Next Article in Journal
Assessment of Different Castration Resistance Definitions and Staging Modalities in Metastatic Castration-Resistant Prostate Cancer
Previous Article in Journal
Prognostic Value of Myosteatosis and Albumin–Bilirubin Grade for Survival in Hepatocellular Carcinoma Post Chemoembolization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Robustness Assessment of Oncology Dose-Finding Trials Using the Modified Fragility Index

1
Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceuticals R&D, AstraZeneca, Durham, NC 27703, USA
2
Biostatistics and Research Decision Sciences, Merck & Co. Inc., Rahway, NJ 07065, USA
3
Division of Biometrics IV, OB/OTS/CDER/FDA, Silver Spring, MD 20993, USA
4
Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, The University of Arizona, Tucson, AZ 85721, USA
5
Cardiovascular, Renal and Metabolism (CVRM), Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
6
Statistical Research and Data Science Center, Pfizer Inc., New York, NY 10001, USA
7
Division of Biostatistics and Health Data Science, The University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(20), 3504; https://doi.org/10.3390/cancers16203504
Submission received: 11 September 2024 / Revised: 9 October 2024 / Accepted: 12 October 2024 / Published: 17 October 2024
(This article belongs to the Special Issue Application of Biostatistics in Cancer Research)

Abstract

:

Simple Summary

In this article, the authors introduce a new metric called the modified Fragility Index (mFI) to assess the accuracy of determining the maximum tolerated dose (MTD) in early oncology clinical trials. The mFI measures how sensitive the MTD decision is to the inclusion of a few more participants in the trial. The authors analyzed three published cancer trials and found that two trials were robust to adding more participants, indicating that the MTD estimate remained stable. However, in the other trial, the MTD estimate was more fragile and could have changed with just one or two more participants. The mFI metric helps researchers make more reliable decisions about the appropriate MTD. By considering the potential impact of additional participants, researchers can improve accuracy and confidence in dose determination, leading to better treatment outcomes for patients.

Abstract

Objectives: The sample sizes of phase I trials are typically small; some designs may lead to inaccurate estimation of the maximum tolerated dose (MTD). The objective of this study was to propose a metric assessing whether the MTD decision is sensitive to enrolling a few additional subjects in a phase I dose-finding trial. Methods: Numerous model-based and model-assisted designs have been proposed to improve the efficiency and accuracy of finding the MTD. The Fragility Index (FI) is a widely used metric quantifying the statistical robustness of randomized controlled trials by estimating the number of events needed to change a statistically significant result to non-significant (or vice versa). We propose a modified Fragility Index (mFI), defined as the minimum number of additional participants required to potentially change the estimated MTD, to supplement existing designs identifying fragile phase I trial results. Findings: Three oncology trials were used to illustrate how to evaluate the fragility of phase I trials using mFI. The results showed that two of the trials were not sensitive to additional subjects’ participation while the third trial was quite fragile to one or two additional subjects. Conclusions: The mFI can be a useful metric assessing the fragility of phase I trials and facilitating robust identification of MTD.

1. Introduction

The concept of fragility index (FI) dates back to the 1990s [1,2] as an additional robustness appraisal of “statistical significance” for assessing a difference between two proportions. The FI is defined as the minimum number of participants in a randomized clinical trial that is required to change a statistically significant result to non-significant (or vice versa). Walsh et al. used FI to assess the robustness of statistical significance of 399 randomized trials with binary outcomes in 2014 [3], and found that in 53% of trials, the FI was less than the number of patients lost to follow-up, suggesting that the trials were frequently fragile. The FI complements the hypothesis testing (e.g., p-value) and helps to identify less robust (or fragile) trial results. Methods for calculating the FI have been further developed for randomized clinical trials with continuous and survival outcomes [4,5], meta-analyses, and network meta-analyses [6,7,8]. The FI has been increasingly applied to many medical fields, including oncology, surgery, obstetrics, and gynecology, during the past decade [9,10,11,12].
In phase I oncology clinical trials, one of the primary goals is to identify the maximum tolerated dose (MTD), which will be used to guide the recommended dose for later phases. However, there may be concerns about the robustness of the chosen MTD, usually determined based on the data from a small number of participants. Researchers and drug developers are often interested in whether the MTD would change if a few additional patients were added to the dose-finding process. If the MTD decision would not be altered in either direction (i.e., downgrading or upgrading) after adding multiple new subjects, we would have more confidence in the dose level chosen as the MTD. Conversely, if the chosen MTD level would be changed right away after including one additional subject, it would raise substantial concerns about whether the selected MTD was reliable. Thus, we propose a modified Fragility Index (mFI), defined as the minimum number of additional participants that is required to potentially change the estimated MTD, to assess robustness and identify fragile phase I trial results.
We begin with an overview on early phase trials and various dose-finding designs in Section 2. Then we cover the definition of mFI and explain how to estimate mFI in Section 3. Three real oncology trials are used to illustrate how to utilize mFI as a convenient and valuable tool for assessing the robustness and validity of the MTD determination in Section 4. Lastly, we examine the relationship between mFI and early stopping, and discuss the limitations of mFI and potential future research topics.

2. Materials and Methods

2.1. Dose Finding Designs

A phase I trial is often the first time a new drug is applied in human beings. One of the primary goals is to examine the highest possible dose level subject to the dose-limiting toxicity (DLT) constraints and identify the MTD for later phases. Assuming monotonicity, the target DLT probability is often set at a value between 20% and 40%.
The traditional approaches to selecting the MTD include the “3+3” design [13] and various up-and-down designs [14]. The “3 +3” design is the most used for phase I dose escalation. The implementation is easy and does not require a computer program. The sample size required is often smaller than for the model-based designs. However, it is generally inferior in identifying the MTD [15].
Many novel model-based and model-assisted designs have been proposed to improve the efficiency and accuracy of phase I trials to find the MTD. Model-based approaches include the continual reassessment method (CRM) [16], escalation with overdose controls (EWOC) [17], the Bayesian logistic regression model (BLRM), and the time-to-event CRM (TITE-CRM) [18]. Model-assisted designs [19] include the modified toxicity probability interval (mTPI) [15,20], keyboard design [21], and Bayesian optimal interval (BOIN) [22]. Researchers have compared the differences and summarized relative pros and cons for some designs [23]. Appendix A presents an overview of commonly used designs in more detail.

2.2. Fragility Index

The FI was developed by Walsh et al. [3] for two-arm randomized controlled trials with binary outcomes that report the numbers of events and non-events in a 2 × 2 frequency table. The aim was to examine whether the statistically significant result of a two-arm trial would be altered with a small change in the number of events. Walsh et al. [3] proposed calculating the FI by changing the event status of a subject in a group with fewer events, re-computing the p-value based on the Fisher exact test, and repeating this process until a non-significant p-value was reached.
For oncology dose-finding studies, because regulators are often interested in whether the MTD would change if a few additional patients were added to the dose-finding process, we propose a modified Fragility Index (mFI), defined as the minimum number of additional participants required to potentially change the estimated MTD, to assess the robustness and identify fragile trial results. The MTD can be altered in either direction: downgrading to a lower dose level or upgrading to a higher dose level. The aim is to investigate if the MTD decision is sensitive to enrolling a few additional subjects in a phase I dose-finding trial.
Suppose that after a dose-finding trial, we collect the following data: d = d 1 , d 2 , d J representing the dosage, n = ( n 1 , n 2 , n J ) the total number of subjects assigned to each dose level, and y = ( y 1 , y 2 , y J ) the observed DLT for each dose level. If t more subjects are included for further investigation, the possible number of DLTs could be any value in the list { 0 ,   1 ,   2 ,   ,   t } . We could iterate through the list to see whether the originally chosen MTD in the trial would be overturned. As long as one value in the list { 0 ,   1 ,   2 ,   ,   t } changes the MTD decision, the mFI is set to be t and we stop the process. If none of the values in the list { 0 ,   1 ,   2 ,   ,   t } changes the MTD decision, we include one more subject and repeat the same process for the t + 1 subjects. If any DLT value in the list { 0 ,   1 ,   2 ,   ,   t + 1 } changes the MTD decision, we stop the process and conclude that m F I = t + 1 . To be consistent, it is recommended to use the same dose-finding design as employed in the original trial. However, other dose-finding designs and models can be implemented as supplementary assessments. Figure 1 illustrates the process of obtaining the mFI for a dose-finding trial. The procedure is summarized as follows:
  • Collect data from a completed dose-finding trial, d = ( d 1 , d 2 , d J ) , n = ( n 1 , n 2 , n J ) , y = ( y 1 , y 2 , y J ) . At the dose level of the MTD ( d M T D ), the number of subjects is n M T D and the number of DLTs is y M T D .
  • Start with t = 1 , i.e., add one additional subject at MTD so that the total number of subjects at the MTD is n M T D + 1 . Let the DLT outcome at the MTD for this new subject be either 0 or 1, hence the total number of DLTs is either y M T D or y M T D + 1 . Use the same statistical method as used in the original study for both numbers of DLTs to see whether the resulting new MTD is different from the original MTD. If it is different, set mFI = 1; otherwise, go to the next step.
  • Let t = t + 1 . The number of subjects at the MTD is n M T D + t and let the number of DLTs at the MTD take any value between 0 and t : y M T D , y M T D + 1 , ,   y M T D + t . Use the same statistical method as used in the original study for all DLT outcomes to assess whether the resulting MTD is different. If it is different, set mFI = t ; otherwise, go to the next step.
  • Repeat step 3 unless MTD has been changed and mFI has been set to a value, or if it reaches a prespecified large value.
  • Once the mFI value is determined, we can calculate the probability of observing the number of DLTs or a more extreme case that would change the MTD decision based on the estimated toxicity probability at the original MTD level, to assess its likelihood. For example, if t patients are added at the original MTD level and if m or fewer DLTs are observed among those new patients, it will change the MTD; the probability of this happening is:
    P r X m = i = 0 m P r X = i = i = 0 m n M T D + t m p M T D m ( 1 p M T D ) n M T D + t m
    where X ~ B i n ( p M T D ,   n M T D + t ) and p M T D can be estimated using the observed DLT rate at the MTD level.
We have developed a publicly available R package to provide a convenient way to implement the mFI calculation.

3. Results: Three Case Studies

3.1. Phase I Dose-Escalation Trial of AUY922

A first-in-human phase I trial was conducted in patients with advanced solid tumors to determine the MTD of AUY922 inhibitor [24]. An adaptive Bayesian logistic regression model (BLRM) with overdose control was used to assess the relation between dose and DLT probability. The dose started at 2 mg/m2 and upgraded to 4, 8, and 16 mg/m2 with no DLTs. At the next higher dose level of 22 mg/m2, one DLT was observed among 11 patients. No DLT was seen at 28 mg/m2, so the dose was upgraded to 40 mg/m2, at which two of the first seven patients experienced DLTs. The BLRM design supported continued dosing at 40 mg/m2 on the basis of the assessment that the probability of a true DLT probability above 33% was less than 0.25. Therefore, nine additional patients were then dosed and no DLT was observed among these patients. The dose was further extended to 54 and 70 mg/m2, where 2 out of 18 at 54 mg/m2 and 3 out of 24 at 70 mg/m2 had DLT. The final recommended phase II dose (RP2D) was declared at 70 mg/m2. Table 1 displays the dose-finding data of the dose levels, total numbers of subjects treated, numbers of DLTs, and DLT rates for all dose levels.
The mFI was found to be 10, based on both the BLRM and BOIN designs: the MTD did not change until 10 extra subjects were added to the trial at the MTD level of 70 mg/m2 and all those 10 subjects experienced DLT. If we tried fewer subjects, the MTD would not be changed no matter how many subjects experienced DLT. This large mFI value suggests that the result for MTD in this trial was quite robust. One can estimate the probability of having a possible number of DLTs, based on the estimated toxicity probability at the MTD dose level, if 10 more subjects were to be recruited onto the trial. Using a binomial distribution, the chance of having 10 DLTs out of 10 additional subjects is very low, 0.125 10 < 10 8 , so it is very unlikely that the MTD result would be altered.
The mFI value was the same with the keyboard design, because both BOIN and keyboard are model-assisted designs and use the same isotonic algorithm to compute the MTD. Other dose-finding designs, such as mTPI, gave the same mFI value of 10, whereas the mFI value was 12 when using the CRM algorithm and the mFI value was 8 when using the EWOC algorithm (Table 2).

3.2. Phase I Trial of Pan-AKT Inhibitor MK-2206

This was a dose-escalation study of continuous oral treatment with the pan-AKT inhibitor MK-2206 in patients with advanced tumors [25]. The drug was administered every other day in 28-day cycles to investigate the safety and MTD. In total, 33 patients were dosed at 30, 60, 75, or 90 mg. The dose finding used a two-stage design. In Stage 1, dose escalation proceeded through dose levels of 30 mg (three subjects), 60 mg (three subjects), and 90 mg (seven subjects). There were 4 out of 7 patients who experienced DLTs at 90 mg. In Stage 2, a new dose of 75 mg was introduced for three patients, whereupon all three developed DLTs. An additional three patients were then enrolled at the lower dose level of 60 mg to check the safety parameters of this dose, and no DLTs were found. Stage 2 included 14 more patients in the expansion phase and observed one DLT. The MTD was established at 60 mg with the mTPI design. The dose-finding data for dose levels, total numbers of subjects treated, numbers of DLTs, and DLT rates are displayed in Table 3.
As shown in Table 3, the 100% observed DLT rate at the dose level of 75 mg makes it impossible to upgrade from 60 mg to 75 mg. When additional subjects are enrolled, the only possible outcome of changing the MTD is to downgrade. The mFI is 18 according to the mTPI design algorithm. The high mFI value is caused by the 100% DLT rate at the next dose level. The MTD level does not change until 18 extra subjects are added in the trial at the MTD level of 60 mg/m2 and all those subjects experience at least one DLT, the probability of which is extremely low. This suggests that the result for MTD is robust.
As summarized in Table 2, if the keyboard or BOIN design is employed, the mFI is 11, which is still large. Other dose-finding designs give similar mFI values around 10: mFI is 10 with CRM and mFI is 9 with BLRM. However, the mFI is only 5 when using the EWOC algorithm, which may be due to EWOC’s over-conservative safety control rule.

3.3. The SPRINT Phase I Trial

SPRINT was an open-label, single arm, multi-center trial of the MEK 1 inhibitor, selumetinib, in children [26]. The phase I portion of the SPRINT trial evaluated three doses of selumetinib, 20 mg/m2, 25 mg/m2, and 30 mg/m2 in pediatric patients, to identify a suitable dose to be used for the next phase based on all available safety, tolerability, pharmacokinetic, and efficacy data. The objective response rate was similar across 20 to 30 mg/m2 doses: 66.7% (8/12) at 20 mg/m2, 83.3% (5/6) at 25 mg/m2, 50.0% (3/6) at 30 mg/ m2 respectively. The best rate was observed at 25 mg/m2. The tolerability was similar between 20 and 25 mg/m2 doses based on the DLT rates: 2 DLTs out of 12 subjects at 20 mg/m2, 1 DLT out of 6 subjects at 25 mg/m2, and 2 DLTs out of 6 subjects at 30 mg/m2, as shown in Table 4. The dose of 25 mg/m2 was identified as the MTD and the recommended dose for phase II. We used only the tolerability data listed in Table 4 to assess the robustness of the MTD result.
The mFI was found to be 1 with the BOIN algorithm. When one patient was added with no DLT for this new patient, it led to an upgrade to a higher dose level; and the probability of this happening is 0.833, using the observed DLT rate of 16.7%. On the other hand, we investigated downgrading: first, if only one patient is added and that patient develops DLT, the MTD remains the same; then, if two patients are tested additionally and both two patients develop DLT, it results in downgrading to a lower dose level. The probability of these two patients experiencing DLT is 0.028. The mFI would be the 1, since this is the smallest number of subjects needed to alter the MTD. The small mFI indicates the trial’s MTD conclusion is not robust if we consider only tolerability data. Other dose-finding designs, such as CRM, EWOC, BLRM, and mTPI, give similar mFI values of either 1 or 2 (Table 2).

3.4. mFI Results Summary

The mFI results are summarized in Table 2 for all three trials using various dose-finding designs, CRM, EWOC, BLRM, mTPI, keyboard, and BOIN. To be consistent with the original dose-finding process, it is recommended to use the same dose-finding design as employed in the trial for calculating mFI. Other dose-finding models can be implemented as supplementary assessments. There are some variations in the mFI results, as demonstrated by the three trials, but there should be a general pattern or signal in terms of robustness assessment.
The results showed that the first two trials were not sensitive to additional participants, while the third trial is quite fragile to one or two additional subjects being added.

4. Discussion and Conclusions

We propose a modified Fragility Index (mFI) to assess the robustness of the MTD determination in early-phase dose-finding trials. The extension of FI in dose-finding trials allows researchers and drug developers to assess whether any additional patients and how many patients should be recruited to achieve a robust MTD decision. Three oncology trials were used to show how to calculate the mFI and assess trial robustness.
In practice, different trials may employ different designs. To be consistent, it is recommended to use the same dose-finding design as employed in the trial for the estimation of mFI. However, other dose-finding designs and models can be implemented as supplementary assessments. Because phase I trials commonly involve a small number of subjects, an mFI value greater than 3 or 5 may intuitively be considered as an indication of robust MTD decision. However, to establish a useful guideline, one would need to systematically evaluate all existing phase I oncology trials and empirically estimate the distribution of mFI. Furthermore, the mFI evaluation may depend on the design used to estimate the MTD. The relative performance of different designs on robust assessment using mFI awaits further research.
Phase I trials can implement rules to stop early if the clinical objectives have been achieved with good confidence. Various stopping rules have been suggested in the literature. O’Quigley et al. [16] proposed a stopping rule based on a confidence interval for CRM and other model-based methods, and Shen and O’Quigley gave a theoretical justification [27]. However, there are some limitations to this approach: (1) the precision level may often require more patients than would be available in an early dose-finding trial and hence, the trial would not halt early in practice; (2) it is questionable whether obtaining a pre-fixed level of precision for the probability of toxicity at the MTD should be a major goal of a trial [28]. On the other hand, O’Quigley and Reiner (1998) [29] estimated the probability that a current recommended dose level will turn out to be the final MTD and the likelihood that all remaining patients will be treated at the current dose level. The trial would be terminated early if the MTD could be predicted with high probability. To implement this stopping rule, one can keep track of the number of times each dose is considered and stop when the dose for an upcoming patient is the same as the dose that was recommended for the previous k (a pre-decided integer) patients in a row. This approach often works out well in practice [28].
Implementing early stopping during a dose-finding trial may first seem quite different from assessing the robustness of a trial using FI, because one is used during a dose-finding trial and the other is considered afterwards. However, they are very much related, because mFI can also be applied in real time during a trial. If a trial has already included a certain number of patients, then what would happen if a few more patients were to be included? Would adding more patients change the current recommended dose? Or would the current recommended dose stay the same, and what probability is associated with that? Therefore, we may want to compare these two approaches via simulations and in practice. As we try to achieve Project Optimus, selecting an optimal biological dose is no longer based just on toxicity, but also other factors, such as efficacy and pharmacokinetic results. Our proposed mFI index can be extended naturally to consider other determining factors beyond safety by incorporating those factors in the decision rule, as shown in the third example.
A related fragility measure is the Robustness Index (RI) proposed by Heston in 2023 [30], which examines how different sample sizes affect statistical significance. When a hypothesis test yields a non-significant result, the original sample size is multiplied by a series of numbers until a significant result is achieved. Conversely, when a test is significant, the original sample size is divided by a series of numbers until the result is no longer significant. The multiplicand or divisor is the RI. However, it is uncertain how the RI can be utilized in a dose-finding trial to determine when the MTD result will change.
The strength of this study is that, to the best of our knowledge, there are scant, if any, existing methods dedicated specifically to evaluating the robustness of results from dose-finding studies. By extending the concept of the FI, this work offers an intuitive way to quantify how the significance of the dose-finding conclusion could change after including additional potential samples. Nevertheless, this study has some limitations. Although our proposed mFI shares the same spirit as the original FI developed by Walsh et al., as both aim at altering the result’s significance, they differ in terms of how they achieve the significance change. Specifically, the original FI was intended for general hypothesis testing in relation to treatment effects with binary outcomes in clinical trials, and it modifies the status of binary outcomes (event or no event), with the size of samples in each group remaining fixed. In contrast, in our proposed mFI, like several other extensions of FI for continuous outcomes and survival outcomes [31], the sample sizes in treatment groups or the study are modified. As such, one may argue that the mFI is not a type of FI measure, and we may consider this as an FI-like measure. In addition, our proposed mFI does not account for the likelihood of DLT outcomes among the assumed additional samples for the mFI calculation. It is possible that the DLT outcomes of some samples may not be clinically practical. Baer et al. proposed generalizing the fragility index to a family of fragility indices called incidence fragility indices, permitting only outcome modifications that are sufficiently likely and providing an exact algorithm to calculate the incidence fragility indices [32]. Such a consideration may also be needed when interpreting the mFI results.

Author Contributions

Conceptualization, A.X.S. and H.C.; Methodology, A.X.S., H.Z., L.N., L.L., H.L. and H.C.; Software, A.X.S. and H.Z.; Validation, A.X.S., H.Z. and H.C.; Formal analysis, A.X.S.; Data curation, A.X.S. and L.N.; Writing—original draft preparation, A.X.S.; writing—review and editing, A.X.S., H.Z., L.N., L.L., H.L. and H.C.; Visualization, A.X.S.; Supervision, H.C.; Project administration, H.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The summary level data that support the findings of this study are included in the paper.

Disclaimer

This article reflects the views of the authors and should not be construed to represent FDA’s views or policies.

Conflicts of Interest

The authors declare no conflicts of interest. A.X.S., H.L., H.Z., and H.C. are employed by AstraZeneca, Merck, and Pfizer, and own stocks in those companies. However, all of the contents in this article are strictly educational, instructive, and methodological.

Appendix A. Overview of Commonly Used Dose Finding Designs

Here, we provide a brief overview of commonly used trial designs. Suppose there are in total J dose levels, d = ( d 1 , d 2 , d J ) , in a phase I dose-finding trial. For dose j (j = 1, 2, …, J), the number of patients with a DLT ( y j ) in a cohort of size n j is assumed to be binomially distributed y j ~ B i n ( p j ,   n j ) with the DLT probabilities ( p j ).

A.1. The Continual Reassessment Method (CRM)

In the CRM design, a parametric model for the dose–toxicity curve is assumed as p j = a j e x p ( α ) , where p j is the true DLT probability at dose level j , α is an unknown parameter, and a j is the initial guess of the DLT probability at dose level j with the constraint of 0 < a 1 < a 2 < < a J < 1 . The CRM updates the dose–toxicity curve with the accumulating DLT data across all dose levels and assigns new patients to the optimal dose level, which is defined as the one with the estimated DLT probability being the closest to the target DLT probability, ϕ . In addition, a safety stopping rule is implemented so that the process is stopped if the DLT probability at the lowest dose ( p 1 ) is greater than the target above a pre-specified threshold (for example,   0.95 ), i.e., P r p 1 > ϕ D > 0.95 , where D denotes the trial data.

A.2. The Escalation with Overdose Control (EWOC)

As a modification of the CRM, the EWOC employs a two-parameter logistic regression model for extra flexibility in modeling the dose–toxicity curve as logit ( p j ) = β 0 + β 1 d j , where ( β 0 ,   β 1 ) are the unknown intercept and slope parameters, and d j is the dosage of dose level j . Thus, the DLT probability of the first dose d 1 is p 1 = e x p ( β 0 + β 1 d 1 ) 1 + e x p ( β 0 + β 1 d 1 ) , and the MTD is λ = 1 β 1 [ log ϕ log 1 ϕ β 0 ] . The EWOC treats the first group of patients at the lowest dose and updates the dose–toxicity curve with the accumulating DLT data. The next group of patients are assigned to the optimal dose, which is defined as the one whose mean estimate of the DLT probability is the closest to the target DLT probability ϕ . The same safety stopping rule is used, i.e., P r p 1 > ϕ D > 0.95 .

A.3. The Bayesian Logistic Regression Model (BLRM)

This is a two-parameter logistic model, logit ( p j ) = l o g ( α ) + β l o g ( d j d * ) , where α , β are the two unknown parameters and d * is the reference dose. Following the paper by Neuenschwander et al. [33], a flat bivariate normal distribution is often used as the prior for log α , log β . The BLRM requires a predefined probability interval ( δ 1 , δ 2 ) as the range of acceptable DLT probabilities. For example, ( δ 1 , δ 2 ) = ( ϕ 0.05 ,   ϕ + 0.05 ) . The BLRM imposes an overdose control rule, as follows: a dose is considered overdosing if the observed data indicate that the DLT probability at a dose level greater than the upper bound is higher than 0.25 , i.e., P r p j > δ 2 D > 0.25 . The same safety stopping rule as the previous two designs is often used in the BLRM.

A.4. The Modified Toxicity Probability Interval (mTPI) Design

The mTPI design uses a beta-binomial model at each dose level: y j | n j , p j ~ B i n n j , p j and p j ~ B e t a 1,1 . Thus, the posterior distribution of the DLT probability at dose j is a beta distribution, i.e., p j | n j , y j ~ B e t a y j + 1 , n j y j + 1 . Given a target toxicity probability ϕ , the mTPI design prespecifies three intervals with two parameters δ 1 = ϕ ε 1 , δ 2 = ϕ + ε 2 , that is, the underdosing interval 0 , δ 1 , the acceptable dosing interval δ 1 , δ 2 , and the overdosing interval ( δ 2 , 1 ) , where 0 < δ 1 < ϕ < δ 2 < 1 . Then, the mTPI design defines a quantity named unit probability mass (UPM), given the posterior distribution of p j for each of the three intervals as follows:
U P M 1 = P r p j < δ 1 | n j , y j / δ 1 ,
U P M 2 = P r δ 1 p j δ 2 | n j , y j / ( δ 2 δ 1 ) ,
U P M 3 = P r p j > δ 2 | n j , y j / ( 1 δ 2 ) .
That is, the UPM is the posterior probability that p j lies in the corresponding interval divided by the length of that interval. The mTPI design determines dose escalation/de-escalation based only on the observed data at the current dose level j as follows:
  • If U P M 1 = m a x { U P M 1 , U P M 2 , U P M 3 } , escalate dose to level j + 1 ;
  • If U P M 2 = m a x { U P M 1 , U P M 2 , U P M 3 } , stay at the current dose level j ;
  • If U P M 3 = m a x { U P M 1 , U P M 2 , U P M 3 } , de-escalate dose to level j 1 .
Because the three UPMs can be calculated for all outcomes of n j and y j , dose escalation and de-escalation rules can be determined before the onset of the trial. To avoid treating excessive numbers of participants at extremely toxic dose levels, the mTPI design implements a dose-exclusion rule as follows: if P r p j > ϕ | n j , y j > 0.95 , dose level j and higher doses are excluded in the trial. If the lowest dose is excluded, the trial is stopped for safety.

A.5. The Keyboard Design

Yan et al. [21] proposed the keyboard design to improve the performance of the mTPI design, noting that the latter has a high risk of overdosing patients due to the use of the UPM to guide dose escalation. The keyboard design starts by specifying a proper probability interval I * = δ 1 , δ 2 (referred to as the target key) and then forms a series of equal-width keys on both sides of the target key. We denote the resulting intervals/keys as I 1 , ,   I K . To make the decision for dose transition, given the observed data ( n j ,   y j ) at the current dose level j , the keyboard design defines the “strongest key” as I m a x = a r g m a x { Pr p j I k ; k = 1 , ,   K } , which can be easily obtained using the beta-binomial model as with the mTPI. Statistically, the strongest key represents the interval in which the true toxicity probability is most likely to be located. This intuitive interpretation of the strongest key naturally leads to the following keyboard dose escalation and de-escalation rule:
  • If the strongest key is on the left side of the target key, escalate to the level j + 1 ;
  • If the strongest key is the target key, stay at the current level j ;
  • If the strongest key is on the right side of the target key, de-escalate to level j 1 .
The same dose-exclusion rule as in the mTPI design is also implemented in the keyboard design.

A.6. The Bayesian Optimal Interval (BOIN) Design

Compared with the mTPI and keyboard designs, which require calculating the posterior distribution of the DLT probabilities, the BOIN design is more straightforward and transparent. Let p ^ j = y j n j denote the observed DLT probability at the current dose level j , and λ e ,   λ d denote the predetermined dose escalation and de-escalation boundaries. The BOIN design determines the next dose level as follows:
  • If p ^ j λ e , escalate to the level j + 1 ;
  • If p ^ j λ d , de-escalate to the level j 1 ;
  • Otherwise, stay at the current level j .
The BOIN design also implements the same dose-exclusion rule as the mTPI and keyboard designs. Due to its straightforward implementation and excellent performance, the BOIN design received a fit-for-purpose designation from FDA as a tool for dose-finding in oncology [34].

References

  1. Feinstein, A.R. The unit fragility index: An additional appraisal of “statistical significance” for a contrast of two proportions. J. Clin. Epidemiol. 1990, 43, 201–209. [Google Scholar] [CrossRef] [PubMed]
  2. Walter, S. Statistical significance and fragility criteria for assessing a difference of two proportions. J. Clin. Epidemiol. 1991, 44, 1373–1378. [Google Scholar] [CrossRef] [PubMed]
  3. Walsh, M.; Srinathan, S.K.; McAuley, D.F.; Mrkobrada, M.; Levine, O.; Ribic, C.; Molnar, A.O.; Dattani, N.D.; Burke, A.; Guyatt, G. The statistical significance of randomized controlled trial results is frequently fragile: A case for a Fragility Index. J. Clin. Epidemiol. 2014, 67, 622–628. [Google Scholar] [CrossRef] [PubMed]
  4. Baer, B.R.; Fremes, S.E.; Gaudino, M.; Charlson, M.; Wells, M.T. On clinical trial fragility due to patients lost to follow up. BMC Med. Res. Methodol. 2021, 21, 1–11. [Google Scholar] [CrossRef] [PubMed]
  5. Bomze, D.; Asher, N.; Ali, O.H.; Flatz, L.; Azoulay, D.; Markel, G.; Meirson, T. Survival-inferred fragility index of phase 3 clinical trials evaluating immune checkpoint inhibitors. JAMA Netw. Open 2020, 3, e2017675. [Google Scholar] [CrossRef]
  6. Atal, I.; Porcher, R.; Boutron, I.; Ravaud, P. The statistical significance of meta-analyses is frequently fragile: Definition of a fragility index for meta-analyses. J. Clin. Epidemiol. 2019, 111, 32–40. [Google Scholar] [CrossRef]
  7. Lin, L. Factors that impact fragility index and their visualizations. J. Eval. Clin. Pract. 2021, 27, 356–364. [Google Scholar] [CrossRef]
  8. Lin, L.; Chu, H. Assessing and visualizing fragility of clinical results with binary outcomes in R using the fragility package. PLoS ONE 2022, 17, e0268754. [Google Scholar] [CrossRef]
  9. Lin, L.; Xing, A.; Chu, H.; Murad, M.H.; Xu, C.; Baer, B.R.; Wells, M.T.; Sanchez-Ramos, L. Assessing the robustness of results from clinical trials and meta-analyses with the fragility index. Am. J. Obstet. Gynecol. 2023, 228, 276–282. [Google Scholar] [CrossRef]
  10. Del Paggio, J.C.; Tannock, I.F. The fragility of phase 3 trials supporting FDA-approved anticancer medicines: A retrospective analysis. Lancet Oncol. 2019, 20, 1065–1069. [Google Scholar] [CrossRef]
  11. Sanchez-Ramos, L.; Lin, L. Cerclage placement in twin pregnancies with short or dilated cervix does not prevent preterm birth: A fragility index assessment. Am. J. Obstet. Gynecol. 2022, 227, 338–339. [Google Scholar] [CrossRef] [PubMed]
  12. Tignanelli, C.J.; Napolitano, L.M. The fragility index in randomized clinical trials as a means of optimizing patient care. JAMA Surg. 2019, 154, 74–79. [Google Scholar] [CrossRef] [PubMed]
  13. Storer, B.E. An evaluation of phase I clinical trial designs in the continuous dose–response setting. Stat. Med. 2001, 20, 2399–2408. [Google Scholar] [CrossRef] [PubMed]
  14. Gezmu, M.; Flournoy, N. Group up-and-down designs for dose-finding. J. Stat. Plan. Inference 2006, 136, 1749–1764. [Google Scholar] [CrossRef]
  15. Ji, Y.; Wang, S.-J. Modified toxicity probability interval design: A safer and more reliable method than the 3 + 3 design for practical phase I trials. J. Clin. Oncol. 2013, 31, 1785. [Google Scholar] [CrossRef]
  16. O’Quigley, J.; Pepe, M.; Fisher, L. Continual reassessment method: A practical design for phase 1 clinical trials in cancer. Biometrics 1990, 46, 33–48. [Google Scholar] [CrossRef]
  17. Babb, J.; Rogatko, A.; Zacks, S. Cancer phase I clinical trials: Efficient dose escalation with overdose control. Stat. Med. 1998, 17, 1103–1120. [Google Scholar] [CrossRef]
  18. Cheung, Y.K.; Chappell, R. Sequential designs for phase I clinical trials with late-onset toxicities. Biometrics 2000, 56, 1177–1182. [Google Scholar] [CrossRef]
  19. Yuan, Y.; Lee, J.J.; Hilsenbeck, S.G. Model-assisted designs for early-phase clinical trials: Simplicity meets superiority. JCO Precis. Oncol. 2019, 3, 1–12. [Google Scholar] [CrossRef]
  20. Guo, W.; Wang, S.-J.; Yang, S.; Lynn, H.; Ji, Y. A Bayesian interval dose-finding design addressingOckham’s razor: mTPI-2. Contemp. Clin. Trials 2017, 58, 23–33. [Google Scholar] [CrossRef]
  21. Yan, F.; Mandrekar, S.J.; Yuan, Y. Keyboard: A novel Bayesian toxicity probability interval design for phase I clinical trials. Clin. Cancer Res. 2017, 23, 3994–4003. [Google Scholar] [CrossRef] [PubMed]
  22. Liu, S.; Yuan, Y. Bayesian optimal interval designs for phase I clinical trials. J. R. Stat. Soc. Ser. C Appl. Stat. 2015, 64, 507–523. [Google Scholar] [CrossRef]
  23. Zhou, H.; Yuan, Y.; Nie, L. Accuracy, safety, and reliability of novel phase I trial designs. Clin. Cancer Res. 2018, 24, 4357–4364. [Google Scholar] [CrossRef] [PubMed]
  24. Sessa, C.; Shapiro, G.I.; Bhalla, K.N.; Britten, C.; Jacks, K.S.; Mita, M.; Papadimitrakopoulou, V.; Pluard, T.; Samuel, T.A.; Akimov, M. First-in-human phase I dose-escalation study of the HSP90 inhibitor AUY922 in patients with advanced solid tumors. Clin. Cancer Res. 2013, 19, 3671–3680. [Google Scholar] [CrossRef]
  25. Yap, T.A.; Yan, L.; Patnaik, A.; Fearen, I.; Olmos, D.; Papadopoulos, K.; Baird, R.D.; Delgado, L.; Taylor, A.; Lupinacci, L. First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors. J. Clin. Oncol. 2011, 29, 4688–4695. [Google Scholar] [CrossRef]
  26. Food and Drug Administration. NDA Multi-Disciplinary Review and Evaluation NDA 213756 for Koselugo (Selumetinib); Food and Drug Administration: Silver Spring, MD, USA, 2020. [Google Scholar]
  27. Shen, L.Z.; O’quigley, J. Consistency of continual reassessment method under model misspecification. Biometrika 1996, 83, 395–405. [Google Scholar] [CrossRef]
  28. Devlin, S.M.; Iasonos, A.; O’Quigley, J. Stopping rules for phase I clinical trials with dose expansion cohorts. Stat. Methods Med. Res. 2022, 31, 334–347. [Google Scholar] [CrossRef]
  29. O’quigley, J.; Reiner, E. A stopping rule for the continual reassessment method. Biometrika 1998, 85, 741–748. [Google Scholar] [CrossRef]
  30. Heston, T.F. The robustness index: Going beyond statistical significance by quantifying fragility. Cureus 2023, 15, e44397. [Google Scholar] [CrossRef]
  31. Caldwell, J.-M.E.; Youssefzadeh, K.; Limpisvasti, O. A method for calculating the fragility index of continuous outcomes. J. Clin. Epidemiol. 2021, 136, 20–25. [Google Scholar] [CrossRef]
  32. Baer, B.R.; Gaudino, M.; Charlson, M.; Fremes, S.E.; Wells, M.T. Fragility indices for only sufficiently likely modifications. Proc. Natl. Acad. Sci. USA 2021, 118, e2105254118. [Google Scholar] [CrossRef] [PubMed]
  33. Neuenschwander, B.; Branson, M.; Gsponer, T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat. Med. 2008, 27, 2420–2439. [Google Scholar] [CrossRef] [PubMed]
  34. Food and Drug Administration. Drug Administration. Drug Development Tools: Fit-for-Purpose Initiative. In Guidance for Industry; FDA (Food and Drug Administration): Silver Spring, MD, USA, 2021. [Google Scholar]
Figure 1. Flowchart of Calculating the modified Fragility Index in a Dose-Finding Trial.
Figure 1. Flowchart of Calculating the modified Fragility Index in a Dose-Finding Trial.
Cancers 16 03504 g001
Table 1. Summary of the Data in the Phase I AUY922 Dose-Escalation Trial.
Table 1. Summary of the Data in the Phase I AUY922 Dose-Escalation Trial.
Dose Level Index
123456789
Dose level (mg/m2)248162228405470
Total # subjects treated3346118161824
# DLT000010223
DLT rate (%)00009.1012.511.112.5
Table 2. The mFI Results of Robustness Assessment for All Three Trials.
Table 2. The mFI Results of Robustness Assessment for All Three Trials.
TrialsDose-Finding Designs
CRMEWOCBLRMmTPIKeyboardBOIN
1. AUY922 Dose Escalation12810101010
2. Pan-AKT Inhibitor MK-2206 1059181111
3. SPRINT Trial222111
Table 3. Summary of the Data in the Phase I Pan-AKT Inhibitor MK-2206 Trial.
Table 3. Summary of the Data in the Phase I Pan-AKT Inhibitor MK-2206 Trial.
Dose Level Index
1234
Dose level (mg)30607590
Total # subjects treated32037
# DLT0134
DLT rate (%)05.010057.1
Table 4. Summary of the Data in the Phase I SPRINT Trial.
Table 4. Summary of the Data in the Phase I SPRINT Trial.
Dose Level Index
123
Dose level (mg/m2)202530
Total # subjects treated1266
# DLT212
DLT rate (%)16.716.733
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shi, A.X.; Zhou, H.; Nie, L.; Lin, L.; Li, H.; Chu, H. Robustness Assessment of Oncology Dose-Finding Trials Using the Modified Fragility Index. Cancers 2024, 16, 3504. https://doi.org/10.3390/cancers16203504

AMA Style

Shi AX, Zhou H, Nie L, Lin L, Li H, Chu H. Robustness Assessment of Oncology Dose-Finding Trials Using the Modified Fragility Index. Cancers. 2024; 16(20):3504. https://doi.org/10.3390/cancers16203504

Chicago/Turabian Style

Shi, Amy X., Heng Zhou, Lei Nie, Lifeng Lin, Hongjian Li, and Haitao Chu. 2024. "Robustness Assessment of Oncology Dose-Finding Trials Using the Modified Fragility Index" Cancers 16, no. 20: 3504. https://doi.org/10.3390/cancers16203504

APA Style

Shi, A. X., Zhou, H., Nie, L., Lin, L., Li, H., & Chu, H. (2024). Robustness Assessment of Oncology Dose-Finding Trials Using the Modified Fragility Index. Cancers, 16(20), 3504. https://doi.org/10.3390/cancers16203504

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop