skip to main content


Title: A phase I‐II design based on periodic and continuous monitoring of disease status and the times to toxicity and death

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
NSF-PAR ID:
10456747
Author(s) / Creator(s):
 ;  ;  
Publisher / Repository:
Wiley Blackwell (John Wiley & Sons)
Date Published:
Journal Name:
Statistics in Medicine
Volume:
39
Issue:
15
ISSN:
0277-6715
Page Range / eLocation ID:
p. 2035-2050
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. 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
  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 STUDY QUESTION

    To what extent is preconception maternal or paternal coronavirus disease 2019 (COVID-19) vaccination associated with miscarriage incidence?

    SUMMARY ANSWER

    COVID-19 vaccination in either partner at any time before conception is not associated with an increased rate of miscarriage.

    WHAT IS KNOWN ALREADY

    Several observational studies have evaluated the safety of COVID-19 vaccination during pregnancy and found no association with miscarriage, though no study prospectively evaluated the risk of early miscarriage (gestational weeks [GW] <8) in relation to COVID-19 vaccination. Moreover, no study has evaluated the role of preconception vaccination in both male and female partners.

    STUDY DESIGN, SIZE, DURATION

    An Internet-based, prospective preconception cohort study of couples residing in the USA and Canada. We analyzed data from 1815 female participants who conceived during December 2020–November 2022, including 1570 couples with data on male partner vaccination.

    PARTICIPANTS/MATERIALS, SETTING, METHODS

    Eligible female participants were aged 21–45 years and were trying to conceive without use of fertility treatment at enrollment. Female participants completed questionnaires at baseline, every 8 weeks until pregnancy, and during early and late pregnancy; they could also invite their male partners to complete a baseline questionnaire. We collected data on COVID-19 vaccination (brand and date of doses), history of SARS-CoV-2 infection (yes/no and date of positive test), potential confounders (demographic, reproductive, and lifestyle characteristics), and pregnancy status on all questionnaires. Vaccination status was categorized as never (0 doses before conception), ever (≥1 dose before conception), having a full primary sequence before conception, and completing the full primary sequence ≤3 months before conception. These categories were not mutually exclusive. Participants were followed up from their first positive pregnancy test until miscarriage or a censoring event (induced abortion, ectopic pregnancy, loss to follow-up, 20 weeks’ gestation), whichever occurred first. We estimated incidence rate ratios (IRRs) for miscarriage and corresponding 95% CIs using Cox proportional hazards models with GW as the time scale. We used propensity score fine stratification weights to adjust for confounding.

    MAIN RESULTS AND THE ROLE OF CHANCE

    Among 1815 eligible female participants, 75% had received at least one dose of a COVID-19 vaccine by the time of conception. Almost one-quarter of pregnancies resulted in miscarriage, and 75% of miscarriages occurred <8 weeks’ gestation. The propensity score-weighted IRR comparing female participants who received at least one dose any time before conception versus those who had not been vaccinated was 0.85 (95% CI: 0.63, 1.14). COVID-19 vaccination was not associated with increased risk of either early miscarriage (GW: <8) or late miscarriage (GW: 8–19). There was no indication of an increased risk of miscarriage associated with male partner vaccination (IRR = 0.90; 95% CI: 0.56, 1.44).

    LIMITATIONS, REASONS FOR CAUTION

    The present study relied on self-reported vaccination status and infection history. Thus, there may be some non-differential misclassification of exposure status. While misclassification of miscarriage is also possible, the preconception cohort design and high prevalence of home pregnancy testing in this cohort reduced the potential for under-ascertainment of miscarriage. As in all observational studies, residual or unmeasured confounding is possible.

    WIDER IMPLICATIONS OF THE FINDINGS

    This is the first study to evaluate prospectively the relation between preconception COVID-19 vaccination in both partners and miscarriage, with more complete ascertainment of early miscarriages than earlier studies of vaccination. The findings are informative for individuals planning a pregnancy and their healthcare providers.

    STUDY FUNDING/COMPETING INTEREST(S)

    This work was supported by the Eunice Kennedy Shriver National Institute of Child Health and Human Development, the National Institute of Health [R01-HD086742 (PI: L.A.W.); R01-HD105863S1 (PI: L.A.W. and M.L.E.)], the National Institute of Allergy and Infectious Diseases (R03-AI154544; PI: A.K.R.), and the National Science Foundation (NSF-1914792; PI: L.A.W.). The funders had no role in the study design, data collection, analysis and interpretation of data, writing of the report, or the decision to submit the paper for publication. L.A.W. is a fibroid consultant for AbbVie, Inc. She also receives in-kind donations from Swiss Precision Diagnostics (Clearblue home pregnancy tests) and Kindara.com (fertility apps). M.L.E. received consulting fees from Ro, Hannah, Dadi, VSeat, and Underdog, holds stock in Ro, Hannah, Dadi, and Underdog, is a past president of SSMR, and is a board member of SMRU. K.F.H. reports being an investigator on grants to her institution from UCB and Takeda, unrelated to this study. S.H.-D. reports being an investigator on grants to her institution from Takeda, unrelated to this study, and a methods consultant for UCB and Roche for unrelated drugs. The authors report no other relationships or activities that could appear to have influenced the submitted work.

    TRIAL REGISTRATION NUMBER

    N/A.

     
    more » « less
  4. Abstract Background

    Entinostat is an oral small molecule inhibitor of class I histone deacetylases (HDAC), which has not previously been evaluated in pediatrics. We conducted a phase I trial to determine the maximum tolerated dose/recommended phase 2 dose (MTD/RP2D), toxicity profile, pharmacokinetics (PK), and pharmacodynamics (PD) of entinostat in children with relapsed or refractory solid tumors including central nervous system (CNS) malignancies.

    Methods

    A rolling six dose escalation design evaluated two dose levels. Entinostat oral tablet formulation was administered once per week, four doses per 28‐day cycle. PK and PD studies were performed.

    Results

    Twenty‐one eligible patients’ median (range) age was 14 years (6‐20). Six subjects were treated at 3 mg/m2dose level and 15 were treated in 4 mg/m2dose level. The study included patients with CNS tumors (n = 12), sarcomas (n = 6), or other solid tumors (n = 3). Eight patients were not fully evaluable for toxicity due to progression of disease prior to receiving the required percentage of protocol therapy. No cycle one dose‐limiting toxicity (DLT) was observed at either dose level. A three‐fold higher area under the curve (AUC) was achieved in our cohort compared to adults using a similar dosing schedule. The PD studies showed increase in acetylated lysine in peripheral blood leukocytes at both doses.

    Conclusions

    Entinostat was well tolerated with no DLT observed. All patients experienced progression within the first two cycles, except one patient with ependymoma with stable disease. Based on PK and PD, the R2PD in pediatric patients with solid tumors is 4 mg/m2orally administered once weekly.

     
    more » « less
  5. 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