skip to main content


Title: Precision Bayesian phase I‐II dose‐finding based on utilities tailored to prognostic subgroups

A Bayesian phase I‐II design is presented that optimizes the dose of a new agent within predefined prognostic subgroups. The design is motivated by a trial to evaluate targeted agents for treating metastatic clear cell renal carcinoma, where a prognostic risk score defined by clinical variables and biomarkers is well established. Two clinical outcomes are used for dose‐finding, time‐to‐toxicity during a prespecified follow‐up period, and efficacy characterized by ordinal disease status evaluated at the end of follow‐up. A joint probability model is constructed for these outcomes as functions of dose and subgroup. The model performs adaptive clustering of adjacent subgroups having similar dose‐outcome distributions to facilitate borrowing information across subgroups. To quantify toxicity‐efficacy risk‐benefit trade‐offs that may differ between subgroups, the objective function is based on outcome utilities elicited separately for each subgroup. In the context of the renal cancer trial, a design is constructed and a simulation study is presented to evaluate the design's reliability, safety, and robustness, and to compare it to designs that either ignore subgroups or run a separate trial within each subgroup.

 
more » « less
NSF-PAR ID:
10445451
Author(s) / Creator(s):
 ;  ;  
Publisher / Repository:
Wiley Blackwell (John Wiley & Sons)
Date Published:
Journal Name:
Statistics in Medicine
Volume:
40
Issue:
24
ISSN:
0277-6715
Page Range / eLocation ID:
p. 5199-5217
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. A Bayesian phase I‐II dose‐finding design is presented for a clinical trial with four coprimary outcomes that reflect the actual clinical observation process. During a prespecified fixed follow‐up period, the times to disease progression, toxicity, and death are monitored continuously, and an ordinal disease status variable, including progressive disease (PD) as one level, is evaluated repeatedly by scheduled imaging. We assume a proportional hazards model with piecewise constant baseline hazard for each continuous variable and a longitudinal multinomial probit model for the ordinal disease status process and include multivariate patient frailties to induce association among the outcomes. A finite partition of the nonfatal outcome combinations during the follow‐up period is constructed, and the utility of each set in the partition is elicited. Posterior mean utility is used to optimize each patient's dose, subject to a safety rule excluding doses with an unacceptably high rate of PD, severe toxicity, or death. A simulation study shows that, compared with the proposed design, a simpler design based on commonly used efficacy and toxicity outcomes obtained by combining the four variables described above performs poorly and has substantially smaller probabilities of correctly choosing truly optimal doses and excluding truly unsafe doses.

     
    more » « less
  2. Summary

    A sequentially adaptive Bayesian design is presented for a clinical trial of cord-blood-derived natural killer cells to treat severe haematologic malignancies. Given six prognostic subgroups defined by disease type and severity, the goal is to optimize cell dose in each subgroup. The trial has five co-primary outcomes: the times to severe toxicity, cytokine release syndrome, disease progression or response and death. The design assumes a multivariate Weibull regression model, with marginals depending on dose, subgroup and patient frailties that induce association between the event times. Utilities of all possible combinations of the non-fatal outcomes over the first 100 days following cell infusion are elicited, with posterior mean utility used as a criterion to optimize the dose. For each subgroup, the design stops accrual to doses having an unacceptably high death rate and at the end of the trial selects the optimal safe dose. A simulation study is presented to validate the design's safety, ability to identify optimal doses and robustness, and to compare it with a simplified design that ignores patient heterogeneity.

     
    more » « less
  3. Abstract

    A Bayesian design is proposed for randomized phase II clinical trials that screen multiple experimental treatments compared to an active control based on ordinal categorical toxicity and response. The underlying model and design account for patient heterogeneity characterized by ordered prognostic subgroups. All decision criteria are subgroup specific, including interim rules for dropping unsafe or ineffective treatments, and criteria for selecting optimal treatments at the end of the trial. The design requires an elicited utility function of the two outcomes that varies with the subgroups. Final treatment selections are based on posterior mean utilities. The methodology is illustrated by a trial of targeted agents for metastatic renal cancer, which motivated the design methodology. In the context of this application, the design is evaluated by computer simulation, including comparison to three designs that conduct separate trials within subgroups, or conduct one trial while ignoring subgroups, or base treatment selection on estimated response rates while ignoring toxicity.

     
    more » « less
  4. An immunotherapy trial often uses the phase I/II design to identify the optimal biological dose, which monitors the efficacy and toxicity outcomes simultaneously in a single trial. The progression-free survival rate is often used as the efficacy outcome in phase I/II immunotherapy trials. As a result, patients developing disease progression in phase I/II immunotherapy trials are generally seriously ill and are often treated off the trial for ethical consideration. Consequently, the happening of disease progression will terminate the toxicity event but not vice versa, so the issue of the semi-competing risks arises. Moreover, this issue can become more intractable with the late-onset outcomes, which happens when a relatively long follow-up time is required to ascertain progression-free survival. This paper proposes a novel Bayesian adaptive phase I/II design accounting for semi-competing risks outcomes for immunotherapy trials, referred to as the dose-finding design accounting for semi-competing risks outcomes for immunotherapy trials (SCI) design. To tackle the issue of the semi-competing risks in the presence of late-onset outcomes, we re-construct the likelihood function based on each patient's actual follow-up time and develop a data augmentation method to efficiently draw posterior samples from a series of Beta-binomial distributions. We propose a concise curve-free dose-finding algorithm to adaptively identify the optimal biological dose using accumulated data without making any parametric dose–response assumptions. Numerical studies show that the proposed SCI design yields good operating characteristics in dose selection, patient allocation, and trial duration. 
    more » « less
  5. Abstract Overly restrictive eligibility criteria for clinical trials may limit the generalizability of the trial results to their target real-world patient populations. We developed a novel machine learning approach using large collections of real-world data (RWD) to better inform clinical trial eligibility criteria design. We extracted patients’ clinical events from electronic health records (EHRs), which include demographics, diagnoses, and drugs, and assumed certain compositions of these clinical events within an individual’s EHRs can determine the subphenotypes—homogeneous clusters of patients, where patients within each subgroup share similar clinical characteristics. We introduced an outcome-guided probabilistic model to identify those subphenotypes, such that the patients within the same subgroup not only share similar clinical characteristics but also at similar risk levels of encountering severe adverse events (SAEs). We evaluated our algorithm on two previously conducted clinical trials with EHRs from the OneFlorida+ Clinical Research Consortium. Our model can clearly identify the patient subgroups who are more likely to suffer or not suffer from SAEs as subphenotypes in a transparent and interpretable way. Our approach identified a set of clinical topics and derived novel patient representations based on them. Each clinical topic represents a certain clinical event composition pattern learned from the patient EHRs. Tested on both trials, patient subgroup (#SAE=0) and patient subgroup (#SAE>0) can be well-separated by k-means clustering using the inferred topics. The inferred topics characterized as likely to align with the patient subgroup (#SAE>0) revealed meaningful combinations of clinical features and can provide data-driven recommendations for refining the exclusion criteria of clinical trials. The proposed supervised topic modeling approach can infer the clinical topics from the subphenotypes with or without SAEs. The potential rules for describing the patient subgroups with SAEs can be further derived to inform the design of clinical trial eligibility criteria. 
    more » « less