1
|
Fang J, Yin G. Fractional accumulative calibration-free odds (f-aCFO) design for delayed toxicity in phase I clinical trials. Stat Med 2024; 43:3210-3226. [PMID: 38816959 DOI: 10.1002/sim.10127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024]
Abstract
The calibration-free odds (CFO) design has been demonstrated to be robust, model-free, and practically useful but faces challenges when dealing with late-onset toxicity. The emergence of the time-to-event (TITE) method and fractional method leads to the development of TITE-CFO and fractional CFO (fCFO) designs to accumulate delayed toxicity. Nevertheless, existing CFO-type designs have untapped potential because they primarily consider dose information from the current position and its two neighboring positions. To incorporate information from all doses, we propose the accumulative CFO (aCFO) design by utilizing data at all dose levels similar to a tug-of-war game where players distant from the center also contribute their strength. This approach enhances full information utilization while still preserving the model-free and calibration-free characteristics. Extensive simulation studies demonstrate performance improvement over the original CFO design, emphasizing the advantages of incorporating information from a broader range of dose levels. Furthermore, we propose to incorporate late-onset outcomes into the TITE-aCFO and f-aCFO designs, with f-aCFO displaying superior performance over existing methods in both fixed and random simulation scenarios. In conclusion, the aCFO and f-aCFO designs can be considered robust, efficient, and user-friendly approaches for conducting phase I trials without or with late-onsite toxicity.
Collapse
Affiliation(s)
- Jialu Fang
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, China
| | - Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
Biard L, Andrillon A, Silva RB, Lee SM. Dose optimization for cancer treatments with considerations for late-onset toxicities. Clin Trials 2024; 21:322-330. [PMID: 38591582 PMCID: PMC11132952 DOI: 10.1177/17407745231221152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Given that novel anticancer therapies have different toxicity profiles and mechanisms of action, it is important to reconsider the current approaches for dose selection. In an effort to move away from considering the maximum tolerated dose as the optimal dose, the Food and Drug Administration Project Optimus points to the need of incorporating long-term toxicity evaluation, given that many of these novel agents lead to late-onset or cumulative toxicities and there are no guidelines on how to handle them. Numerous methods have been proposed to handle late-onset toxicities in dose-finding clinical trials. A summary and comparison of these methods are provided. Moreover, using PI3K inhibitors as a case study, we show how late-onset toxicity can be integrated into the dose-optimization strategy using current available approaches. We illustrate a re-design of this trial to compare the approach to those that only consider early toxicity outcomes and disregard late-onset toxicities. We also provide proposals going forward for dose optimization in early development of novel anticancer agents with considerations for late-onset toxicities.
Collapse
Affiliation(s)
- Lucie Biard
- INSERM U1153 Team ECSTRRA, Université Paris Cité, Paris, France
| | - Anaïs Andrillon
- INSERM U1153 Team ECSTRRA, Université Paris Cité, Paris, France
- Department of Statistical Methodology, Saryga, Tournus, France
| | - Rebecca B Silva
- Columbia University, Mailman School of Public Health, Department of Biostatistics, New York, USA
| | - Shing M Lee
- Columbia University, Mailman School of Public Health, Department of Biostatistics, New York, USA
| |
Collapse
|
3
|
Lee SY. A flexible dose-response modeling framework based on continuous toxicity outcomes in phase I cancer clinical trials. Trials 2023; 24:745. [PMID: 37990281 PMCID: PMC10664620 DOI: 10.1186/s13063-023-07793-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND The past few decades have seen remarkable developments in dose-finding designs for phase I cancer clinical trials. While many of these designs rely on a binary toxicity response, there is an increasing focus on leveraging continuous toxicity responses. A continuous toxicity response pertains to a quantitative measure represented by real numbers. A higher value corresponds not only to an elevated likelihood of side effects for patients but also to an increased probability of treatment efficacy. This relationship between toxicity and dose is often nonlinear, necessitating flexibility in the quest to find an optimal dose. METHODS A flexible, fully Bayesian dose-finding design is proposed to capitalize on continuous toxicity information, operating under the assumption that the true shape of the dose-toxicity curve is nonlinear. RESULTS We conduct simulations of clinical trials across varying scenarios of non-linearity to evaluate the operational characteristics of the proposed design. Additionally, we apply the proposed design to a real-world problem to determine an optimal dose for a molecularly targeted agent. CONCLUSIONS Phase I cancer clinical trials, designed within a fully Bayesian framework with the utilization of continuous toxicity outcomes, offer an alternative approach to finding an optimal dose, providing unique benefits compared to trials designed based on binary toxicity outcomes.
Collapse
Affiliation(s)
- Se Yoon Lee
- Department of Statistics, Texas A &M University, 3143 TAMU, College Station, 77843, TX, USA.
| |
Collapse
|
4
|
Jin H, Yin G. Time-to-event calibration-free odds design: A robust efficient design for phase I trials with late-onset outcomes. Pharm Stat 2023; 22:773-783. [PMID: 37095681 DOI: 10.1002/pst.2304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 04/26/2023]
Abstract
Compared with most of the existing phase I designs, the recently proposed calibration-free odds (CFO) design has been demonstrated to be robust, model-free, and easy to use in practice. However, the original CFO design cannot handle late-onset toxicities, which have been commonly encountered in phase I oncology dose-finding trials with targeted agents or immunotherapies. To account for late-onset outcomes, we extend the CFO design to its time-to-event (TITE) version, which inherits the calibration-free and model-free properties. One salient feature of CFO-type designs is to adopt game theory by competing three doses at a time, including the current dose and the two neighboring doses, while interval-based designs only use the data at the current dose and is thus less efficient. We conduct comprehensive numerical studies for the TITE-CFO design under both fixed and randomly generated scenarios. TITE-CFO shows robust and efficient performances compared with interval-based and model-based counterparts. As a conclusion, the TITE-CFO design provides robust, efficient, and easy-to-use alternatives for phase I trials when the toxicity outcome is late-onset.
Collapse
Affiliation(s)
- Huaqing Jin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Guosheng Yin
- Department of Mathematics, Imperial College London, London, UK
| |
Collapse
|
5
|
Brown SR, Hinsley S, Hall E, Hurt C, Baird RD, Forster M, Scarsbrook AF, Adams RA. A Road Map for Designing Phase I Clinical Trials of Radiotherapy-Novel Agent Combinations. Clin Cancer Res 2022; 28:3639-3651. [PMID: 35552622 PMCID: PMC9433953 DOI: 10.1158/1078-0432.ccr-21-4087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/26/2022] [Accepted: 04/28/2022] [Indexed: 01/07/2023]
Abstract
Radiotherapy has proven efficacy in a wide range of cancers. There is growing interest in evaluating radiotherapy-novel agent combinations and a drive to initiate this earlier in the clinical development of the novel agent, where the scientific rationale and preclinical evidence for a radiotherapy combination approach are high. Optimal design, delivery, and interpretation of studies are essential. In particular, the design of phase I studies to determine safety and dosing is critical to an efficient development strategy. There is significant interest in early-phase research among scientific and clinical communities over recent years, at a time when the scrutiny of the trial methodology has significantly increased. To enhance trial design, optimize safety, and promote efficient trial conduct, this position paper reviews the current phase I trial design landscape. Key design characteristics extracted from 37 methodology papers were used to define a road map and a design selection process for phase I radiotherapy-novel agent trials. Design selection is based on single- or dual-therapy dose escalation, dose-limiting toxicity categorization, maximum tolerated dose determination, subgroup evaluation, software availability, and design performance. Fifteen of the 37 designs were identified as being immediately accessible and relevant to radiotherapy-novel agent phase I trials. Applied examples of using the road map are presented. Developing these studies is intensive, highlighting the need for funding and statistical input early in the trial development to ensure appropriate design and implementation from the outset. The application of this road map will improve the design of phase I radiotherapy-novel agent combination trials, enabling a more efficient development pathway.
Collapse
Affiliation(s)
- Sarah R. Brown
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Samantha Hinsley
- Clinical Trials Unit Glasgow, University of Glasgow, Glasgow, United Kingdom
| | - Emma Hall
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Chris Hurt
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | | | | | - Andrew F. Scarsbrook
- Radiotherapy Research Group, Leeds Institute of Medical Research at St James's, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Richard A. Adams
- Centre for Trials Research, Cardiff University and Velindre Cancer Centre, Cardiff, United Kingdom
| |
Collapse
|
6
|
Lee SY, Munafo A, Girard P, Goteti K. Optimization of dose selection using multiple surrogates of toxicity as a continuous variable in phase I cancer trial. Contemp Clin Trials 2021; 113:106657. [PMID: 34954097 DOI: 10.1016/j.cct.2021.106657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/03/2022]
Abstract
In phase I trials, it is the top priority of clinicians to effectively treat patients and minimize the chance of exposing them to subtherapeutic and overly toxic doses, while exploiting patient information. Motived by this practical consideration, we revive the one parameter linear dose-finder developed in 1970s to accommodate a continuous toxicity response in the phase I cancer clinical trials, which is called the two parameters linear dose-finder (2PLD). The 2PLD is a fully Bayesian model that assumes a linear relationship between toxicity response and dose. We suggest a dose search algorithm based on the 2PLD to exploit the grades of toxicities from multiple adverse events to align with Common Toxicity Criteria for Adverse Events provided by the National Cancer Institute. The proposed search procedure suggests an optimal dose to each patient by using accrued patients' information while controlling the posterior probability of overdose. The heterogeneity of patients in dose reaction is addressed by making a fully Bayesian inference about the standard deviation of toxicity responses. The 2PLD can be an attractive tool for clinical scientists due to its parsimonious description of a toxicity-dose curve and medical interpretation as well as an automatic posterior computation. We illustrate the performance of this design using simulation data to identify the maximum tolerated dose.
Collapse
Affiliation(s)
- Se Yoon Lee
- Pharmacometrics, EMD Serono R&D Institute, 45A Middlesex Turnpike, Billerica, MA 01821, USA; Department of Statistics, Texas A&M University, College Station, TX 77843, USA
| | - Alain Munafo
- Merck Institute for Pharmacometrics, EPFL Innovation Park, Building I, CH-1015 Lausanne, Switzerland
| | - Pascal Girard
- Merck Institute for Pharmacometrics, EPFL Innovation Park, Building I, CH-1015 Lausanne, Switzerland
| | - Kosalaram Goteti
- Pharmacometrics, EMD Serono R&D Institute, 45A Middlesex Turnpike, Billerica, MA 01821, USA.
| |
Collapse
|
7
|
Zhu J, Sabanés Bové D, Liao Z, Beyer U, Yung G, Sarkar S. Rolling continual reassessment method with overdose control: An efficient and safe dose escalation design. Contemp Clin Trials 2021; 107:106436. [PMID: 34000410 DOI: 10.1016/j.cct.2021.106436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 02/27/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022]
Abstract
In phase 1 dose escalation studies, dose limiting toxicities (DLTs) are defined as adverse events of concern occurring during a predefined time window after first dosing of patients. Standard dose escalation designs, such as the continual reassessment method (CRM), only utilize this binary DLT information. Thus, late-onset DLTs are usually not accounted for when CRM guiding the dose escalation and finally defining the maximum tolerated dose (MTD) of the drug, which brings safety concerns for patients. Previously, several extensions of CRMs, such as the time-to-event CRM (TITE-CRM), fractional CRM (fCRM) and the data augmented CRM (DA-CRM), have been proposed to handle this issue without prolonging trial duration. However, among the model-based designs, none of the designs have explicitly controlled the risk of overdosing as in the escalation with overdose control (EWOC) design. Here we propose a novel dose escalation with overdose control design using a two-parameter logistic regression model for the probability of DLT depending on the dose and a piecewise exponential model for the time to DLT distribution, which we call rolling-CRM design. A comprehensive simulation study has been conducted to compare the performance of the rolling-CRM design with other dose escalation designs. Of note, the trial duration is significantly shorter compared to traditional CRM designs. The proposed design also retains overdose control characteristics, but might require a larger sample size compared to traditional CRM designs.
Collapse
Affiliation(s)
- Jiawen Zhu
- Genentech, Product Development Data Sciences, South San Francisco, CA 94080, USA.
| | - Daniel Sabanés Bové
- F. Hoffmann-La Roche, Product Development Data Sciences, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Ziwei Liao
- Dept. of Biostatistics, Mailman School of Public Health, Columbia University, USA
| | - Ulrich Beyer
- F. Hoffmann-La Roche, Product Development Data Sciences, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Godwin Yung
- Genentech, Product Development Data Sciences, South San Francisco, CA 94080, USA
| | | |
Collapse
|
8
|
Zhang W, Wang X, Muthukumarana S, Yang P. A continual reassessment method without undue risk of toxicity. COMMUN STAT-SIMUL C 2021. [DOI: 10.1080/03610918.2021.1877306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Weijia Zhang
- Chongqing Key Laboratory of Social Economy and Applied Statistics, College of Mathematics and Statistics, Chongqing Technology and Business University, Chongqing, P. R. China
| | - Xikui Wang
- Warren Centre for Actuarial Studies and Research, I.H. Asper School of Business, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Saman Muthukumarana
- Department of Statistics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Po Yang
- Department of Statistics, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
9
|
Yin G, Yang Z. Fractional design: An alternative paradigm for late-onset toxicities in oncology dose-finding studies. Contemp Clin Trials Commun 2020; 19:100650. [PMID: 32875142 PMCID: PMC7451759 DOI: 10.1016/j.conctc.2020.100650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/05/2020] [Accepted: 08/16/2020] [Indexed: 11/17/2022] Open
Abstract
Late-onset (LO) toxicities often arise in the new era of phase I oncology dose-finding trials with targeted agents or immunotherapies. The current LO toxicities modelling is often formulated in a weighted likelihood framework, where the time-to-event continual reassessment method (TITE-CRM) is commonly used. The TITE-CRM uses the patient exposure time as a weight for the censored observation, while there is large uncertainty on which weight function to be used. As an alternative, the fractional scheme formulates an efficient and robust paradigm to address LO toxicity issues in dose finding. We review the fractional continual reassessment method (fCRM) and compare its operating characteristics with those of the TITE-CRM as well as other competitive designs via extensive simulation studies based on both the fixed and randomly generated scenarios. The fCRM is shown to possess desirable operating characteristics in identifying the maximum tolerated dose (MTD) and deliver competitive performances in comparison with other designs. It provides an alternative efficient and robust paradigm for interpreting and addressing LO toxicities in the new era of phase I dose-finding trials in precision oncology. A real trial example is used to illustrate the practical use of the fCRM design.
Collapse
Affiliation(s)
- Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zhao Yang
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
10
|
Dinart D, Fraisse J, Tosi D, Mauguen A, Touraine C, Gourgou S, Le Deley MC, Bellera C, Mollevi C. GUIP1: a R package for dose escalation strategies in phase I cancer clinical trials. BMC Med Inform Decis Mak 2020; 20:134. [PMID: 32580715 PMCID: PMC7469913 DOI: 10.1186/s12911-020-01149-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 06/05/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The main objective of phase I cancer clinical trials is to identify the maximum tolerated dose, usually defined as the highest dose associated with an acceptable level of severe toxicity during the first cycle of treatment. Several dose-escalation designs based on mathematical modeling of the dose-toxicity relationship have been developed. The main ones are: the continual reassessment method (CRM), the escalation with overdose control (EWOC) method and, for late-onset and cumulative toxicities, the time-to-event continual reassessment method (TITE-CRM) and the time-to-event escalation with overdose control (TITE-EWOC) methods. The objective of this work was to perform a user-friendly R package that combines the latter model-guided adaptive designs. RESULTS GUIP1 is an R Graphical User Interface for dose escalation strategies in Phase 1 cancer clinical trials. It implements the CRM (based on Bayesian or maximum likelihood estimation), EWOC and TITE-CRM methods using the dfcrm and bcrm R packages, while the TITE-EWOC method has been specifically developed. The program is built using the TCL/TK programming language, which can be compiled via R software libraries (tcltk, tkrplot, tcltk2). GUIP1 offers the possibility of simulating and/or conducting and managing phase I clinical trials in real-time using file management options with automatic backup of study and/or simulation results. CONCLUSIONS GUIP1 is implemented using the software R, which is widely used by statisticians in oncology. This package simplifies the use of the main model-based dose escalation methods and is designed to be fairly simple for beginners in R. Furthermore, it offers multiple possibilities such as a full traceability of the study. By including multiple innovative adaptive methods in a free and user-friendly program, we hope that GUIP1 will promote and facilitate their use in designing future phase I cancer clinical trials.
Collapse
Affiliation(s)
- D. Dinart
- Inserm CIC1401, Module Epidémiologie Clinique, Institut Bergonié, Bordeaux, France
| | - J. Fraisse
- Institut du Cancer Montpellier (ICM), Université de Montpellier, Montpellier, France
| | - D. Tosi
- Institut du Cancer Montpellier (ICM), Université de Montpellier, Montpellier, France
| | - A. Mauguen
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - C. Touraine
- Institut du Cancer Montpellier (ICM), Université de Montpellier, Montpellier, France
| | - S. Gourgou
- Institut du Cancer Montpellier (ICM), Université de Montpellier, Montpellier, France
| | - M. C. Le Deley
- Centre Oscar Lambret, Lille, France
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - C. Bellera
- Inserm CIC1401, Module Epidémiologie Clinique, Institut Bergonié, Bordeaux, France
| | - C. Mollevi
- Institut du Cancer Montpellier (ICM), Université de Montpellier, Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier INSERM U1194, Université de Montpellier, Montpellier, France
| |
Collapse
|
11
|
van Werkhoven E, Hinsley S, Frangou E, Holmes J, de Haan R, Hawkins M, Brown S, Love SB. Practicalities in running early-phase trials using the time-to-event continual reassessment method (TiTE-CRM) for interventions with long toxicity periods using two radiotherapy oncology trials as examples. BMC Med Res Methodol 2020; 20:162. [PMID: 32571298 PMCID: PMC7477911 DOI: 10.1186/s12874-020-01012-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 05/10/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Awareness of model-based designs for dose-finding studies such as the Continual Reassessment Method (CRM) is now becoming more commonplace amongst clinicians, statisticians and trial management staff. In some settings toxicities can occur a long time after treatment has finished, resulting in extremely long, interrupted, CRM design trials. The Time-to-Event CRM (TiTE-CRM), a modification to the original CRM, accounts for the timing of late-onset toxicities and results in shorter trial duration. In this article, we discuss how to design and deliver a trial using this method, from the grant application stage through to dissemination, using two radiotherapy trials as examples. METHODS The TiTE-CRM encapsulates the dose-toxicity relationship with a statistical model. The model incorporates observed toxicities and uses a weight to account for the proportion of completed follow-up of participants without toxicity. This model uses all available data to determine the next participant's dose and subsequently declare the maximum tolerated dose. We focus on two trials designed by the authors to illustrate practical issues when designing, setting up, and running such studies. RESULTS In setting up a TiTE-CRM trial, model parameters need to be defined and the time element involved might cause complications, therefore looking at operating characteristics through simulations is essential. At the grant application stage, we suggest resources to fund statisticians' time before funding is awarded and make recommendations for the level of detail to include in funding applications. While running the trial, close contact of all involved staff is required as a dose decision is made each time a participant is recruited. We suggest ways of capturing data in a timely manner and give example code in R for design and delivery of the trial. Finally, we touch upon dissemination issues while the trial is running and upon completion. CONCLUSION Model-based designs can be complex. We hope this paper will help clinical trial teams to demystify the conduct of TiTE-CRM trials and be a starting point for using this methodology in practice.
Collapse
Affiliation(s)
| | - Samantha Hinsley
- Cancer Research UK Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | | | - Jane Holmes
- Centre for Statistics in Medicine, NDORMS, University of Oxford, Oxford, UK
| | | | - Maria Hawkins
- CRUK MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Oxford, UK
| | - Sarah Brown
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | | |
Collapse
|
12
|
Huang B. Some statistical considerations in the clinical development of cancer immunotherapies. Pharm Stat 2017; 17:49-60. [PMID: 29098766 DOI: 10.1002/pst.1835] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 09/17/2017] [Accepted: 09/29/2017] [Indexed: 12/11/2022]
Abstract
Immuno-oncology has emerged as an exciting new approach to cancer treatment. Common immunotherapy approaches include cancer vaccine, effector cell therapy, and T-cell-stimulating antibody. Checkpoint inhibitors such as cytotoxic T lymphocyte-associated antigen 4 and programmed death-1/L1 antagonists have shown promising results in multiple indications in solid tumors and hematology. However, the mechanisms of action of these novel drugs pose unique statistical challenges in the accurate evaluation of clinical safety and efficacy, including late-onset toxicity, dose optimization, evaluation of combination agents, pseudoprogression, and delayed and lasting clinical activity. Traditional statistical methods may not be the most accurate or efficient. It is highly desirable to develop the most suitable statistical methodologies and tools to efficiently investigate cancer immunotherapies. In this paper, we summarize these issues and discuss alternative methods to meet the challenges in the clinical development of these novel agents. For safety evaluation and dose-finding trials, we recommend the use of a time-to-event model-based design to handle late toxicities, a simple 3-step procedure for dose optimization, and flexible rule-based or model-based designs for combination agents. For efficacy evaluation, we discuss alternative endpoints/designs/tests including the time-specific probability endpoint, the restricted mean survival time, the generalized pairwise comparison method, the immune-related response criteria, and the weighted log-rank or weighted Kaplan-Meier test. The benefits and limitations of these methods are discussed, and some recommendations are provided for applied researchers to implement these methods in clinical practice.
Collapse
Affiliation(s)
- Bo Huang
- Pfizer Inc, Groton, 06340, CT, USA
| |
Collapse
|
13
|
Colin P, Delattre M, Minini P, Micallef S. An Escalation for Bivariate Binary Endpoints Controlling the Risk of Overtoxicity (EBE-CRO): Managing Efficacy and Toxicity in Early Oncology Clinical Trials. J Biopharm Stat 2017; 27:1054-1072. [DOI: 10.1080/10543406.2017.1295248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- P. Colin
- AgroParisTech, UMR 518 MIA, Paris, France
- Statistical Science & Modeling, Sanofi R&D, Chilly-Mazarin, France
| | - M. Delattre
- AgroParisTech, UMR 518 MIA, Paris, France
- INRA, UMR 518 MIA, Paris, France
| | - P. Minini
- Biostatistiques, Sanofi R&D, Chilly-Mazarin, France
| | - S. Micallef
- Clinical Pharmacometrics, Roche Pharma Research and Early Development, Basel, Switzerland
| |
Collapse
|
14
|
Lin R, Yin G. Nonparametric overdose control with late-onset toxicity in phase I clinical trials. Biostatistics 2016; 18:180-194. [DOI: 10.1093/biostatistics/kxw038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 11/12/2022] Open
|
15
|
Doussau A, Geoerger B, Jiménez I, Paoletti X. Innovations for phase I dose-finding designs in pediatric oncology clinical trials. Contemp Clin Trials 2016; 47:217-27. [PMID: 26825023 DOI: 10.1016/j.cct.2016.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/14/2016] [Accepted: 01/23/2016] [Indexed: 01/24/2023]
Abstract
Phase I oncology clinical trials are designed to identify the optimal dose that will be recommended for phase II trials. In pediatric oncology, the conduct of those trials raises specific challenges, as the disease is rare with limited therapeutic options. In addition, the tolerance profile is known from adult trials. This paper provides a review of the major recent developments in the design of these trials, inspired by the need to cope with the specific challenges of dose finding in cancer pediatric oncology. We reviewed simulation studies comparing designs dedicated to address these challenges. We also reviewed the design used in published dose-finding trials in pediatric oncology over the period 2009-2014. Three main fields of innovation were identified. First, designs that were developed in order to relax the rules for more flexible inclusions. Second, methods to incorporate data emerging from adult studies. Third, designs accounting for toxicity evaluation at repeated cycles in pediatric oncology. In addition to this overview, we propose some further directions for designing pediatric dose-finding trials.
Collapse
Affiliation(s)
- Adelaide Doussau
- National Institutes of Health, Clinical Center, Department of Bioethics, Bethesda, MD, USA.
| | - Birgit Geoerger
- Gustave Roussy, Pediatric and Adolescent Oncology, Villejuif, France; CNRS UMR8203, Univ. Paris-Sud, Univ. Paris-Saclay, Villejuif, France
| | - Irene Jiménez
- Institut Curie, Pediatric, Adolescent and Young Adults Department, Paris, France
| | - Xavier Paoletti
- Gustave Roussy, Biostatistics and Epidemiology unit, Villejuif, France; INSERM U1018, CESP, Univ. Paris-Sud, Univ. Paris-Saclay, Villejuif, France
| |
Collapse
|
16
|
Neuenschwander B, Wandel S, Roychoudhury S, Bailey S. Robust exchangeability designs for early phase clinical trials with multiple strata. Pharm Stat 2015; 15:123-34. [DOI: 10.1002/pst.1730] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/28/2015] [Accepted: 11/02/2015] [Indexed: 11/09/2022]
|
17
|
Chen Z, Yuan Y, Li Z, Kutner M, Owonikoko T, Curran WJ, Khuri F, Kowalski J. Dose escalation with over-dose and under-dose controls in Phase I/II clinical trials. Contemp Clin Trials 2015; 43:133-41. [PMID: 26012358 DOI: 10.1016/j.cct.2015.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 10/23/2022]
Abstract
To save valuable time and resources in new drug development, Phase I/II clinical trials with toxicity control and drug efficacy as dual primary endpoints have become increasingly popular. Escalation with over-dose control (the EWOC) is a Bayesian adaptive Phase I clinical trial design that can accurately estimate the maximum tolerated dose (MTD) level and control the probability of overdosing patients during the dose allocation phase. In this paper, we extend EWOC to Phase I/II clinical trials by controlling for under-dosing with a Gumbel Copula model to provide patients with at least minimum drug efficacy. We propose a utility function to measure the composite effect of toxicity and efficacy and select the optimal dose. To deal with the common issue that the efficacy endpoint often cannot be quickly ascertained, we employ Bayesian data augmentation to handle delayed efficacy and allow for flexible patient accrual without a waiting period. Extensive simulations demonstrate that the proposed new design not only provides better therapeutic effect by reducing the probability of treating patients at under-dose levels while protecting patients from being overdosed, but also improves trial efficiency and increases the accuracy of dose recommendation for subsequent clinical trials. We apply the proposed design to a Phase I/II solid tumor trial.
Collapse
Affiliation(s)
- Zhengjia Chen
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States; Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States; Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, United States; Department of Radiology and Imaging Science, Emory University, Atlanta, GA 30322, United States.
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Zheng Li
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States
| | - Michael Kutner
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States
| | - Taofeek Owonikoko
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, United States
| | - Walter J Curran
- Department of Radiation Oncology, Emory University, Atlanta, GA 30322, United States
| | - Fadlo Khuri
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, United States
| | - Jeanne Kowalski
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States; Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States
| |
Collapse
|
18
|
Huang B, Kuan PF. Time-to-event continual reassessment method incorporating treatment cycle information with application to an oncology phase I trial. Biom J 2014; 56:933-46. [DOI: 10.1002/bimj.201300261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/05/2014] [Accepted: 04/07/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Bo Huang
- Pfizer Inc; 445 Eastern Point Road Groton CT 06340 USA
| | - Pei Fen Kuan
- Department of Applied Mathematics and Statistics; Stony Brook University; Stony Brook NY 11790 USA
| |
Collapse
|
19
|
Tighiouart M, Piantadosi S, Rogatko A. Dose finding with drug combinations in cancer phase I clinical trials using conditional escalation with overdose control. Stat Med 2014; 33:3815-29. [PMID: 24825779 DOI: 10.1002/sim.6201] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 02/28/2014] [Accepted: 04/19/2014] [Indexed: 11/12/2022]
Abstract
We present a Bayesian adaptive design for dose finding of a combination of two drugs in cancer phase I clinical trials. The goal is to estimate the maximum tolerated dose (MTD) as a curve in the two-dimensional Cartesian plane. We use a logistic model to describe the relationship between the doses of the two agents and the probability of dose limiting toxicity. The model is re-parameterized in terms of parameters clinicians can easily interpret. Trial design proceeds using univariate escalation with overdose control, where at each stage of the trial, we seek a dose of one agent using the current posterior distribution of the MTD of this agent given the current dose of the other agent. At the end of the trial, an estimate of the MTD curve is proposed as a function of Bayes estimates of the model parameters. We evaluate design operating characteristics in terms of safety of the trial design and percent of dose recommendation at dose combination neighborhoods around the true MTD curve. We also examine the performance of the approach under model misspecifications for the true dose-toxicity relationship.
Collapse
Affiliation(s)
- Mourad Tighiouart
- Samuel Oschin Comprehensive Cancer Institute, 8700 Beverly Blvd., Los Angeles, CA, 90048, U.S.A
| | | | | |
Collapse
|
20
|
Escalation with overdose control using time to toxicity for cancer phase I clinical trials. PLoS One 2014; 9:e93070. [PMID: 24663812 PMCID: PMC3963973 DOI: 10.1371/journal.pone.0093070] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 03/01/2014] [Indexed: 11/19/2022] Open
Abstract
Escalation with overdose control (EWOC) is a Bayesian adaptive phase I clinical trial design that produces consistent sequences of doses while controlling the probability that patients are overdosed. However, this design does not take explicitly into account the time it takes for a patient to exhibit dose limiting toxicity (DLT) since the occurrence of DLT is ascertained within a predetermined window of time. Models to estimate the Maximum Tolerated Dose (MTD) that use the exact time when the DLT occurs are expected to be more precise than those where the variable of interest is categorized as presence or absence of DLT, given that information is lost in the process of categorization of the variable. We develop a class of parametric models for time to toxicity data in order to estimate the MTD efficiently, and present extensive simulations showing that the method has good design operating characteristics relative to the original EWOC and a version of time to event EWOC (TITE-EWOC) which allocates weights to account for the time it takes for a patient to exhibit DLT. The methodology is exemplified by a cancer phase I clinical trial we designed in order to estimate the MTD of Veliparib (ABT-888) in combination with fixed doses of gemcitabine and intensity modulated radiation therapy in patients with locally advanced, un-resectable pancreatic cancer.
Collapse
|
21
|
Chen Z, Cui Y, Owonikoko TK, Wang Z, Li Z, Luo R, Kutner M, Khuri FR, Kowalski J. Escalation with overdose control using all toxicities and time to event toxicity data in cancer Phase I clinical trials. Contemp Clin Trials 2014; 37:322-32. [PMID: 24530487 DOI: 10.1016/j.cct.2014.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 01/27/2014] [Accepted: 02/05/2014] [Indexed: 11/18/2022]
Abstract
The primary purposes of Phase I cancer clinical trials are to determine the maximum tolerated dose (MTD) and the treatment schedule of a new drug. Phase I trials usually involve a small number of patients so that fully utilizing all toxicity information including time to event toxicity data is key to improving the trial efficiency and the accuracy of MTD estimation. Chen et al. proposed a novel normalized equivalent toxicity score (NETS) system to fully utilize multiple toxicities per patient instead of a binary indicator of dose limiting toxicity (DLT). Cheung and Chappell developed the time to toxicity event (TITE) approach to incorporate time to toxicity event data. Escalation with overdose control (EWOC) is an adaptive Bayesian Phase I design which can allow rapid dose escalation while controlling the probability of overdosing patients. In this manuscript, we use EWOC as a framework and integrate it with the NETS system and the TITE approach to develop an advanced Phase I design entitled EWOC-NETS-TITE. We have conducted simulation studies to compare its operating characteristics using selected derived versions of EWOC because EWOC itself has already been extensively compared with common Phase I designs [3]. Simulation results demonstrate that EWOC-NETS-TITE can substantially improve the trial efficiency and accuracy of MTD determination as well as allow patients to be entered in a staggered fashion to significantly shorten trial duration. Moreover, user-friendly software for EWOC-NETS-TITE is under development.
Collapse
Affiliation(s)
- Zhengjia Chen
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States; Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States.
| | - Ye Cui
- ICF International, 3 Corporate Square, NE, Suite 370, Atlanta, GA 30329, United States
| | - Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, United States
| | - Zhibo Wang
- Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States
| | - Zheng Li
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States
| | - Ruiyan Luo
- School of Public Health, Georgia State University, Atlanta, GA 30303, United States
| | - Michael Kutner
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, United States
| | - Jeanne Kowalski
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, United States; Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States
| |
Collapse
|
22
|
Yin G, Zheng S, Xu J. Fractional dose-finding methods with late-onset toxicity in phase I clinical trials. J Biopharm Stat 2014; 23:856-70. [PMID: 23786314 DOI: 10.1080/10543406.2013.789892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In Phase I clinical trials, the algorithm-based dose-finding methods, such as the 3 + 3 and up-and-down designs, do not impose any dose-toxicity curve. In contrast, model-based designs, such as the continual reassessment method (CRM), assume a parametric model to borrow information from all the doses under consideration. For these conventional dose-finding methods, toxicity outcomes need to be observed shortly after the treatment, so that newly enrolled patients can be treated without delay. However, in the case of late-onset toxicity, patients' outcomes may not be observed quickly enough to keep up with the speed of enrollment, and thus toxicity data may not be available when that information is needed. Patients who have not experienced toxicity by the decision-making time may yet experience toxicity later during the rest of the follow-up. Ignoring such late-onset toxicity information may lead to biased estimation of the dose toxicity probabilities and thus compromise the trial's performance. To expand the applicability of the 3 + 3, up-and-down, and CRM designs with late-onset toxicity, we propose to redistribute the mass of the censored observation to the right and utilize the fractional contribution for the unobserved toxicity outcome. We evaluate the operating characteristics of the proposed fractional designs through extensive simulation studies. The fractional designs satisfactorily resolve the issues associated with late-onset toxicity, and are compared favorably with other available methods.
Collapse
Affiliation(s)
- Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, China.
| | | | | |
Collapse
|
23
|
Sverdlov O, Wong WK, Ryeznik Y. Adaptive clinical trial designs for phase I cancer studies. STATISTICS SURVEYS 2014. [DOI: 10.1214/14-ss106] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
24
|
Liu S, Yin G, Yuan Y. BAYESIAN DATA AUGMENTATION DOSE FINDING WITH CONTINUAL REASSESSMENT METHOD AND DELAYED TOXICITY. Ann Appl Stat 2013; 7:1837-2457. [PMID: 24707327 DOI: 10.1214/13-aoas661] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A major practical impediment when implementing adaptive dose-finding designs is that the toxicity outcome used by the decision rules may not be observed shortly after the initiation of the treatment. To address this issue, we propose the data augmentation continual re-assessment method (DA-CRM) for dose finding. By naturally treating the unobserved toxicities as missing data, we show that such missing data are nonignorable in the sense that the missingness depends on the unobserved outcomes. The Bayesian data augmentation approach is used to sample both the missing data and model parameters from their posterior full conditional distributions. We evaluate the performance of the DA-CRM through extensive simulation studies, and also compare it with other existing methods. The results show that the proposed design satisfactorily resolves the issues related to late-onset toxicities and possesses desirable operating characteristics: treating patients more safely, and also selecting the maximum tolerated dose with a higher probability. The new DA-CRM is illustrated with two phase I cancer clinical trials.
Collapse
|
25
|
Doussau A, Thiébaut R, Paoletti X. Dose-finding design using mixed-effect proportional odds model for longitudinal graded toxicity data in phase I oncology clinical trials. Stat Med 2013; 32:5430-47. [PMID: 24018535 DOI: 10.1002/sim.5960] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 06/20/2013] [Accepted: 08/05/2013] [Indexed: 11/06/2022]
Abstract
Phase I oncology clinical trials are designed to identify the optimal dose that will be recommended for phase II trials. This dose is typically defined as the dose associated with a certain probability of severe toxicity during the first cycle of treatment, although toxicity is repeatedly measured over cycles on an ordinal scale. We propose a new adaptive dose-finding design using longitudinal measurements of ordinal toxic adverse events, with proportional odds mixed-effect models. Likelihood-based inference is implemented. The optimal dose is then the dose producing a target rate of severe toxicity per cycle. This model can also be used to identify cumulative or late toxicities. The performances of this approach were compared with those of the continual reassessment method in a simulation study. Operating characteristics were evaluated in terms of correct identification of the target dose, distribution of the doses allocated and power to detect trends in the risk of toxicities over time. This approach was also used to reanalyse data from a phase I oncology trial. Use of a proportional odds mixed-effect model appears to be feasible in phase I dose-finding trials, increases the ability of selecting the correct dose and provides a tool to detect cumulative effects.
Collapse
Affiliation(s)
- Adélaïde Doussau
- INSERM, ISPED, Centre INSERM U897-Epidemiologie-Biostatistique, F-33000 Bordeaux, France; CHU de Bordeaux, Pole de Santé Publique, F-33000 Bordeaux, France; Univ. Bordeaux, ISPED, CIC-EC7, F-33000 Bordeaux, France; INSERM, U900, F-75005 Paris, France
| | | | | |
Collapse
|
26
|
Liu S, Ning J. A Bayesian Dose-finding Design for Drug Combination Trials with Delayed Toxicities. BAYESIAN ANALYSIS 2013; 8:703-722. [PMID: 27924182 PMCID: PMC5136476 DOI: 10.1214/13-ba839] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We propose a Bayesian adaptive dose-finding design for drug combination trials with delayed toxicity. We model the dose-toxicity relationship using the Finney model, a model widely used in drug-drug interaction studies. The intuitive interpretations of the Finney model facilitate incorporating the available prior dose-toxicity information from single-agent trials into combination trials through prior elicitation. We treat unobserved delayed toxicity outcomes as missing data and handle them using Bayesian data augmentation. We conduct extensive simulation studies to examine the operating characteristics of the proposed method under various practical scenarios. Results show that the proposed design is safe and able to select the target dose combinations with high probabilities.
Collapse
Affiliation(s)
- Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX,
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX,
| |
Collapse
|
27
|
Shi Y, Yin G. Escalation with overdose control for phase I drug-combination trials. Stat Med 2013; 32:4400-12. [PMID: 23630103 DOI: 10.1002/sim.5832] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 04/02/2013] [Indexed: 11/06/2022]
Abstract
Dose finding for combined drugs has grown rapidly in oncology drug development. The escalation with overdose control (EWOC) method is a popular model-based dose-finding approach to single-agent phase I clinical trials. When two drugs are combined as a treatment, we propose a two-dimensional EWOC design for dose finding on the basis of a four-parameter logistic regression model. During trial conduct, we continuously update the posterior distribution of the maximum tolerated dose (MTD) combination to find the most appropriate dose combination for each cohort of patients. The probability that the next assigned dose combination exceeds the MTD combination can be controlled by a feasibility bound, which is based on a prespecified quantile level of the MTD distribution such as to reduce the possibility of overdosing. We determine dose escalation, de-escalation, or staying at the same doses by searching the MTD combination along the rows and columns in a two-drug combination matrix, respectively. We conduct simulation studies to examine the performance of the two-dimensional EWOC design under various practical scenarios, and illustrate it with a trial example.
Collapse
Affiliation(s)
- Yun Shi
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong
| | | |
Collapse
|
28
|
Vassal G, Zwaan CM, Ashley D, Le Deley MC, Hargrave D, Blanc P, Adamson PC. New drugs for children and adolescents with cancer: the need for novel development pathways. Lancet Oncol 2013; 14:e117-24. [PMID: 23434337 DOI: 10.1016/s1470-2045(13)70013-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite major progress in the past 40 years, 20% of children with cancer die from the disease, and 40% of survivors have late adverse effects. Innovative, safe, and effective medicines are needed. Although regulatory initiatives in the past 15 years in the USA and Europe have been introduced, new drug development for children with cancer is insufficient. Children and families face major inequity between countries in terms of access to innovative drugs in development. Hurdles and bottlenecks are well known-eg, small numbers of patients, the complexity of developing targeted agents and their biomarkers for selected patients, limitations of US and EU regulations for paediatric medicines, insufficient return on investment, and the global economic crisis facing drug companies. New drug development pathways could efficiently address the challenges with innovative methods and trial designs, investment in biology and preclinical research, new models of partnership and funding including public-private partnerships and precompetitive research consortia, improved regulatory requirements, initiatives and incentives that better address these needs, and increased collaboration between paediatric oncology cooperative groups worldwide. Increased cooperation between all stakeholders-academia, parents' organisations and advocacy groups, regulatory bodies, pharmaceutical companies, philanthropic organisations, and government-will be essential.
Collapse
Affiliation(s)
- Gilles Vassal
- Division of Clinical Research, Institut Gustave Roussy, Paris-Sud University, Paris, France.
| | | | | | | | | | | | | |
Collapse
|