharmacodynamics (PK/PD), and to the diversity of their individual values and goals of care. Even as Bayesianism has made progress toward the ethical imperative of efficient use of data in such studies, it has neglected to confront the distinct ethical dimension of individualism6 This seems a great irony, as the dynamic learning model of Bayesianism is suited, and essential, to solving the latter problem. The drug’s population pharmacokinetics and myelosuppression dynamics are simulated using published parameter estimates for docetaxel The amenability of this model to linearization is explored empirically.

Objectives
Methods
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call