1
|
Yuan Y, Zhou H, Liu S. Statistical and practical considerations in planning and conduct of dose-optimization trials. Clin Trials 2024; 21:273-286. [PMID: 38243399 PMCID: PMC11134987 DOI: 10.1177/17407745231207085] [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] [Indexed: 01/21/2024]
Abstract
The U.S. Food and Drug Administration launched Project Optimus with the aim of shifting the paradigm of dose-finding and selection toward identifying the optimal biological dose that offers the best balance between benefit and risk, rather than the maximum tolerated dose. However, achieving dose optimization is a challenging task that involves a variety of factors and is considerably more complicated than identifying the maximum tolerated dose, both in terms of design and implementation. This article provides a comprehensive review of various design strategies for dose-optimization trials, including phase 1/2 and 2/3 designs, and highlights their respective advantages and disadvantages. In addition, practical considerations for selecting an appropriate design and planning and executing the trial are discussed. The article also presents freely available software tools that can be utilized for designing and implementing dose-optimization trials. The approaches and their implementation are illustrated through real-world examples.
Collapse
Affiliation(s)
- Ying Yuan
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heng Zhou
- Biostatistics and Research Decision Sciences, Merck and Co., Inc, Rahway, NJ, USA
| | - Suyu Liu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
2
|
Celum C, Horton BJ, Conaway M. The quasi-CRM shift method for partially ordered groups. Contemp Clin Trials 2024; 136:107400. [PMID: 38000453 DOI: 10.1016/j.cct.2023.107400] [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: 07/03/2023] [Revised: 11/01/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023]
Abstract
This paper proposes a phase-I clinical trial design that uses ordinal toxicity to locate group-specific doses when groups are partially or completely ordered prior to the start of the trial. There has been previous work on dose-finding for groups and on dose-finding with ordinal toxicity but a solution to the problem of dose-finding for groups with ordinal toxicity has not been proposed. Simulations compared the proposed method against two methods; one that uses ordinal toxicity but does not use group information and one that uses group information but does not use ordinal toxicity. One issue with the first method is the potential for reversals, when the recommended dose for a more sensitive group is higher than the recommended dose for a less sensitive group. The proposed method avoids reversals, allocates patients to optimal doses more frequently during the trial, and selects optimal doses more frequently at the end of the trial.
Collapse
Affiliation(s)
- Connor Celum
- Department of Statistics, University of Virginia, Charlottesville, VA, USA.
| | - Bethany Jablonski Horton
- Division Of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Mark Conaway
- Department of Statistics, University of Virginia, Charlottesville, VA, USA; Division Of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
3
|
Takeda K, Yamaguchi Y, Taguri M, Morita S. TITE-gBOIN-ET: Time-to-event generalized Bayesian optimal interval design to accelerate dose-finding accounting for ordinal graded efficacy and toxicity outcomes. Biom J 2023; 65:e2200265. [PMID: 37309248 DOI: 10.1002/bimj.202200265] [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: 09/23/2022] [Revised: 03/17/2023] [Accepted: 05/08/2023] [Indexed: 06/14/2023]
Abstract
One of the primary objectives of an oncology dose-finding trial for novel therapies, such as molecular-targeted agents and immune-oncology therapies, is to identify an optimal dose (OD) that is tolerable and therapeutically beneficial for subjects in subsequent clinical trials. These new therapeutic agents appear more likely to induce multiple low or moderate-grade toxicities than dose-limiting toxicities. Besides, for efficacy, evaluating the overall response and long-term stable disease in solid tumors and considering the difference between complete remission and partial remission in lymphoma are preferable. It is also essential to accelerate early-stage trials to shorten the entire period of drug development. However, it is often challenging to make real-time adaptive decisions due to late-onset outcomes, fast accrual rates, and differences in outcome evaluation periods for efficacy and toxicity. To solve the issues, we propose a time-to-event generalized Bayesian optimal interval design to accelerate dose finding, accounting for efficacy and toxicity grades. The new design named "TITE-gBOIN-ET" design is model-assisted and straightforward to implement in actual oncology dose-finding trials. Simulation studies show that the TITE-gBOIN-ET design significantly shortens the trial duration compared with the designs without sequential enrollment while having comparable or higher performance in the percentage of correct OD selection and the average number of patients allocated to the ODs across various realistic settings.
Collapse
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Yusuke Yamaguchi
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Masataka Taguri
- Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Ling H, Shi H, Yuan N, Ji Y, Lin X. qTPI: A quasi-toxicity probability interval design for phase I trials with multiple-grade toxicities. Stat Methods Med Res 2023; 32:1389-1402. [PMID: 37278183 DOI: 10.1177/09622802231176034] [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] [Indexed: 06/07/2023]
Abstract
The common terminology criteria for adverse events by the National Cancer Institute has greatly facilitated the revolution of drug development and an increasing number of Phase I trials have started to collect multiple-grade toxicity endpoints. Appropriate and yet transparent Phase I statistical designs for multiple-grade toxicities are therefore in great needs. In this article, we propose a quasi-toxicity probability interval (qTPI) design that incorporates a quasi-continuous measure of the toxicity probability (q T P ) into the Bayesian theoretic framework of the interval based designs. Multiple-grade toxicity outcomes of each patient are mapped to q T P according to a severity weight matrix. Dose-toxicity curve underlying the dosing decisions in the qTPI design is continuously updated using accumulating trial data. Numerical simulations investigating the operating characteristics of qTPI show that qTPI achieved better safety, accuracy and reliability compared to designs that rely on binary toxicity data. Furthermore, parameter elicitation in qTPI is simple and does not involve multiple hypothetical cohorts specification. Finally, a hypothetical soft tissue sarcoma trial with six toxicity types and grade 0 to grade 4 severity grades is illustrated with patient-by-patient dose allocation under the qTPI design.
Collapse
Affiliation(s)
- Haodong Ling
- School of Data Science, Fudan University, Shanghai, China
| | - Haolun Shi
- Department of Statistics and Actuarial Science, Simon Fraser University, Burnaby, Canada
| | - Nan Yuan
- School of Data Science, Fudan University, Shanghai, China
| | - Yuan Ji
- Department of Public Health Sciences, University of Chicago Chicago, IL, USA
| | - Xiaolei Lin
- School of Data Science, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Wages NA, Nelson B, Kharofa J, Meier T. Application of the patient-reported outcomes continual reassessment method to a phase I study of radiotherapy in endometrial cancer. Int J Biostat 2023; 19:163-176. [PMID: 36394530 PMCID: PMC10238853 DOI: 10.1515/ijb-2022-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/29/2022] [Accepted: 08/07/2022] [Indexed: 07/28/2023]
Abstract
This article considers the concept of designing Phase I clinical trials using both clinician- and patient-reported outcomes to adaptively allocate study participants to tolerable doses and determine the maximum tolerated dose (MTD) at the study conclusion. We describe an application of a Bayesian form of the patient-reported outcomes continual reassessment method (PRO-CRMB) in an ongoing Phase I study of adjuvant hypofractionated whole pelvis radiation therapy (WPRT) in endometrial cancer (NCT04458402). The study's primary objective is to determine the MTD per fraction of WPRT, defined by acceptable clinician- and patient-reported DLT rates. We conduct simulation studies of the operating characteristics of the design and compared them to a rule-based approach. We illustrate that the PRO-CRMB makes appropriate dose assignments during the study to give investigators and reviewers an idea of how the method behaves. In simulation studies, the PRO-CRMB demonstrates superior performance to a 5 + 2 stepwise design in terms of recommending target treatment courses and allocating patients to these courses. The design is accompanied by an easy-to-use R shiny web application to simulate operating characteristics at the design stage and sequentially update dose assignments throughout the trial's conduct.
Collapse
Affiliation(s)
- Nolan A. Wages
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Bailey Nelson
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Jordan Kharofa
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Teresa Meier
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
6
|
O'Connell NS, Wages NA, Garrett-Mayer E. Quasi-partial order continual reassessment method: Applying toxicity scores to cancer dose-finding drug combination trials. Contemp Clin Trials 2023; 125:107050. [PMID: 36529437 DOI: 10.1016/j.cct.2022.107050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
The primary endpoint of most dose-finding cancer trials is patient toxicity, and the primary goal is to identify the maximum tolerated dose (MTD), that is, the highest dose that falls below or within a pre-specified toxicity tolerability threshold. Conventionally, dose-finding methods have utilized a binary toxicity endpoint based on whether or not a patient experiences a dose limiting-toxicity (DLT). Improving upon this, in recent years several methods have been developed for modeling toxicity scores, a novel continuous endpoint designed to more precisely estimate patient toxicity burden. Separately, drug-combination trials have become increasingly prevalent, and due to added complexities regarding estimating 'true' dose ordering and potential for more complex patient toxicity profiles, provide an ideal setting which may benefit from the improved precision of toxicity scores. In this paper, we merge two frameworks based on the Continual Reassessment Method (CRM) - the Quasi-CRM and the Partial Order CRM (POCRM) - to propose a novel approach for modeling toxicity scores in a combination-trial setting. We demonstrate that utilizing toxicity scores has the potential to greatly improve correct dose-selection over a variety of trial scenarios. We further present a simple adaptation to the toxicity-score model to control for potential over-dosing issues such that it adheres to the conventional DLT definition and will, at worst, perform equivalently to that of the traditional binary DLT framework. We demonstrate that extending toxicity scores to the combination-trial setting offers potential for improvement over the conventional binary endpoint models.
Collapse
Affiliation(s)
- Nathaniel S O'Connell
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston Salem, NC, USA.
| | - Nolan A Wages
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Elizabeth Garrett-Mayer
- Center for Research and Analytics, American Society for Clinical Oncology, Alexandria, VA, USA
| |
Collapse
|
7
|
Marchenko O, Sridhara R, Jiang Q, Barksdale E, Ando Y, Alwis DD, Brown K, Fernandes L, van Bussel MT, Choo Q, Coory M, Garrett-Mayer E, Gwise T, Hess L, Liu R, Mandrekar S, Ouellet D, Pinheiro J, Posch M, Rahman NA, Rantell KR, Raven A, Sarem S, Sen S, Shah M, Shen YL, Simon R, Theoret M, Yuan Y, Pazdur R. Designing Dose-Optimization Studies in Cancer Drug Development: Discussions with Regulators. Stat Biopharm Res 2023. [DOI: 10.1080/19466315.2023.2166099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Thomas Gwise
- Office of Biostatistics, CDER US FDA, Silver Spring, MD
| | | | - Rong Liu
- Bristol Myers Squibb, Berkeley Heights, NJ
| | | | | | | | - Martin Posch
- Institute for Medical Statistics at the Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Mirat Shah
- Office of Oncologic Diseases, CDER, US FDA, Silver Spring, MD
| | - Yuan Li Shen
- Office of Biostatistics, CDER US FDA, Silver Spring, MD
| | | | - Marc Theoret
- Oncology Center of Excellence, US FDA, Silver Spring, MD
| | - Ying Yuan
- MD Anderson Cancer Center, Houston, TX
| | - Richard Pazdur
- Oncology Center of Excellence, US FDA, Silver Spring, MD
| |
Collapse
|
8
|
Ananthakrishnan R, Lin R, He C, Chen Y, Li D, LaValley M. An overview of the BOIN design and its current extensions for novel early-phase oncology trials. Contemp Clin Trials Commun 2022; 28:100943. [PMID: 35812822 PMCID: PMC9260438 DOI: 10.1016/j.conctc.2022.100943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/02/2022] [Accepted: 06/07/2022] [Indexed: 11/24/2022] Open
Abstract
Bayesian Optimal Interval (BOIN) designs are a class of model-assisted dose-finding designs that can be used in oncology trials to determine the maximum tolerated dose (MTD) of a study drug based on safety or the optimal biological dose (OBD) based on safety and efficacy. BOIN designs provide a complete suite for dose finding in early phase trials, as well as a consistent way to explore different scenarios such as toxicity, efficacy, continuous outcomes, delayed toxicity or efficacy and drug combinations in a unified manner with easy access to software to implement most of these designs. Although built upon Bayesian probability models, BOIN designs are operationally simple in general and have good statistical operating characteristics compared to other dose-finding designs. This review paper describes the original BOIN design and its many extensions, their advantages and limitations, the software used to implement them, and the most suitable situation for use of each of these designs. Published examples of the implementation of BOIN designs are provided in the Appendix.
Collapse
Affiliation(s)
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chunsheng He
- Bristol-Myers Squibb (BMS), 300 Connell Drive, Berkeley Heights, NJ, 07922, USA
| | - Yanping Chen
- Bristol-Myers Squibb (BMS), 300 Connell Drive, Berkeley Heights, NJ, 07922, USA
| | | | - Michael LaValley
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| |
Collapse
|
9
|
An R package UnifiedDoseFinding for continuous and ordinal outcomes in Phase I dose-finding trials. COMMUNICATIONS FOR STATISTICAL APPLICATIONS AND METHODS 2022. [DOI: 10.29220/csam.2022.29.4.421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
10
|
Takeda K, Morita S, Taguri M. gBOIN-ET: The generalized Bayesian optimal interval design for optimal dose-finding accounting for ordinal graded efficacy and toxicity in early clinical trials. Biom J 2022; 64:1178-1191. [PMID: 35561046 DOI: 10.1002/bimj.202100263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/22/2022] [Accepted: 04/03/2022] [Indexed: 12/19/2022]
Abstract
One of the primary objectives of an oncology dose-finding trial for novel therapies, such as molecular targeted agents and immune-oncology therapies, is to identify an optimal dose (OD) that is tolerable and therapeutically beneficial for subjects in subsequent clinical trials. These new therapeutic agents appear more likely to induce multiple low- or moderate-grade toxicities than dose-limiting toxicities. Besides, efficacy should be evaluated as an overall response and stable disease in solid tumors and the difference between complete remission and partial remission in lymphoma. This paper proposes the generalized Bayesian optimal interval design for dose-finding accounting for efficacy and toxicity grades. The new design, named "gBOIN-ET" design, is model-assisted, simple, and straightforward to implement in actual oncology dose-finding trials than model-based approaches. These characteristics are quite valuable in practice. A simulation study shows that the gBOIN-ET design has advantages compared with the other model-assisted designs in the percentage of correct OD selection and the average number of patients allocated to the ODs across various realistic settings.
Collapse
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, IL, USA
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Taguri
- Department of Data Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
11
|
Mu R, Hu Z, Xu G, Pan H. An adaptive gBOIN design with shrinkage boundaries for phase I dose-finding trials. BMC Med Res Methodol 2021; 21:278. [PMID: 34895153 PMCID: PMC8667395 DOI: 10.1186/s12874-021-01455-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 10/19/2021] [Indexed: 11/21/2022] Open
Abstract
Background With the emergence of molecularly targeted agents and immunotherapies, the landscape of phase I trials in oncology has been changed. Though these new therapeutic agents are very likely induce multiple low- or moderate-grade toxicities instead of DLT, most of the existing phase I trial designs account for the binary toxicity outcomes. Motivated by a pediatric phase I trial of solid tumor with a continuous outcome, we propose an adaptive generalized Bayesian optimal interval design with shrinkage boundaries, gBOINS, which can account for continuous, toxicity grades endpoints and regard the conventional binary endpoint as a special case. Result The proposed gBOINS design enjoys convergence properties, e.g., the induced interval shrinks to the toxicity target and the recommended dose converges to the true maximum tolerated dose with increased sample size. Conclusion The proposed gBOINS design is transparent and simple to implement. We show that the gBOINS design has the desirable finite property of coherence and large-sample property of consistency. Numerical studies show that the proposed gBOINS design yields good performance and is comparable with or superior to the competing design.
Collapse
Affiliation(s)
- Rongji Mu
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zongliang Hu
- College of Mathematics and Statistics, Shenzhen University, Shenzhen, 518060, China.
| | - Guoying Xu
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai, 201203, China
| | - Haitao Pan
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, 38105, TN, USA.
| |
Collapse
|
12
|
Takeda K, Xia Q, Liu S, Rong A. TITE-gBOIN: Time-to-event Bayesian optimal interval design to accelerate dose-finding accounting for toxicity grades. Pharm Stat 2021; 21:496-506. [PMID: 34862715 DOI: 10.1002/pst.2182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 11/08/2022]
Abstract
The new therapeutic agents, such as molecular targeted agents and immuno-oncology therapies, appear more likely to induce multiple toxicities at different grades than dose-limiting toxicities defined in traditional dose-finding trials. In addition, it is often challenging to make adaptive decisions on dose escalation and de-escalation on time because of the fast accrual rate and/or the late-onset toxicity outcomes, causing the potential suspension of the enrollment and the delay of the trials. To address these issues, we propose a time-to-event Bayesian optimal interval design to accelerate the dose-finding process utilizing toxicity grades based on both cumulative and pending toxicity outcomes. The proposed design, named "TITE-gBOIN" design, is a nonparametric and model-assisted design and has the virtues of robustness, simplicity and straightforward to implement in actual oncology dose-finding trials. A simulation study shows that the TITE-gBOIN design has a higher probability of selecting the MTDs correctly and allocating more patients to the MTDs across various realistic settings while reducing the trial duration significantly, therefore can accelerate early-stage dose-finding trials.
Collapse
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Qing Xia
- Biostatistics & Data Science, University of Kansas Medical Center, Kansas, USA
| | - Shufang Liu
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Alan Rong
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| |
Collapse
|
13
|
Yin J, Du Y, Qin R, Shen S, Mandrekar S. phase1RMD: An R package for repeated measures dose-finding designs with novel toxicity and efficacy endpoints. PLoS One 2021; 16:e0256391. [PMID: 34473708 PMCID: PMC8412295 DOI: 10.1371/journal.pone.0256391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
Traditional dose-finding designs are substantially inefficient for targeted agents and cancer immunotherapies by failing to incorporate efficacy signals, mild and moderate adverse events, and late, cumulative toxicities. However, the lack of user-friendly software is a barrier to the practical use of the novel phase I designs, despite their demonstrated superiority of traditional 3+3 designs. To overcome these barriers, we present an R package, phase1RMD, which provides a comprehensive implementation of novel designs with repeated toxicity measures and early efficacy. A novel phase I repeated measures design that used a continuous toxicity score from multiple treatment cycles was implemented. Furthermore, in studies where preliminary efficacy is evaluated, an adaptive, multi-stage design to identify the most efficacious dose with acceptable toxicity was demonstrated. Functions are provided to recommend the next dose based on the data collected in a phase I trial, as well as to assess trial characteristics given design parameters via simulations. The repeated measure designs accurately estimated both the magnitude and direction of toxicity trends in late treatment cycles, and allocated more patients at therapeutic doses. The R package for implementing these designs is available from the Comprehensive R Archive Network. To our best knowledge, this is the first software that implement novel phase I dose-finding designs that simultaneously accounts for the multiple-grade toxicity events over multiple treatment cycles and a continuous early efficacy outcome. With the software published on CRAN, we will pursue the implementation of these designs in phase I trials in real-life settings.
Collapse
Affiliation(s)
- Jun Yin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| | - Yu Du
- Statistics-Diabetes/Endocrine, Eli Lilly and Company, Indianapolis, IN, United States of America
| | - Rui Qin
- Clinical Biostatistics, Janssen Research and Development, Raritan, NJ, United States of America
| | - Shihao Shen
- Data Science & Biostatistics, DermBiont, Boston, MA, United States of America
| | - Sumithra Mandrekar
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| |
Collapse
|
14
|
Jin L, Pang G, Alemayehu D. Multiarmed Bandit Designs for Phase I Dose-Finding Clinical Trials With Multiple Toxicity Types. Stat Biopharm Res 2021. [DOI: 10.1080/19466315.2021.1962402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Lan Jin
- The Harold and Inge Marcus Department of Industrial and Manufacturing Engineering, Pennsylvania State University, Pennsylvania, PA
| | - Guodong Pang
- The Harold and Inge Marcus Department of Industrial and Manufacturing Engineering, Pennsylvania State University, Pennsylvania, PA
| | | |
Collapse
|
15
|
Gerard E, Zohar S, Lorenzato C, Ursino M, Riviere MK. Bayesian modeling of a bivariate toxicity outcome for early phase oncology trials evaluating dose regimens. Stat Med 2021; 40:5096-5114. [PMID: 34259343 PMCID: PMC9292544 DOI: 10.1002/sim.9113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/23/2021] [Accepted: 05/25/2021] [Indexed: 11/05/2022]
Abstract
Most phase I trials in oncology aim to find the maximum tolerated dose (MTD) based on the occurrence of dose limiting toxicities (DLT). Evaluating the schedule of administration in addition to the dose may improve drug tolerance. Moreover, for some molecules, a bivariate toxicity endpoint may be more appropriate than a single endpoint. However, standard dose‐finding designs do not account for multiple dose regimens and bivariate toxicity endpoint within the same design. In this context, following a phase I motivating trial, we proposed modeling the first type of DLT, cytokine release syndrome, with the entire dose regimen using pharmacokinetics and pharmacodynamics (PK/PD), whereas the other DLT (DLTo) was modeled with the cumulative dose. We developed three approaches to model the joint distribution of DLT, defining it as a bivariate binary outcome from the two toxicity types, under various assumptions about the correlation between toxicities: an independent model, a copula model and a conditional model. Our Bayesian approaches were developed to be applied at the end of the dose‐allocation stage of the trial, once all data, including PK/PD measurements, were available. The approaches were evaluated through an extensive simulation study that showed that they can improve the performance of selecting the true MTD‐regimen compared to the recommendation of the dose‐allocation method implemented. Our joint approaches can also predict the DLT probabilities of new dose regimens that were not tested in the study and could be investigated in further stages of the trial.
Collapse
Affiliation(s)
- Emma Gerard
- Inserm, Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, Paris, France.,HeKA, Inria, Paris, France.,Oncology Biostatistics, Biostatistics and Programming Department, Sanofi R&D, Vitry-sur-Seine, France.,Statistical Methodology Group, Biostatistics and Programming Department, Sanofi R&D, Chilly-Mazarin, France
| | - Sarah Zohar
- Inserm, Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, Paris, France.,HeKA, Inria, Paris, France
| | - Christelle Lorenzato
- Oncology Biostatistics, Biostatistics and Programming Department, Sanofi R&D, Vitry-sur-Seine, France
| | - Moreno Ursino
- Inserm, Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, Paris, France.,HeKA, Inria, Paris, France.,Unit of Clinical Epidemiology, AP-HP, CHU Robert Debré, Université de Paris, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, Paris, France
| | - Marie-Karelle Riviere
- Statistical Methodology Group, Biostatistics and Programming Department, Sanofi R&D, Chilly-Mazarin, France
| |
Collapse
|
16
|
Lee SM, Wages NA, Goodman KA, Lockhart AC. Designing Dose-Finding Phase I Clinical Trials: Top 10 Questions That Should Be Discussed With Your Statistician. JCO Precis Oncol 2021; 5:317-324. [PMID: 34151131 DOI: 10.1200/po.20.00379] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/08/2020] [Accepted: 12/21/2020] [Indexed: 01/22/2023] Open
Abstract
In recent years, the landscape in clinical trial development has changed to involve many molecularly targeted agents, immunotherapies, or radiotherapy, as a single agent or in combination. Given their different mechanisms of action and lengths of administration, these agents have different toxicity profiles, which has resulted in numerous challenges when applying traditional designs such as the 3 + 3 design in dose-finding clinical trials. Novel methods have been proposed to address these design challenges such as combinations of therapies or late-onset toxicities. However, their design and implementation require close collaboration between clinicians and statisticians to ensure that the appropriate design is selected to address the aims of the study and that the design assumptions are pertinent to the study drug. The goal of this paper is to provide guidelines for appropriate questions that should be considered early in the design stage to facilitate the interactions between clinical and statistical teams and to improve the design of dose-finding clinical trials for novel anticancer agents.
Collapse
Affiliation(s)
- Shing M Lee
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY
| | - Nolan A Wages
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - A Craig Lockhart
- Division of Medical Oncology, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL
| |
Collapse
|
17
|
Zhao D, Zhu J, Wang L. Bayesian interval-based oncology dose-finding design with repeated quasi-continuous toxicity model. Contemp Clin Trials 2021; 102:106265. [PMID: 33418097 DOI: 10.1016/j.cct.2021.106265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/20/2020] [Accepted: 12/28/2020] [Indexed: 11/25/2022]
Abstract
In oncology dose-finding clinical trials, the key to accurately estimating the maximum tolerated dose (MTD) is to use all data efficiently given small sample sizes. Currently, popular designs dichotomize adverse events of various types and grades that occur within the first treatment cycle into binary toxicity outcomes of dose-limiting toxicity (DLT) events. Such compression of toxicity data from multiple treatment cycles causes huge loss of information, often resulting in MTD estimation with large bias and variance. To improve this, a continuous endpoint (the total toxicity profile, TTP) was proposed to incorporate adverse event types and grades. The Bayesian Repeated Measures Design (RMD) was further developed by Yin et al. (2017) to account for the cumulative toxicity information from multiple treatment cycles. However, the existing RMD method selects the dose that minimizes the loss function based on point estimates, which may generate inconsistent results due to small sample sizes in phase I trials. To reduce the variability in dose escalation decision-making, we propose an improved repeated measures design with an interval-based decision rule that selects the dose with the highest posterior probability of falling in a pre-specified target toxicity interval. Through comprehensive simulations, we compared this proposed design with the existing RMD design, along with well-established DLT-based designs such as Continual Reassessment Method (CRM) and Bayesian Logistic Regression Model (BLRM). The results demonstrated that our proposed design outperforms all other designs in terms of accurately identifying the MTD and assigning fewer patients to sub-therapeutic or overly toxic doses.
Collapse
Affiliation(s)
- Dan Zhao
- Statistical and Quantitative Science, Data Science Institute, Takeda Pharmaceutical Co. Limited, Cambridge, MA 02139, USA
| | - Jian Zhu
- Servier Pharmaceuticals, Boston, MA 02210, USA
| | - Ling Wang
- Pfizer Inc, Cambridge, MA 02139, USA.
| |
Collapse
|
18
|
Diniz MA, Kim S, Tighiouart M. A Bayesian Adaptive Design in Cancer Phase I Trials Using Dose Combinations with Ordinal Toxicity Grades. STATS 2020; 3:221-238. [PMID: 33073179 PMCID: PMC7561046 DOI: 10.3390/stats3030017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We propose a Bayesian adaptive design for early phase drug combination cancer trials incorporating ordinal grade of toxicities. Parametric models are used to describe the relationship between the dose combinations and the probabilities of the ordinal toxicities under the proportional odds assumption. Trial design proceeds by treating cohorts of two patients simultaneously receiving different dose combinations. Specifically, at each stage of the trial, we seek the dose of one agent by minimizing the Bayes risk with respect to a loss function given the current dose of the other agent. We consider two types of loss functions corresponding to the Continual Reassessment Method (CRM) and Escalation with Overdose Control (EWOC). At the end of the trial, we estimate the MTD curve as a function of Bayes estimates of the model parameters. We evaluate design operating characteristics in terms of safety of the trial and percent of dose recommendation at dose combination neighborhoods around the true MTD by comparing this design to the one that uses a binary indicator of DLT. The methodology is further adapted to the case of a pre-specified discrete set of dose combinations.
Collapse
|
19
|
Rosner GL. Bayesian Methods in Regulatory Science. Stat Biopharm Res 2019; 12:130-136. [PMID: 32489520 PMCID: PMC7265656 DOI: 10.1080/19466315.2019.1668843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 08/22/2019] [Accepted: 09/08/2019] [Indexed: 10/26/2022]
Abstract
Regulatory science comprises the tools, standards, and approaches that regulators use to assess safety, efficacy, quality, and performance of drugs and medical devices. A major focus of regulatory science is the design and analysis of clinical trials. Clinical trials are an essential part of clinical research programs that aim to improve therapies and reduce the burden of disease. These clinical experiments help us learn about what works clinically and what does not work. The results of clinical trials support therapeutic and policy decisions. When designing clinical trials, investigators make many decisions regarding various aspects of how they will carry out the study, such as the primary objective of the study, primary and secondary endpoints, methods of analysis, sample size, etc. This paper provides a brief review of the clinical development of new treatments and argues for the use of Bayesian methods and decision theory in clinical research.
Collapse
Affiliation(s)
- Gary L Rosner
- Division of Oncology Biostatistics & Bioinformatics, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore MD 21205
| |
Collapse
|
20
|
Lee SM, Ursino M, Cheung YK, Zohar S. Dose-finding designs for cumulative toxicities using multiple constraints. Biostatistics 2019; 20:17-29. [PMID: 29140414 PMCID: PMC6296314 DOI: 10.1093/biostatistics/kxx059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 10/18/2017] [Indexed: 11/14/2022] Open
Abstract
This article addresses the concern regarding late-onset dose-limiting toxicities (DLT), moderate toxicities below the threshold of a DLT and cumulative toxicities that may lead to a DLT, which are mostly disregarded or handled in an ad hoc manner when determining the maximum tolerated dose (MTD) in dose-finding cancer clinical trials. An extension of the Time-to-Event Continual Reassessment Method (TITE-CRM) which allows for the specification of toxicity constraints on both DLT and moderate toxicities, and can account for partial information is proposed. The method is illustrated in the context of an Erlotinib dose-finding trial with low DLT rates, but a significant number of moderate toxicities leading to treatment discontinuation in later cycles. Based on simulations, our method performs well at selecting the dose level that satisfies both the DLT and moderate-toxicity constraints. Moreover, it has similar probability of correct selection compared to the TITE-CRM when the true MTD based on DLT only and the true MTD based on grade 2 or higher toxicities alone coincide, but reduces the probability of recommending a dose above the MTD.
Collapse
Affiliation(s)
- Shing M Lee
- Department of Biostatistics, Mailman School of Public Health, Columbia University, 722 W. 168th St, New York, NY, USA
| | - Moreno Ursino
- INSERM, UMRS 1138, Team 22, CRC, University Paris 5, University Paris 6, Paris, France
| | - Ying Kuen Cheung
- Department of Biostatistics, Mailman School of Public Health, Columbia University, 722 W. 168th St, New York, NY, USA
| | - Sarah Zohar
- INSERM, UMRS 1138, Team 22, CRC, University Paris 5, University Paris 6, Paris, France
| |
Collapse
|
21
|
Du Y, Yin J, Sargent DJ, Mandrekar SJ. An adaptive multi-stage phase I dose-finding design incorporating continuous efficacy and toxicity data from multiple treatment cycles. J Biopharm Stat 2018; 29:271-286. [PMID: 30403559 DOI: 10.1080/10543406.2018.1535497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Phase I designs traditionally use the dose-limiting toxicity (DLT), a binary endpoint from the first treatment cycle, to identify the maximum-tolerated dose (MTD) assuming a monotonically increasing relationship between dose and efficacy. In this article, we establish a general framework for a multi-stage adaptive design where we jointly model a continuous efficacy outcome and continuous/quasi-continuous toxicity endpoints from multiple treatment cycles. The normalized Total Toxicity Profile (nTTP) is used as an illustration for quasi-continuous toxicity endpoints, and we replace DLT with nTTP to take into account multiple grades and types of toxicities. In addition, the proposed design accommodates non-monotone dose-efficacy relationships, and longitudinal toxicity data in effort to capture the adverse events from multiple cycles. Stage 1 of our design uses toxicity data to perform dose-escalation and identify a set of initially allowable (safe) doses; stage 2 of our design incorporates an efficacy outcome to update the set of allowable doses for each new cohort and randomizes the new cohort of patients to the allowable doses with emphasis towards those with higher predicted efficacy. Stage 3 uses all data from all treated patients at the end of the trial to make final recommendations. Simulations showed that the design had a high probability of making the correct dose selection and good overdose control across various dose-efficacy and dose-toxicity scenarios. In addition, the proposed design allows for early termination when all doses are too toxic. To our best knowledge, the proposed dual-endpoint dose-finding design is the first such study to incorporate multiple cycles of toxicities and a continuous efficacy outcome.
Collapse
Affiliation(s)
- Yu Du
- a Department of Biostatistics , Johns Hopkins University , Baltimore , MD , USA
| | - Jun Yin
- b Cancer Center Statistics , Mayo Clinic , Rochester , MN , USA
| | | | | |
Collapse
|
22
|
Ezzalfani M, Burzykowski T, Paoletti X. Joint modelling of a binary and a continuous outcome measured at two cycles to determine the optimal dose. J R Stat Soc Ser C Appl Stat 2018. [DOI: 10.1111/rssc.12305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Monia Ezzalfani
- Institut Curie and Institut National de la Recherche Médicale Paris
- Centre Léon Bérard Lyon France
| | | | - Xavier Paoletti
- Université, Paris‐Sud and Université Paris‐Saclay Villejuif France
| |
Collapse
|
23
|
Lin R. Bayesian optimal interval design with multiple toxicity constraints. Biometrics 2018; 74:1320-1330. [DOI: 10.1111/biom.12912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 04/01/2018] [Accepted: 04/01/2018] [Indexed: 11/27/2022]
Affiliation(s)
- Ruitao Lin
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexas 77030U.S.A
- Key Laboratory for Applied Statistics of MOENortheast Normal UniversityChangchunJilinChina
| |
Collapse
|
24
|
Muenz DG, Braun TM, Taylor JM. Modeling adverse event counts in phase I clinical trials of a cytotoxic agent. Clin Trials 2018; 15:386-397. [PMID: 29779418 DOI: 10.1177/1740774518772309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background/Aims The goal of phase I clinical trials for cytotoxic agents is to find the maximum dose with an acceptable risk of severe toxicity. The most common designs for these dose-finding trials use a binary outcome indicating whether a patient had a dose-limiting toxicity. However, a patient may experience multiple toxicities, with each toxicity assigned an ordinal severity score. The binary response is then obtained by dichotomizing a patient's richer set of data. We contribute to the growing literature on new models to exploit this richer toxicity data, with the goal of improving the efficiency in estimating the maximum tolerated dose. Methods We develop three new, related models that make use of the total number of dose-limiting and low-level toxicities a patient experiences. We use these models to estimate the probability of having at least one dose-limiting toxicity as a function of dose. In a simulation study, we evaluate how often our models select the true maximum tolerated dose, and we compare our models with the continual reassessment method, which uses binary data. Results Across a variety of simulation settings, we find that our models compare well against the continual reassessment method in terms of selecting the true optimal dose. In particular, one of our models which uses dose-limiting and low-level toxicity counts beats or ties the other models, including the continual reassessment method, in all scenarios except the one in which the true optimal dose is the highest dose available. We also find that our models, when not selecting the true optimal dose, tend to err by picking lower, safer doses, while the continual reassessment method errs more toward toxic doses. Conclusion Using dose-limiting and low-level toxicity counts, which are easily obtained from data already routinely collected, is a promising way to improve the efficiency in finding the true maximum tolerated dose in phase I trials.
Collapse
Affiliation(s)
- Daniel G Muenz
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Thomas M Braun
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jeremy Mg Taylor
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
25
|
Mu R, Yuan Y, Xu J, Mandrekar SJ, Yin J. gBOIN: a unified model-assisted phase I trial design accounting for toxicity grades, and binary or continuous end points. J R Stat Soc Ser C Appl Stat 2018. [DOI: 10.1111/rssc.12263] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Rongji Mu
- East China Normal University; Shanghai People's Republic of China
- University of Texas MD Anderson Cancer Center; Houston USA
| | - Ying Yuan
- University of Texas MD Anderson Cancer Center; Houston USA
| | - Jin Xu
- East China Normal University; Shanghai People's Republic of China
| | | | | |
Collapse
|
26
|
Yin J, Paoletti X, Sargent DJ, Mandrekar SJ. Repeated measures dose-finding design with time-trend detection in the presence of correlated toxicity data. Clin Trials 2017; 14:611-620. [PMID: 28764555 DOI: 10.1177/1740774517723829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Phase I trials are designed to determine the safety, tolerability, and recommended phase 2 dose of therapeutic agents for subsequent testing. The dose-finding paradigm has thus traditionally focused on identifying the maximum tolerable dose of an agent or combination therapy under the assumption that there is a non-decreasing relationship between dose-toxicity and dose-efficacy. The dose is typically determined based on the probability of severe toxicity observed during the first treatment cycle. A novel endpoint, the total toxicity profile, was previously developed to account for the multiple toxicity types and grades experienced in the first cycle. More recently, this was extended to a repeated measures design based on the total toxicity profile to account for longitudinal toxicities over multiple treatment cycles in the absence of within-patient correlation. METHODS In this work, we propose to extend the design in the presence of within-patient correlation. Furthermore, we provide a framework to detect a toxicity time trend (toxicity increasing, decreasing, or stable) over multiple treatment cycles. We utilize a linear mixed model in the Bayesian framework, with the addition of Bayesian risk functions for decision-making in dose assignment. RESULTS The performance of this design was evaluated using simulation studies and real data from a phase I trial. We demonstrated that using available toxicity data from all cycles of treatment improves the accuracy of maximum tolerated dose identification and allows for the detection of a time trend. The performance is consistent regardless of the strength of the within-patient correlation. In addition, the use of a quasi-continuous total toxicity profile score significantly increased the power to detect time trends compared to when binary data only were used. CONCLUSION The increased interest in molecularly targeted agents and immunotherapies in oncology necessitates innovative phase I study designs. Our proposed framework provides a tool to tackle some of the challenges presented by these novel agents, specifically through the ability to understand patterns of toxicity over time, which is important in the cases of cumulative or late toxicities.
Collapse
Affiliation(s)
- Jun Yin
- 1 Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Xavier Paoletti
- 2 Biostatistics and Epidemiology Department, INSERM CESP, OncoStat, Institut Gustave Roussy, Villejuif, France
| | - Daniel J Sargent
- 1 Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
27
|
Yin J, Shen S. Challenges and Innovations in Phase I Dose-Finding Designs for Molecularly Targeted Agents and Cancer Immunotherapies. JOURNAL OF BIOMETRICS & BIOSTATISTICS 2017; 7. [PMID: 28616356 PMCID: PMC5467542 DOI: 10.4172/2155-6180.1000324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Phase I oncology trials are designed to identify a safe dose with an acceptable toxicity profile. In traditional phase I dose-finding design, the dose is typically determined based on the probability of severe toxicity observed during the first treatment cycle. The recent development of molecularly targeted agents and cancer immunotherapies call for new innovations in phase I designs, because of prolonged treatment cycles often involved. Various phase I designs using toxicity and efficacy endpoints from multiple treatment cycles have been developed for these new treatment agents. Here, we will review the novel endpoints and designs for the phase I oncology clinical trials.
Collapse
Affiliation(s)
- Jun Yin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Shihao Shen
- Department of Microbiology, Immunology, & Molecular Genetics, UCLA, Los Angeles, CA 90024, USA
| |
Collapse
|
28
|
Adaptive Estimation of Personalized Maximum Tolerated Dose in Cancer Phase I Clinical Trials Based on All Toxicities and Individual Genomic Profile. PLoS One 2017; 12:e0170187. [PMID: 28125617 PMCID: PMC5268707 DOI: 10.1371/journal.pone.0170187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/30/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Many biomarkers have been shown to be associated with the efficacy of cancer therapy. Estimation of personalized maximum tolerated doses (pMTDs) is a critical step toward personalized medicine, which aims to maximize the therapeutic effect of a treatment for individual patients. In this study, we have established a Bayesian adaptive Phase I design which can estimate pMTDs by utilizing patient biomarkers that can predict susceptibility to specific adverse events and response as covariates. METHODS Based on a cutting-edge cancer Phase I clinical trial design called escalation with overdose control using normalized equivalent toxicity score (EWOC-NETS), which fully utilizes all toxicities, we propose new models to incorporate patient biomarker information in the estimation of pMTDs for novel cancer therapeutic agents. The methodology is fully elaborated and the design operating characteristics are evaluated with extensive simulations. RESULTS Simulation studies demonstrate that the utilization of biomarkers in EWOC-NETS can estimate pMTDs while maintaining the original merits of this Phase I trial design, such as ethical constraint of overdose control and full utilization of all toxicity information, to improve the accuracy and efficiency of the pMTD estimation. CONCLUSIONS Our novel cancer Phase I designs with inclusion of covariate(s) in the EWOC-NETS model are useful to estimate a personalized MTD and have substantial potential to improve the therapeutic effect of drug treatment.
Collapse
|
29
|
Yin J, Qin R, Ezzalfani M, Sargent DJ, Mandrekar SJ. A Bayesian dose-finding design incorporating toxicity data from multiple treatment cycles. Stat Med 2016; 36:67-80. [PMID: 27633877 DOI: 10.1002/sim.7134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 08/16/2016] [Accepted: 08/25/2016] [Indexed: 01/05/2023]
Abstract
Phase I oncology trials are designed to identify a safe dose with an acceptable toxicity profile. The dose is typically determined based on the probability of severe toxicity observed during the first treatment cycle, although patients continue to receive treatment for multiple cycles. In addition, the toxicity data from multiple types and grades are typically summarized into a single binary outcome of dose-limiting toxicity. A novel endpoint, the total toxicity profile, was previously developed to account for the multiple toxicity types and grades. In this work, we propose to account for longitudinal repeated measures of total toxicity profile over multiple treatment cycles, accounting for cumulative toxicity during dosing-finding. A linear mixed model was utilized in the Bayesian framework, with addition of Bayesian risk functions for decision-making in dose assignment. The performance of this design is evaluated using simulation studies and compared with the previously proposed quasi-likelihood continual reassessment method (QLCRM) design. Twelve clinical scenarios incorporating four different locations of maximum tolerated dose and three different time trends (decreasing, increasing, and no effect) were investigated. The proposed repeated measures design was comparable with the QLCRM when only cycle 1 data were utilized in dose-finding; however, it demonstrated an improvement over the QLCRM when data from multiple cycles were used across all scenarios. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jun Yin
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| | - Rui Qin
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| | - Monia Ezzalfani
- Biostatistics Department, Institut Gustave-Roussy, Villejuif, France
| | - Daniel J Sargent
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| | - Sumithra J Mandrekar
- Department of Health Sciences Research, Mayo Clinic, 55905, Rochester, MN, U.S.A
| |
Collapse
|
30
|
|
31
|
Tseng WW, Zhou S, To CA, Thall PF, Lazar AJ, Pollock RE, Lin PP, Cormier JN, Lewis VO, Feig BW, Hunt KK, Ballo MT, Patel S, Pisters PWT. Phase 1 adaptive dose-finding study of neoadjuvant gemcitabine combined with radiation therapy for patients with high-risk extremity and trunk soft tissue sarcoma. Cancer 2015; 121:3659-67. [PMID: 26177983 DOI: 10.1002/cncr.29544] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/23/2015] [Accepted: 05/29/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND This study was performed to determine the maximum tolerated dose (MTD) of gemcitabine given concurrently with preoperative, fixed-dose external-beam radiation therapy (EBRT) for patients with resectable, high-risk extremity and trunk soft tissue sarcoma (STS). METHODS Gemcitabine was administered on days 1, 8, 22, 29, 43, and 50 with EBRT (50 Gy in 25 fractions over 5 weeks). The gemcitabine MTD was determined with a toxicity severity weight method (TSWM) incorporating 6 toxicity types. The TSWM is a Bayesian procedure that choses each cohort's dose to have a posterior mean total toxicity burden closest to a predetermined clinician-defined target. Clinicopathologic and outcome data were also collected. RESULTS Thirty-six patients completed the study. According to the TSWM, the gemcitabine MTD was 700 mg/m(2). At this dose level, 4 patients (24%) experienced grade 4 toxicity; no toxicity-related deaths occurred. All tumors were resected with microscopically negative margins. Pathologic responses of >90% tumor necrosis were achieved in 17 patients (47%); 14 (39%) had complete responses. With a median follow-up of 6.2 years, the 5-year locoregional recurrence-free survival, distant metastasis-free survival, and overall survival rates were 85%, 80%, and 86%, respectively. CONCLUSIONS The TSWM combines data from qualitatively different toxicities and can be used to determine the MTD for a drug given as part of a multimodality treatment. Neoadjuvant gemcitabine plus radiation therapy is feasible and safe in patients with high-risk extremity and trunk STS. Major pathologic responses can be achieved, and after complete resection, long-term clinical outcomes are encouraging.
Collapse
Affiliation(s)
- William W Tseng
- Section of Surgical Oncology, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California.,Sarcoma Program, Hoag Family Cancer Institute and Hoag Memorial Hospital Presbyterian, Newport Beach, California
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christina A To
- Department of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Peter F Thall
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raphael E Pollock
- Division of Surgical Oncology, James Comprehensive Cancer Center, Ohio State University, Columbus, Ohio
| | - Patrick P Lin
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Janice N Cormier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Valerae O Lewis
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barry W Feig
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kelly K Hunt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew T Ballo
- Department of Radiation Oncology, University of Tennessee Health Sciences Center, Memphis, Tennessee
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
32
|
Paoletti X, Doussau A, Ezzalfani M, Rizzo E, Thiébaut R. Dose finding with longitudinal data: simpler models, richer outcomes. Stat Med 2015; 34:2983-98. [PMID: 26109523 DOI: 10.1002/sim.6552] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 05/17/2015] [Indexed: 01/29/2023]
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 at cycle 1, although toxicity is repeatedly measured over cycles on an ordinal scale. Recently, a proportional odds mixed-effect model for ordinal outcomes has been proposed to (i) identify the optimal dose accounting for repeated events and (ii) to provide some framework to explore time trend. We compare this approach to a method based on repeated binary variables and to a method based on an under-parameterized model of the dose-time toxicity relationship. We show that repeated binary and ordinal outcomes both improve the accuracy of dose-finding trials in the same proportion; ordinal outcomes are, however, superior to detect time trend even in the presence of nonproportional odds models. Moreover, less parameterized models led to the best operating characteristics. These approaches are illustrated on two dose-finding phase I trials. Integration of repeated measurements is appealing in phase I dose-finding trials.
Collapse
Affiliation(s)
| | - Adélaïde Doussau
- INSERM U900, Institut Curie, Paris, France
- Centre de recherche INSERM U897, & INRIA SISTM & Université de Bordeaux, ISPED, France
| | | | - Elisa Rizzo
- European Organization for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Rodolphe Thiébaut
- Centre de recherche INSERM U897, & INRIA SISTM & Université de Bordeaux, ISPED, France
| |
Collapse
|
33
|
Mussai FJ, Yap C, Mitchell C, Kearns P. Challenges of clinical trial design for targeted agents against pediatric leukemias. Front Oncol 2015; 4:374. [PMID: 25610810 PMCID: PMC4285052 DOI: 10.3389/fonc.2014.00374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 12/15/2014] [Indexed: 12/16/2022] Open
Abstract
The past 40 years have seen significant improvements in both event-free and overall survival for children with acute lymphoblastic and acute myeloid leukemia (ALL and AML, respectively). Serial national and international clinical trials have optimized the use of conventional chemotherapeutic drugs and, along with improvements in supportive care that have enabled the delivery of more intensive regimens, have been responsible for the major improvements in patient outcome seen over the past few decades. However, the benefits of dose intensification have likely now been maximized, and over the same period, the identification of new cytotoxic drugs has been limited. Therefore, challenges remain if survival is to be improved further. In pediatric ALL, 5-year-survival rates of over 85% have been achieved with risk-stratified therapy, but a notable minority of patients will still not be cured. In pediatric AML, different challenges remain. A slower improvement in overall survival has taken place in this patient population. Despite the obvious morphological heterogeneity of AML blasts, biological stratification is comparatively limited, and translation into risk-stratified therapeutic approaches has only best characterized by the use of retinoic acid for t(15;17)-positive AML. Even where prognostic markers have been identified, limited therapeutic options or multi-drug resistance of AML blasts has limited the impact on patient benefit. For both, the acute morbidities of current treatment remain significant and may be life-threatening alone. In addition, the Childhood Cancer Survivor Study (CCSS) highlighted many leukemia survivors develop one or more chronic medical conditions attributable to treatment (1, 2). As the biology of leukemogenesis has become better understood, key molecules and intracellular pathways have been identified that offer the possibility of targeting directly the leukemia cells while sparing normal cells. Consequently, there is now a drive to develop novel leukemia-specific or "targeted" therapies. These new classes of drugs will have mechanisms of action, toxicities, and therapeutic indices quite different from conventional cytotoxic drugs previously encountered, thus rendering current clinical trial methodologies inappropriate. Clinical trial methods will need to be adapted to accommodate these features of these new classes of drugs. This review will address the challenges and some of the techniques for developing clinical trials for targeted therapies.
Collapse
Affiliation(s)
| | - Christina Yap
- Cancer Research UK Clinical Trials Unit, School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | - Christopher Mitchell
- Department of Paediatric Oncology, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Pamela Kearns
- Cancer Research UK Clinical Trials Unit, School of Cancer Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
34
|
Wang Y, Ivanova A. Dose finding with continuous outcome in phase I oncology trials. Pharm Stat 2014; 14:102-7. [PMID: 25408518 DOI: 10.1002/pst.1662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 10/07/2014] [Accepted: 10/28/2014] [Indexed: 11/10/2022]
Abstract
The goal of a phase I clinical trial in oncology is to find a dose with acceptable dose-limiting toxicity rate. Often, when a cytostatic drug is investigated or when the maximum tolerated dose is defined using a toxicity score, the main endpoint in a phase I trial is continuous. We propose a new method to use in a dose-finding trial with continuous endpoints. The new method selects the right dose on par with other methods and provides more flexibility in assigning patients to doses in the course of the trial when the rate of accrual is fast relative to the follow-up time.
Collapse
Affiliation(s)
- Yunfei Wang
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7420, USA
| | | |
Collapse
|
35
|
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
|
36
|
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
|