1
|
Fang J, Zhang N, Wang W, Yin G. CFO: Calibration-Free Odds Bayesian Designs for Dose Finding in Clinical Trials. JCO Clin Cancer Inform 2025; 9:e2400184. [PMID: 39889256 DOI: 10.1200/cci-24-00184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/17/2024] [Accepted: 12/10/2024] [Indexed: 02/02/2025] Open
Abstract
PURPOSE Calibration-free odds type (CFO-type) designs have been demonstrated to be robust, model-free, and practically useful, which have become the state-of-the-art approach for dose finding. However, a key challenge for implementing such designs is a lack of accessible tools. We develop a user-friendly R package and Shiny web-based software to facilitate easy implementation of CFO-type designs. Moreover, we incorporate randomization into the CFO framework. METHODS We created the R package CFO and leveraged R Shiny to build an interactive web application, CFO suite, for implementing CFO-type designs. We introduce the randomized CFO (rCFO) design by integrating the exploration-exploitation mechanism into the CFO framework. RESULTS The CFO package and CFO suite encompass various variants tailored to different clinical settings. Beyond the fundamental CFO design, these include the two-dimensional CFO (2dCFO) for drug-combination trials, accumulative CFO (aCFO) for accommodating all dose information, rCFO for integrating exploration-exploitation via randomization, time-to-event CFO (TITE-CFO), and fractional CFO (fCFO) for addressing late-onset toxicity. Using all information and addressing delayed toxicity outcomes, TITE-aCFO and fractional-aCFO are also included. The package provides functions for determining the subsequent cohort dose, selecting the maximum tolerated dose, and conducting simulations to evaluate performance, with results presented through textual and graphical outputs. CONCLUSION The CFO package and CFO suite provide comprehensive and flexible tools for implementing CFO-type designs in phase I clinical trials. This work is highly significant as it integrates all existing CFO-type designs to facilitate novel trial designs with enhanced performance. In addition, this promotes the spread of statistical methods using a user-friendly R package and Shiny software. It strengthens collaborations between biostatisticians and clinicians, further enhancing trial performance in terms of efficiency and accuracy.
Collapse
Affiliation(s)
- Jialu Fang
- School of Computing and Data Science, The University of Hong Kong, Hong Kong, China
| | - Ninghao Zhang
- School of Computing and Data Science, The University of Hong Kong, Hong Kong, China
| | - Wenliang Wang
- School of Computing and Data Science, The University of Hong Kong, Hong Kong, China
| | - Guosheng Yin
- School of Computing and Data Science, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
Msaouel P, Yu K, Yuan Y, Chen J, Yan X, Karki M, Duan F, Sheth RA, Rao P, Sircar K, Shah AY, Zurita AJ, Genovese G, Li M, Yeh CC, Dang M, Han G, Chu Y, Hallin M, Olson P, Yang R, Slavin D, Der-Torossian H, Chin CD, Tannir NM, Wang L, Gao J. Sitravatinib in combination with nivolumab plus ipilimumab in patients with advanced clear cell renal cell carcinoma: a phase 1 trial. Nat Commun 2025; 16:578. [PMID: 39794332 PMCID: PMC11724043 DOI: 10.1038/s41467-024-55642-8] [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] [Received: 08/20/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
We conducted a phase I trial to determine the optimal dose of triplet therapy with the tyrosine kinase inhibitor sitravatinib plus nivolumab plus ipilimumab in 22 previously untreated patients with advanced clear cell renal cell carcinoma. The primary endpoint was safety. Secondary endpoints were objective response rate (ORR), disease control rate (DCR), duration of response (DOR), progression-free survival (PFS), overall survival (OS), 1-year survival probability, and sitravatinib pharmacokinetics. Sitravatinib dose of 35 mg daily plus nivolumab 3 mg/kg and ipilimumab 1 mg/kg resulted in high frequency of immune-related adverse events. Subsequent dose reduction of ipilimumab to 0.7 mg/kg allowed safe escalation of sitravatinib up to 100 mg daily. Overall, the triplet combination achieved ORR 45.5%, DCR 86.4%, median PFS 14.5 months, and 1-year survival 80.8%. Median OS and DOR were not reached. Sitravatinib exposure increased dose-dependently. Single-cell RNA-seq of longitudinally collected tumor biopsies from 12 patients identified a tumor cell-specific epithelial-mesenchymal transition-like program associated with treatment resistance and poor outcomes. Treatment resistance was characterized by a transition from cytotoxic to exhausted T cell state and enrichment for M2-like myeloid cells. The observed hypothesis-generating changes in gene expression dynamics and cellular states may help inform future strategies to optimize immunotherapy efficacy. Clinical Trials.gov identifier: NCT04518046.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Kai Yu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianfeng Chen
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Xinmiao Yan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Menuka Karki
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Fei Duan
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Rahul A Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priya Rao
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanishka Sircar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amishi Y Shah
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amado J Zurita
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Min Li
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Chih-Chen Yeh
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Minghao Dang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Guangchun Han
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Yanshuo Chu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Max Hallin
- Mirati Therapeutics, Inc, San Diego, CA, USA
| | - Peter Olson
- Mirati Therapeutics, Inc, San Diego, CA, USA
| | - Rui Yang
- Mirati Therapeutics, Inc, San Diego, CA, USA
| | | | | | | | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA.
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Sadachi R, Sato H, Fujiwara T, Hirakawa A. Enhancement of Bayesian optimal interval design by accounting for overdose and underdose errors trade-offs. J Biopharm Stat 2025; 35:1-20. [PMID: 37966109 DOI: 10.1080/10543406.2023.2275766] [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: 02/28/2023] [Accepted: 10/22/2023] [Indexed: 11/16/2023]
Abstract
Model-assisted designs, a new class of dose-finding designs for determining the maximum tolerated dose (MTD), model only the dose-limiting toxicity (DLT) data observed at the current dose based on a simple binomial model and offer the boundaries of DLT for the determination of dose escalation, retention, or de-escalation before beginning the trials. The boundaries for dose-escalation and de-escalation decisions are relevant to the operating characteristics of the design. The well-known model-assisted design, Bayesian Optimal Interval (BOIN), selects these boundaries to minimize the probability of incorrect decisions at each dose allocation but does not distinguish between overdose and underdose allocations caused by incorrect decisions when calculating the probability of incorrect decisions. Distinguishing between overdose and underdose based on the decision error in the BOIN design is expected to increase the accuracy of MTD determination. In this study, we extended the BOIN design to account for the decision probabilities of incorrect overdose and underdose allocations separately. To minimize the two probabilities simultaneously, we propose utilizing multiple objective optimizations and formulating an approach for determining the boundaries for dose escalation and de-escalation. Comprehensive simulation studies using fixed and randomly generated scenarios of DLT probability demonstrated that the proposed method is superior or comparable to existing interval designs, along with notably better operating characteristics of the proposed method.
Collapse
Affiliation(s)
- Ryo Sadachi
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo, Japan
- Department of Global Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Sato
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeo Fujiwara
- Department of Global Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
4
|
Willmann J, Balermpas P, Rimner A, Appelt AL, Vasquez Osorio EM, Rønde HS, Day M, Embring A, Gabryś D, Guren MG, Hoskin P, Massaccesi M, Mayo C, Murray L, Nieder C, Guckenberger M, Andratschke N. Ongoing prospective studies on reirradiation: A systematic review of a clinical trials database. Radiother Oncol 2025; 202:110624. [PMID: 39532233 DOI: 10.1016/j.radonc.2024.110624] [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: 09/27/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Reirradiation has gained increasing interest, as advances in systemic therapy increase the survival of patients with cancer, and modern radiation techniques allow more precise treatments. However, high-quality prospective evidence on the safety and efficacy of reirradiation to guide clinical practice remains scarce. This systematic review evaluates ongoing prospective studies on reirradiation to identify research gaps and priorities. METHODS A systematic review of ClinicalTrials.gov was conducted on July 11, 2024, using search terms related to reirradiation. Inclusion criteria were prospective studies that were "recruiting," "not yet recruiting," or "active, not recruiting." Studies with published results, retrospective, and in-silico studies were excluded. The review followed PRISMA 2020 guidelines and recommendations for systematic searches of clinical trial registries. RESULTS Among 1026 identified studies, 307 were screened, 99 were included. Fourty (40%) focused on central nervous system (CNS), 23 (23%) head and neck, and 17 (17%) on pelvic reirradiation. Most studies (90%) were interventional, with 32 (32%) phase II and 4 (4%) phase III trials. Sixteen trials were randomized (RCTs), including the 4 phase III trials for recurrent glioblastoma, rectal and nasopharyngeal cancer. Ten dose escalation trials focus on recurrent prostate, rectal, and non-small cell lung cancer as well as glioma. Modern high-precision radiotherapy techniques were frequently used, with 21 (21%) studies using stereotactic radiotherapy and 17 (17%) using particle therapy. Combinations with systemic therapies were investigated in 41 (41%) studies. CONCLUSION Ongoing studies most frequently focus on CNS, head and neck, and pelvic reirradiation. There remains a critical need for RCTs, in particular for lung, breast, and gynecological cancers. Dose escalation trials, application of precision radiation techniques and combinations with modern systemic therapy may help define the optimal multimodality treatment schedules.
Collapse
Affiliation(s)
- Jonas Willmann
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Panagiotis Balermpas
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andreas Rimner
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg, Germany
| | - Ane L Appelt
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Eliana Maria Vasquez Osorio
- Division of Cancer Sciences, The University of Manchester & The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Heidi S Rønde
- Danish Center for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Madalyne Day
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anna Embring
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Dorota Gabryś
- Department of Radiation Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Marianne G Guren
- Department of Oncology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peter Hoskin
- Mount Vernon Cancer Centre, London, United Kingdom; Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Mariangela Massaccesi
- Department of Radiology, Radiation Oncology and Hematology, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Charles Mayo
- University of Michigan, Ann Arbor, United States of America
| | - Louise Murray
- University of Leeds and Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Carsten Nieder
- Department of Oncology and Palliative Medicine, Nordland Hospital Trust, Bodø, Norway
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Nicolaus Andratschke
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Chen X, Zhang J, Li B, Yan F. Determining doses for backfill cohorts based on patient-reported outcome. BMC Med Res Methodol 2024; 24:270. [PMID: 39516724 PMCID: PMC11546322 DOI: 10.1186/s12874-024-02398-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Incorporating backfill cohorts in phase I oncology trials is a recently developed strategy for dose optimization. However, the efficacy assessment window is long in general, causing a lag in identifying ineffective doses and more patients being backfilled to those doses. There is necessity to investigate how to use patient-reported outcomes (PRO) to determine doses for backfill cohorts. METHODS We propose a unified Bayesian design framework, called 'Backfill-QoL', to utilize patient-reported quality of life (QoL) data into phase I oncology trials with backfill cohorts, including methods for trial monitoring, algorithm for dose-finding, and criteria for dose selection. Simulation studies and sensitivity analyses are conducted to evaluate the proposed Backfill-QoL design. RESULTS The simulation studies demonstrate that the Backfill-QoL design is more efficient than traditional dose-expansion strategy, and fewer patients would be allocated to doses with unacceptable QoL profiles. A user-friendly Windows desktop application is developed and freely available for implementing the proposed design. CONCLUSIONS The Backfill-QoL design enables continuous monitoring of safety, efficacy and QoL outcomes, and the recommended phase II dose (RP2D) can be identified in a more patient-centered perspective.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Jingyi Zhang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Bosheng Li
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
6
|
Kojima M, Wende W, Zhao H. Tips for Accelerating BOIN Design. Ther Innov Regul Sci 2024; 58:1129-1137. [PMID: 39222294 DOI: 10.1007/s43441-024-00692-9] [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: 03/04/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
During discussions at the Data Science Roundtable meeting in Japan, there were instances where the adoption of the BOIN design was declined, attributed to the extension of study duration and increased sample size in comparison to the 3 + 3 design. We introduce an accelerated BOIN design aimed at completing a clinical phase I trial at a pace comparable to the 3 + 3 design. Additionally, we introduce how we could have applied the BOIN design within our company, which predominantly utilized the 3 + 3 design for most of its clinical oncology dose escalation trials. The accelerated BOIN design is adaptable by using efficiently designated stopping criterion for the existing BOIN framework. Our approach is to terminate the dose escalation study if the number of evaluable patients treated at the current dose reaches 6 and the decision is to stay at the current dose for the next cohort of patients. In addition, for lower dosage levels, considering a cohort size smaller than 3 may be feasible when there are no safety concerns from non-clinical studies. We demonstrate the accelerated BOIN design using a case study and subsequently evaluate the performance of our proposed design through a simulation study. In the simulation study, the average difference in the percentage of correct MTD selection between the accelerated BOIN design and the standard BOIN design was - 2.43%, the average study duration and the average sample size of the accelerated BOIN design was reduced by 14.8 months and 9.22, respectively, compared with the standard BOIN design.
Collapse
Affiliation(s)
- Masahiro Kojima
- Biometrics Department, Research Division, Kyowa Kirin Co., Ltd. Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo, 100-004, Japan.
| | - Wu Wende
- Biometrics Department, Development Division, Kyowa Kirin, Inc, Princeton, NJ, USA
| | - Henry Zhao
- Biometrics Department, Development Division, Kyowa Kirin, Inc, Princeton, NJ, USA
| |
Collapse
|
7
|
Guo J, Lu M, Wan I, Wang Y, Han L, Zang Y. T3 + 3: 3 + 3 Design With Delayed Outcomes. Pharm Stat 2024; 23:959-970. [PMID: 38923150 PMCID: PMC11602947 DOI: 10.1002/pst.2414] [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] [Received: 02/14/2024] [Revised: 05/22/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
Delayed outcome is common in phase I oncology clinical trials. It causes logistic difficulty, wastes resources, and prolongs the trial duration. This article investigates this issue and proposes the time-to-event 3 + 3 (T3 + 3) design, which utilizes the actual follow-up time for at-risk patients with pending toxicity outcomes. The T3 + 3 design allows continuous accrual without unnecessary trial suspension and is costless and implementable with pretabulated dose decision rules. Besides, the T3 + 3 design uses the isotonic regression to estimate the toxicity rates across dose levels and therefore can accommodate for any targeted toxicity rate for maximum tolerated dose (MTD). It dramatically facilitates the trial preparation and conduct without intensive computation and statistical consultation. The extension to other algorithm-based phase I dose-finding designs (e.g., i3 + 3 design) is also studied. Comprehensive computer simulation studies are conducted to investigate the performance of the T3 + 3 design under various dose-toxicity scenarios. The results confirm that the T3 + 3 design substantially shortens the trial duration compared with the conventional 3 + 3 design and yields much higher accuracy in MTD identification than the rolling six design. In summary, the T3 + 3 design addresses the delayed outcome issue while keeping the desirable features of the 3 + 3 design, such as simplicity, transparency, and costless implementation. It has great potential to accelerate early-phase drug development.
Collapse
Affiliation(s)
- Jiaying Guo
- Department of Biostatistics and Health Data ScienceIndiana UniversityIndianapolisIndianaUSA
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Mengyi Lu
- Department of BiostatisticsNanjing Medical UniversityNanjingChina
| | - Isabella Wan
- Faculty of Arts and ScienceUniversity of TorontoTorontoCanada
| | | | - Leng Han
- Department of Biostatistics and Health Data ScienceIndiana UniversityIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana UniversityIndianapolisIndianaUSA
| | - Yong Zang
- Department of Biostatistics and Health Data ScienceIndiana UniversityIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
8
|
Sun H, Tu J. PKBOIN-12: A Bayesian Optimal Interval Phase I/II Design Incorporating Pharmacokinetics Outcomes to Find the Optimal Biological Dose. Pharm Stat 2024. [PMID: 39448544 DOI: 10.1002/pst.2444] [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: 04/03/2024] [Revised: 07/17/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024]
Abstract
Immunotherapies and targeted therapies have gained popularity due to their promising therapeutic effects across multiple treatment areas. The focus of early phase dose-finding clinical trials has shifted from finding the maximum tolerated dose (MTD) to identifying the optimal biological dose (OBD), which aims to balance the toxicity and efficacy outcomes, thus optimizing the risk-benefit trade-off. These trials often collect multiple pharmacokinetics (PK) outcomes to assess drug exposure, which has shown correlations with toxicity and efficacy outcomes but has not been utilized in the current dose-finding designs for OBD selection. Moreover, PK outcomes are usually available within days after initial treatment, much faster than toxicity and efficacy outcomes. To bridge this gap, we introduce the innovative model-assisted PKBOIN-12 design, which enhances BOIN12 by integrating PK information into both the dose-finding algorithm and the final OBD determination process. We further extend PKBOIN-12 to TITE-PKBOIN-12 to address the challenges of late-onset toxicity and efficacy outcomes. Simulation results demonstrate that PKBOIN-12 more effectively identifies the OBD and allocates a greater number of patients to it than BOIN12. Additionally, PKBOIN-12 decreases the probability of selecting inefficacious doses as the OBD by excluding those with low drug exposure. Comprehensive simulation studies and sensitivity analysis confirm the robustness of both PKBOIN-12 and TITE-PKBOIN-12 in various scenarios.
Collapse
Affiliation(s)
- Hao Sun
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Jieqi Tu
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, Illinois, USA
| |
Collapse
|
9
|
Zumsteg ZS, Sheth S, Jabbour SK, Patel KR, Kimple RJ, Williams TM, Xu-Welliver M, Torres-Saavedra PA, Monjazeb AM, Mayadev J, Finkelstein SE, Buatti JM, Patel SP, Lin SH. Challenges and opportunities for early phase clinical trials of novel drug-radiotherapy combinations: recommendations from NRG Oncology, the American Society for Radiation Oncology (ASTRO), the American College of Radiology (ACR), the Sarah Cannon Research Institute, and the American College of Radiation Oncology (ACRO). Lancet Oncol 2024; 25:e489-e500. [PMID: 39362260 PMCID: PMC11778933 DOI: 10.1016/s1470-2045(24)00264-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 10/05/2024]
Abstract
NRG Oncology's Developmental Therapeutics and Radiation Therapy Subcommittee assembled an interdisciplinary group of investigators to address barriers to successful early phase clinical trials of novel combination therapies involving radiation. This Policy Review elucidates some of the many challenges associated with study design for early phase trials combining radiotherapy with novel systemic agents, which are distinct from drug-drug combination development and are often overlooked. We also advocate for potential solutions that could mitigate or eliminate some of these barriers, providing examples of specific clinical trial designs that could help facilitate efficient and effective evaluation of novel drug-radiotherapy combinations.
Collapse
Affiliation(s)
- Zachary S Zumsteg
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Siddharth Sheth
- Division of Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Krishnan R Patel
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Randall J Kimple
- Department of Human Oncology, Univeristy of Wisconsin, Madison, WI, USA
| | | | - Meng Xu-Welliver
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Pedro A Torres-Saavedra
- Division of Cancer Treatment and Diagnosis, Biometric Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California, San Diego, CA, USA
| | - Jyoti Mayadev
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, CA, USA
| | - Steven E Finkelstein
- The US Oncology Network, Florida Cancer Affiliates, Panama City, FL, USA; Sarah Cannon Research Institute, Nashville, TN, USA; Associated Medical Professional of NY, US Urology Partners, Syracuse, NY, USA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Sandip P Patel
- Division of Medical Oncology, University of California, San Diego, CA, USA
| | - Steven H Lin
- Department of Thoracic Radiation Oncology, Division of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Li N, Zhou X, Yan D. Phase I clinical trial designs in oncology: A systematic literature review from 2020 to 2022. J Clin Transl Sci 2024; 8:e134. [PMID: 39345694 PMCID: PMC11428115 DOI: 10.1017/cts.2024.599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/10/2024] [Accepted: 07/19/2024] [Indexed: 10/01/2024] Open
Abstract
Background Phase I clinical trials aim to find the highest dose of a novel drug that may be administrated safely without having serious adverse effects. Model-based designs have recently become popular in dose-finding procedures. Our objective is to provide an overview of phase I clinical trials in oncology. Methods A retrospective analysis of phase I clinical trials in oncology was performed by using the PubMed database between January 1, 2020, and December 31, 2022. We extracted all papers with the inclusion of trials in oncology and kept only those in which dose escalation or/ and dose expansion were conducted. We also compared the study parameters, design parameters, and patient parameters between industry-sponsored studies and academia-sponsored research. Result Among the 1450 papers retrieved, 256 trials described phase I clinical trials in oncology. Overall, 71.1% of trials were done with a single study cohort, 56.64% of trials collected a group of at least 20 study volunteers, 55.1% were sponsored by industry, and 99.2% of trials had less than 10 patients who experienced DLTs.The traditional 3 + 3 (73.85%) was still the most prevailing method for the dose-escalation approach. More than 50% of the trials did not reach MTDs. Industry-sponsored study enrolled more patients in dose-escalation trials with benefits of continental cooperation. Compared to previous findings, the usage of model-based design increased to about 10%, and the percentage of traditional 3 + 3 design decreased to 74%. Conclusions Phase I traditional 3 + 3 designs perform well, but there is still room for development in novel model-based dose-escalation designs in clinical practice.
Collapse
Affiliation(s)
- Ning Li
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Xitong Zhou
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, USA
| | - Donglin Yan
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
11
|
Lu M, Yuan Y, Liu S. A Bayesian pharmacokinetics integrated phase I-II design to optimize dose-schedule regimes. Biostatistics 2024:kxae034. [PMID: 39275895 DOI: 10.1093/biostatistics/kxae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/16/2024] Open
Abstract
The schedule of administering a drug has profound impact on the toxicity and efficacy profiles of the drug through changing its pharmacokinetics (PK). PK is an innate and indispensable component of the dose-schedule optimization. Motivated by this, we propose a Bayesian PK integrated dose-schedule finding (PKIDS) design to identify the optimal dose-schedule regime by integrating PK, toxicity, and efficacy data. Based on the causal pathway that dose and schedule affect PK, which in turn affects efficacy and toxicity, we jointly model the three endpoints by first specifying a Bayesian hierarchical model for the marginal distribution of the longitudinal dose-concentration process. Conditional on the drug concentration in plasma, we jointly model toxicity and efficacy as a function of the concentration. We quantify the risk-benefit of regimes using utility-continuously updating the estimates of PK, toxicity, and efficacy based on interim data-and make adaptive decisions to assign new patients to appropriate dose-schedule regimes via adaptive randomization. The simulation study shows that the PKIDS design has desirable operating characteristics.
Collapse
Affiliation(s)
- Mengyi Lu
- Department of Biostatistics, Nanjing Medical University, Nanjing 211166, China
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
12
|
Deng Q, Zhu L, Weiss B, Aanur P, Gao L. Strategies for successful dose optimization in oncology drug development: a practical guide. J Biopharm Stat 2024:1-15. [PMID: 39127994 DOI: 10.1080/10543406.2024.2387364] [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: 09/28/2023] [Accepted: 07/27/2024] [Indexed: 08/12/2024]
Abstract
Dose optimization is a critical challenge in drug development. Historically, dose determination in oncology has followed a divergent path from other non-oncology therapeutic areas due to the unique characteristics and requirements in Oncology. However, with the emergence of new drug modalities and mechanisms of drugs in oncology, such as immune therapies, radiopharmaceuticals, targeted therapies, cytostatic agents, and others, the dose-response relationship for efficacy and toxicity could be vastly varied compared to the cytotoxic chemotherapies. The doses below the MTD may demonstrate similar efficacy to the MTD with an improved tolerability profile, resembling what is commonly observed in non-oncology treatments. Hence, alternate strategies for dose optimization are required for new modalities in oncology drug development. This paper delves into the historical evolution of dose finding methods from non-oncology to oncology, highlighting examples and summarizing the underlying drivers of change. Subsequently, a practical framework and guidance are provided to illustrate how dose optimization can be incorporated into various stages of the development program. We provide the following general recommendations: 1) The objective for phase I is to identify a dose range rather than a single MTD dose for subsequent development to better characterize the safety and tolerability profile within the dose range. 2) At least two doses separable by PK are recommended for dose optimization in phase II. 3) Ideally, dose optimization should be performed before launching the confirmatory study. Nevertheless, innovative designs such as seamless II/III design can be implemented for dose selection and may accelerate the drug development program.
Collapse
Affiliation(s)
- Qiqi Deng
- Biostatistics and Programming, Moderna Inc., Cambridge, MA, USA
| | - Lili Zhu
- Biostatistics and Programming, Moderna Inc., Cambridge, MA, USA
| | - Brendan Weiss
- Clinical Development Oncology, Moderna Inc., Cambridge, MA, USA
| | - Praveen Aanur
- Clinical Development Oncology, Moderna Inc., Cambridge, MA, USA
| | - Lei Gao
- Biostatistics and Programming, Moderna Inc., Cambridge, MA, USA
| |
Collapse
|
13
|
Jiang L, Yin Z, Yan F, Yuan Y. MC-Keyboard: A Practical Phase I Trial Design for Targeted Therapies and Immunotherapies Integrating Multiple-Grade Toxicities. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:159-167. [PMID: 39219992 PMCID: PMC11361345 DOI: 10.36401/jipo-23-35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 09/04/2024]
Abstract
In targeted therapies and immunotherapies, the occurrence of low-grade (e.g., grade 1-2) toxicities (LGT) is common, while dose-limiting toxicities (DLT) are relatively rare. As a result, conventional phase I trial designs, solely based on DLTs and disregarding milder toxicities, are problematic when evaluating these novel therapies. Methods: To address this issue, we propose a novel phase I design called a multiple-constraint keyboard (MC-Keyboard) that integrates multiple toxicity constraints, accounting for both DLT and LGT, for precise dose escalation and de-escalation, and identification of the maximum tolerated dose (MTD). As a model-assisted design, an important feature of MC-Keyboard is that its dose-escalation or de-escalation rule can be pretabulated and incorporated into the trial protocol before the initiation of the trial, greatly simplifying its implementation. Results: The simulation study showed that the MC-Keyboard had high accuracy in identifying the MTD and is safer than some existing designs. Conclusion: The MC-Keyboard provides a novel, simple, and safe approach to assessing safety and identifying the MTD for targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Liyun Jiang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhulin Yin
- Clinical Trials and Statistics Unit, Institute of Cancer Research, London, UK
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
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
|
15
|
Franceschini D, Loi M, Marzo AM, Dominici L, Spoto R, Bertolini A, Lo Faro L, La Fauci F, Marini B, Di Cristina L, Scorsetti M. STRILL: Phase I Trial Evaluating Stereotactic Body Radiotherapy (SBRT) Dose Escalation for Re-Irradiation of Inoperable Peripheral Lung Lesions. Diseases 2024; 12:153. [PMID: 39057124 PMCID: PMC11276608 DOI: 10.3390/diseases12070153] [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: 06/17/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Few data are available on the role of SBRT re-irradiation for isolated recurrences. We designed a prospective phase I study to evaluate the maximum tolerated dose (MTD) of SBRT for thoracic re-irradiation, for peripheral lung lesions. RT was delivered with a dose escalation design from 30 Gy in five fractions up to 50 Gy in five fractions. The primary end point was the definition of the maximum tolerated dose (MTD) of SBRT for thoracic re-irradiation. The dose-limiting toxicity was pneumonia ≥G3. Fifteen patients were enrolled. No cases of pneumonia ≥G3 occurred in any of our cohorts. Only one patient developed pneumonia G1 during treatment. Three patients developed acute toxicities that included dyspnea G1, cardiac failure G3, and chest wall pain. One patient developed G3 late toxicity with acute coronary syndrome. After a median follow-up of 21 months (range 3.6-29.1 months), six patients (40%) had a local relapse. Distant relapse occurred in five patients (33.3%). At the last follow-up, six patients died, all but two due to progressive disease. SBRT dose escalation for thoracic re-irradiation is an effective and well-tolerated option for patients with inoperable lung lesions after a first thoracic RT with acceptable acute and late toxicities.
Collapse
Affiliation(s)
- Davide Franceschini
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
| | - Mauro Loi
- Department of Radiation Oncology, Azienda Universitaria Ospedaliera Careggi, 50134 Florence, Italy;
| | - Antonio Marco Marzo
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
| | - Luca Dominici
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
| | - Ruggero Spoto
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
| | - Anna Bertolini
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Lorenzo Lo Faro
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
| | - Francesco La Fauci
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
| | - Beatrice Marini
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Luciana Di Cristina
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Marta Scorsetti
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.M.M.); (L.D.); (R.S.); (A.B.); (L.L.F.); (F.L.F.); (B.M.); (L.D.C.); (M.S.)
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| |
Collapse
|
16
|
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
|
17
|
Chiuzan C, Dehbi HM. The 3 + 3 design in dose-finding studies with small sample sizes: Pitfalls and possible remedies. Clin Trials 2024; 21:350-357. [PMID: 38618916 DOI: 10.1177/17407745241240401] [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: 04/16/2024]
Abstract
In the last few years, numerous novel designs have been proposed to improve the efficiency and accuracy of phase I trials to identify the maximum-tolerated dose (MTD) or the optimal biological dose (OBD) for noncytotoxic agents. However, the conventional 3+3 approach, known for its and poor performance, continues to be an attractive choice for many trials despite these alternative suggestions. The article seeks to underscore the importance of moving beyond the 3+3 design by highlighting a different key element in trial design: the estimation of sample size and its crucial role in predicting toxicity and determining the MTD. We use simulation studies to compare the performance of the most used phase I approaches: 3+3, Continual Reassessment Method (CRM), Keyboard and Bayesian Optimal Interval (BOIN) designs regarding three key operating characteristics: the percentage of correct selection of the true MTD, the average number of patients allocated per dose level, and the average total sample size. The simulation results consistently show that the 3+3 algorithm underperforms in comparison to model-based and model-assisted designs across all scenarios and metrics. The 3+3 method yields significantly lower (up to three times) probabilities in identifying the correct MTD, often selecting doses one or even two levels below the actual MTD. The 3+3 design allocates significantly fewer patients at the true MTD, assigns higher numbers to lower dose levels, and rarely explores doses above the target dose-limiting toxicity (DLT) rate. The overall performance of the 3+3 method is suboptimal, with a high level of unexplained uncertainty and significant implications for accurately determining the MTD. While the primary focus of the article is to demonstrate the limitations of the 3+3 algorithm, the question remains about the preferred alternative approach. The intention is not to definitively recommend one model-based or model-assisted method over others, as their performance can vary based on parameters and model specifications. However, the presented results indicate that the CRM, Keyboard, and BOIN designs consistently outperform the 3+3 and offer improved efficiency and precision in determining the MTD, which is crucial in early-phase clinical trials.
Collapse
Affiliation(s)
- Cody Chiuzan
- Northwell Health, New Hyde Park, NY, USA
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Hakim-Moulay Dehbi
- Comprehensive Clinical Trials Unit, University College London, London, UK
| |
Collapse
|
18
|
Viraswami-Appanna K, Buenconsejo J, Baidoo C, Chan I, Li D, Micsinai-Balan M, Tiwari R, Yang L, Sethuraman V. Accelerating drug development at Bristol Myers Squibb through innovation. Drug Discov Today 2024; 29:103952. [PMID: 38508230 DOI: 10.1016/j.drudis.2024.103952] [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: 12/30/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
This paper focuses on the use of novel technologies and innovative trial designs to accelerate evidence generation and increase pharmaceutical Research and Development (R&D) productivity, at Bristol Myers Squibb. We summarize learnings with case examples, on how we prepared and continuously evolved to address the increasing cost, complexities, and external pressures in drug development, to bring innovative medicines to patients much faster. These learnings were based on review of internal efforts toward accelerating R&D focusing on four key areas: adopting innovative trial designs, optimizing trial designs, leveraging external control data, and implementing novel methods using artificial intelligence and machine learning.
Collapse
Affiliation(s)
| | - Joan Buenconsejo
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Charlotte Baidoo
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Ivan Chan
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Daniel Li
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | | | - Ram Tiwari
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Ling Yang
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Venkat Sethuraman
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
19
|
Fogel EL, Easler JJ, Yuan Y, Yadav D, Conwell DL, Vege SS, Han SY, Park W, Patrick V, White FA. Safety, Tolerability, and Dose-Limiting Toxicity of Lacosamide in Patients With Painful Chronic Pancreatitis: Protocol for a Phase 1 Clinical Trial to Determine Safety and Identify Side Effects. JMIR Res Protoc 2024; 13:e50513. [PMID: 38451604 PMCID: PMC10958339 DOI: 10.2196/50513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Chronic abdominal pain is the hallmark symptom of chronic pancreatitis (CP), with 50% to 80% of patients seeking medical attention for pain control. Although several management options are available, outcomes are often disappointing, and opioids remain a mainstay of therapy. Opioid-induced hyperalgesia is a phenomenon resulting in dose escalation, which may occur partly because of the effects of opioids on voltage-gated sodium channels associated with pain. Preclinical observations demonstrate that the combination of an opioid and the antiseizure drug lacosamide diminishes opioid-induced hyperalgesia and improves pain control. OBJECTIVE In this phase 1 trial, we aim to determine the safety, tolerability, and dose-limiting toxicity of adding lacosamide to opioids for the treatment of painful CP and assess the feasibility of performance of a pilot study of adding lacosamide to opioid therapy in patients with CP. As an exploratory aim, we will assess the efficacy of adding lacosamide to opioid therapy in patients with painful CP. METHODS Using the Bayesian optimal interval design, we will conduct a dose-escalation trial of adding lacosamide to opioid therapy in patients with painful CP enrolled in cohorts of size 3. The initial dose will be 50 mg taken orally twice a day, followed by incremental increases to a maximum dose of 400 mg/day, with lacosamide administered for 7 days at each dose level. Adverse events will be documented according to Common Terminology Criteria for Adverse Events (version 5.0). RESULTS As of December 2023, we have currently enrolled 6 participants. The minimum number of participants to be enrolled is 12 with a maximum of 24. We expect to publish the results by March 2025. CONCLUSIONS This trial will test the feasibility of the study design and provide reassurance regarding the tolerability and safety of opioids in treating painful CP. It is anticipated that lacosamide will prove to be safe and well tolerated, supporting a subsequent phase 2 trial assessing the efficacy of lacosamide+opioid therapy in patients with painful CP, and that lacosamide combined with opiates will lower the opioid dose necessary for pain relief and improve the safety profile of opioid use in treating painful CP. TRIAL REGISTRATION Clinicaltrials.gov NCT05603702; https://clinicaltrials.gov/study/NCT05603702. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/50513.
Collapse
Affiliation(s)
- Evan L Fogel
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Jeffrey J Easler
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Darwin L Conwell
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Samuel Y Han
- Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Walter Park
- Department of Medicine, Stanford University Medical Center, Stanford, CA, United States
| | - Vanessa Patrick
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Fletcher A White
- Department of Anesthesia, School of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
20
|
Rodney S, Banerji U. Optimizing the FDA's Project Optimus: opportunities and challenges. Nat Rev Clin Oncol 2024; 21:165-166. [PMID: 38129533 DOI: 10.1038/s41571-023-00853-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Affiliation(s)
- Simon Rodney
- Drug Development Unit, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK.
| | - Udai Banerji
- Drug Development Unit, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
21
|
Yin Z, Mander AP, de Bono JS, Zheng H, Yap C. Handling Incomplete or Late-Onset Toxicities in Early-Phase Dose-Finding Clinical Trials: Current Practice and Future Prospects. JCO Precis Oncol 2024; 8:e2300441. [PMID: 38181316 DOI: 10.1200/po.23.00441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/28/2023] [Accepted: 10/12/2023] [Indexed: 01/07/2024] Open
Abstract
PURPOSE The way late-onset toxicities are managed can affect trial outcomes and participant safety. Specifically, participants often might not have completed their entire follow-up period to observe any toxicities before new participants would be recruited. We conducted a methodological review of published early-phase dose-finding clinical trials that used designs accounting for partial and complete toxicity information, aiming to understand (1) how such designs were implemented and reported and (2) if sufficient information was provided to enable the replicability of trial results. METHODS Until March 26, 2023, we identified 141 trials using the rolling 6 design, the time-to-event continuous reassessment method (TITE-CRM), the TITE-CRM with cycle information, the TITE Bayesian optimal interval design, the TITE cumulative cohort design, and the rapid enrollment design. Clinical settings, design parameters, practical considerations, and dose-limiting toxicity (DLT) information were extracted from these published trials. RESULTS The TITE-CRM (61, 43.3%) and the rolling 6 design (76, 53.9%) were most frequently implemented in practice. Trials using the TITE-CRM had longer DLT assessment windows beyond the first cycle compared with the rolling 6 design (52.5% v 6.6%). Most trials implementing the TITE-CRM (91.8%, 56 of 61) failed to describe essential parameters in the protocols or the study result papers. Only five TITE-CRM trials (8.2%, 5 of 61) reported sufficient information to enable replication of the final analysis. CONCLUSION When compared with trials using the rolling 6 design, those implementing the TITE-CRM design exhibited notable deficiencies in reporting essential details necessary for reproducibility. Inadequate reporting quality of advanced model-based trial designs hinders their credibility. We provide recommendations that can improve transparency, reproducibility, and accurate interpretation of the results for such designs.
Collapse
Affiliation(s)
- Zhulin Yin
- Clinical Trials and Statistics Unit, Institute of Cancer Research, London, United Kingdom
| | - Adrian P Mander
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - Johann S de Bono
- Drug Development Unit, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Haiyan Zheng
- Department of Mathematical Sciences, University of Bath, Bath, United Kingdom
| | - Christina Yap
- Clinical Trials and Statistics Unit, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
22
|
You ZY, Wu MF, Li H, Ye YF, Wang LJ, Lin ZQ, Li J. A phase I dose-finding trial of hyperthermic intraperitoneal docetaxel combined with cisplatin in patients with advanced-stage ovarian cancer. J Gynecol Oncol 2024; 35:e1. [PMID: 37477105 PMCID: PMC10792218 DOI: 10.3802/jgo.2024.35.e1] [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: 02/09/2023] [Revised: 05/11/2023] [Accepted: 06/24/2023] [Indexed: 07/22/2023] Open
Abstract
OBJECTIVE To identify the maximum tolerated dose (MTD) of docetaxel combined with a fixed dose of cisplatin (75 mg/m²) delivered as hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with ovarian cancer. METHODS In this phase I trial, a time-to-event Bayesian optimal interval design was used. Docetaxel was given at a starting dose of 60 mg/m² and was increased in 5 mg/m² increments until the MTD was determined or the maximum dose level of 75 mg/m² was reached. The dose-limiting toxicity (DLT) rate was set at 25%, with a total sample size of 30 patients. HIPEC was delivered immediately following debulking surgery at a target temperature of 43°C for 90 minutes. RESULTS From August 2022 to November 2022, 30 patients were enrolled. Among the patients who received a dose of docetaxel ≤65 mg/m², no DLT was reported. DLTs were observed in one patient who received 70 mg/m² docetaxel (grade 3 anaemia) and in three patients who received 75 mg/m² docetaxel (one case of grade 3 anaemia, one case of grade 3 hepatic impairment and one case of grade 4 thrombocytopenia). Patients treated with docetaxel 75 mg/m² in combination with cisplatin 75 mg/m² had an estimated DLT rate of 25%, which was the closest to the target DLT rate and was therefore chosen as the MTD. CONCLUSION Docetaxel, in combination with a fixed dose of cisplatin (75 mg/m²), can be used safely at intraperitoneal doses of 75 mg/m² in ovarian cancer patients who received HIPEC (43°C, 90 minutes) following debulking surgery. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05410483.
Collapse
Affiliation(s)
- Zhi-Yao You
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Miao-Fang Wu
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Li
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Fang Ye
- Clinical research design division, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Juan Wang
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong-Qiu Lin
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Li
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Gynecology, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China.
| |
Collapse
|
23
|
Takeda K, Zhu J, Li R, Yamaguchi Y. A Bayesian optimal interval design for dose optimization with a randomization scheme based on pharmacokinetics outcomes in oncology. Pharm Stat 2023; 22:1104-1115. [PMID: 37545018 DOI: 10.1002/pst.2332] [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: 02/04/2023] [Revised: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
The primary objective of an oncology dose-finding trial for novel therapies, such as molecularly targeted agents and immune-oncology therapies, is to identify the optimal dose (OD) that is tolerable and therapeutically beneficial for subjects in subsequent clinical trials. Pharmacokinetic (PK) information is considered an appropriate indicator for evaluating the level of drug intervention in humans from a pharmacological perspective. Several novel anticancer agents have been shown to have significant exposure-efficacy relationships, and some PK information has been considered an important predictor of efficacy. This paper proposes a Bayesian optimal interval design for dose optimization with a randomization scheme based on PK outcomes in oncology. A simulation study shows that the proposed design has advantages compared to the other designs in the percentage of correct OD selection and the average number of patients allocated to OD in various realistic settings.
Collapse
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Jing Zhu
- Data Science, Astellas Pharma China, Beijing, China
| | - Ran Li
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Yusuke Yamaguchi
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| |
Collapse
|
24
|
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: 3] [Impact Index Per Article: 1.5] [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
|
25
|
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
|
26
|
Zhang J, Chen X, Li B, Yan F. A comparative study of adaptive trial designs for dose optimization. Pharm Stat 2023; 22:797-814. [PMID: 37156731 DOI: 10.1002/pst.2306] [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] [Received: 05/18/2022] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Recently, the US Food and Drug Administration Oncology Center of Excellence initiated Project Optimus to reform the dose optimization and dose selection paradigm in oncology drug development. The agency pointed out that the current paradigm for dose selection-based on the maximum tolerated dose (MTD)-is not sufficient for molecularly targeted therapies and immunotherapies, for which efficacy may not increase after the dose reaches a certain level. In these cases, it is more appropriate to identify the optimal biological dose (OBD) that optimizes the risk-benefit tradeoff of the drug. Project Optimus has spurred tremendous interest and urgent need for guidance on designing dose optimization trials. In this article, we review several representative dose optimization designs, including model-based and model-assisted designs, and compare their operating characteristics based on 10,000 randomly generated scenarios with various dose-toxicity and dose-efficacy curves and some fixed representative scenarios. The results show that, compared with model-based designs, model-assisted methods have advantages of easy-to-implement, robustness, and high accuracy to identify OBD. Some guidance is provided to help biostatisticians and clinicians to choose appropriate dose optimization methods in practice.
Collapse
Affiliation(s)
- Jingyi Zhang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bosheng Li
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
27
|
Andrillon A, Biard L, Lee SM. Incorporating patient-reported outcomes in dose-finding clinical trials with continuous patient enrollment. J Biopharm Stat 2023:1-12. [PMID: 37496233 PMCID: PMC10811281 DOI: 10.1080/10543406.2023.2236216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 07/09/2023] [Indexed: 07/28/2023]
Abstract
Dose-finding clinical trials in oncology estimate the maximum tolerated dose (MTD), based on toxicity obtained from the clinician's perspective. While the collection of patient-reported outcomes (PROs) has been advocated to better inform treatment tolerability, there is a lack of guidance and methods on how to use PROs for dose assignments and recommendations. The PRO continual reassessment method (PRO-CRM) has been proposed to formally incorporate PROs into dose-finding trials. In this paper, we propose two extensions of the PRO-CRM, which allow continuous enrollment of patients and longer toxicity observation windows to capture late-onset or cumulative toxicities by using a weighted likelihood to include the partial toxicity follow-up information. The TITE-PRO-CRM uses both the PRO and the clinician's information during the trial for dose assignment decisions and at the end of the trial to estimate the MTD. The TITE-CRM + PRO uses clinician's information solely to inform dose assignments during the trial and incorporates PRO at the end of the trial for the estimation of the MTD. Simulation studies show that the TITE-PRO-CRM performs similarly to the PRO-CRM in terms of dose recommendation and assignments during the trial while almost halving trial duration in case of an accrual of two patients per observation window. The TITE-CRM + PRO slightly underperforms compared to the TITE-PRO-CRM, but similar performance can be attained by requiring larger sample sizes. We also show that the performance of the proposed methods is robust to higher accrual rates, different toxicity hazards, and correlated time-to-clinician toxicity and time-to-patient toxicity data.
Collapse
Affiliation(s)
- Anaïs Andrillon
- INSERM U1153 Team ECSTRRA, Université Paris Cité, Paris, France
- Department of Statistical Methodology, Saryga, Tournus, France
| | - Lucie Biard
- INSERM U1153 Team ECSTRRA, Université Paris Cité, Paris, France
| | - Shing M. Lee
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
28
|
Li R, Takeda K, Rong A. Comparison Between Simultaneous and Sequential Utilization of Safety and Efficacy for Optimal Dose Determination in Bayesian Model-Assisted Designs. Ther Innov Regul Sci 2023; 57:728-736. [PMID: 37087525 DOI: 10.1007/s43441-023-00517-1] [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: 11/21/2022] [Accepted: 03/21/2023] [Indexed: 04/24/2023]
Abstract
It has become quite common in recent early oncology trials to include both the dose-finding and the dose-expansion parts within the same study. This shift can be viewed as a seamless way of conducting the trials to obtain information on safety and efficacy hence identifying an optimal dose (OD) rather than just the maximum tolerated dose (MTD). One approach is to conduct a dose-finding part based solely on toxicity outcomes, followed by a dose expansion part to evaluate efficacy outcomes. Another approach employs only the dose-finding part, where the dose-finding decisions are made utilizing both the efficacy and toxicity outcomes of those enrolled patients. In this paper, we compared the two approaches through simulation studies under various realistic settings. The percentage of correct ODs selection, the average number of patients allocated to the ODs, and the average trial duration are reported in choosing the appropriate designs for their early-stage dose-finding trials, including expansion cohorts.
Collapse
Affiliation(s)
- Ran Li
- Biostatistics, Data Science, Astellas Pharma Inc, 1 Astellas Way, N3.272.A, Northbrook, IL, 60062, USA.
| | - Kentaro Takeda
- Biostatistics, Data Science, Astellas Pharma Inc, 1 Astellas Way, N3.272.A, Northbrook, IL, 60062, USA
| | - Alan Rong
- Biostatistics, Data Science, Astellas Pharma Inc, 1 Astellas Way, N3.272.A, Northbrook, IL, 60062, USA
| |
Collapse
|
29
|
Lancman G, Moshier E, Cho HJ, Parekh S, Richard S, Richter J, Rodriguez C, Rossi A, Sanchez L, Jagannath S, Chari A. Trial designs and endpoints for immune therapies in multiple myeloma. Am J Hematol 2023; 98 Suppl 2:S35-S45. [PMID: 36200130 DOI: 10.1002/ajh.26753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/18/2022] [Accepted: 09/29/2022] [Indexed: 11/08/2022]
Abstract
Immune therapies, including CAR-T cells, bispecific antibodies, and antibody-drug conjugates, are revolutionizing the treatment of multiple myeloma. In this review, we discuss clinical trial design considerations relevant to immune therapies. We first examine issues pertinent to specific populations, including elderly, patients with renal impairment, high-risk/extramedullary disease, and prior immune therapies. We then highlight trial designs to optimize the selection of dose and schedule, explore rational combination therapies based on preclinical data, and evaluate the nuances of commonly used endpoints. By exploiting their pharmacokinetic/pharmacodynamic profiles and utilizing novel translational insights, we can optimize the use of immune therapies in multiple myeloma.
Collapse
Affiliation(s)
- Guido Lancman
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Erin Moshier
- Department of Population Health Science and Policy, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Hearn Jay Cho
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Samir Parekh
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Shambavi Richard
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Joshua Richter
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Cesar Rodriguez
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Adriana Rossi
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Larysa Sanchez
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Sundar Jagannath
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Ajai Chari
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
30
|
Kojima M. Data-dependent early completion of dose-finding trials for drug-combination. Stat Methods Med Res 2023; 32:820-828. [PMID: 36775992 DOI: 10.1177/09622802231155094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
PURPOSE Model-assisted designs for drug combination trials have been proposed as novel designs with simple and superior performance. However, model-assisted designs have the disadvantage that the sample size must be set in advance, and trials cannot be completed until the number of patients treated reaches the pre-set sample size. Model-assisted designs have a stopping rule that can be used to terminate the trial if the number of patients treated exceeds the predetermined number, there is no statistical basis for the predetermined number. Here, I propose two methods for data-dependent early completion of dose-finding trials for drug combination: (1) an early completion method based on dose retainment probability, and (2) an early completion method in which the dose retainment probability is adjusted by a bivariate isotonic regression. METHODS Early completion is determined when the dose retainment probability using both trial data and the number of remaining patients is high. Early completion of a virtual trial was demonstrated. The performances of the early completion methods were evaluated by simulation studies with 12 scenarios. RESULTS The simulation studies showed that the percentage of early completion was an average of approximately 70%, and the number of patients treated was 25% less than the planned sample size. The percentage of correct maximum tolerated dose combination selection for the early completion methods was similar to that of non-early completion methods with an average difference of approximately 3%. CONCLUSION The performance of the proposed early completion methods was similar to that of the non-early completion methods. Furthermore, the number of patients for determining early completion before the trial starts was determined and a program code for calculating the dose retainment probability was proposed.
Collapse
Affiliation(s)
- Masahiro Kojima
- Biometrics Department, R&D Division, 13486Kyowa Kirin Co., Ltd, Tokyo, Japan.,Research Center for Medical and Health Data Science, The Institute of Statistical Mathematics, Tokyo, Japan
| |
Collapse
|
31
|
Wages NA, Braun TM, Conaway MR. Isotonic design for phase I cancer clinical trials with late-onset toxicities. J Biopharm Stat 2023; 33:357-370. [PMID: 36606874 DOI: 10.1080/10543406.2022.2162068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This article addresses the problem of identifying the maximum tolerated dose (MTD) in Phase I dose-finding clinical trials with late-onset toxicities. The main design challenge is how best to adaptively allocate study participants to tolerable doses when the evaluation window for the toxicity endpoint is long relative to the accrual rate of new participants. We propose a new design framework based on order-restricted statistical inference that addresses this challenge in sequential dose assignments. We illustrate the proposed method on real data from a Phase I trial of bortezomib in lymphoma patients and apply it to a Phase I trial of radiotherapy in prostate cancer patients. We conduct extensive simulation studies to compare our design's operating characteristics to existing published methods. Overall, our proposed design demonstrates good performance relative to existing methods in allocating participants at and around the MTD during the study and accurately recommending the MTD at the study conclusion.
Collapse
Affiliation(s)
- Nolan A Wages
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Thomas M Braun
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Mark R Conaway
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
32
|
Andrillon A, Chevret S, Lee SM, Biard L. Surv-CRM-12: A Bayesian phase I/II survival CRM for right-censored toxicity endpoints with competing disease progression. Stat Med 2022; 41:5753-5766. [PMID: 36259523 PMCID: PMC9691552 DOI: 10.1002/sim.9591] [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: 12/16/2021] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 01/12/2023]
Abstract
The growing interest in new classes of anti-cancer agents, such as molecularly-targeted therapies and immunotherapies with modes of action different from those of cytotoxic chemotherapies, has changed the dose-finding paradigm. In this setting, the observation of late-onset toxicity endpoints may be precluded by treatment and trial discontinuation due to disease progression, defining a competing event to toxicity. Trial designs where dose-finding is modeled in the framework of a survival competing risks model appear particularly well-suited. We aim to provide a phase I/II dose-finding design that allows dose-limiting toxicity (DLT) outcomes to be delayed or unobserved due to competing progression within the possibly long observation window. The proposed design named the Survival-continual reassessment method-12, uses survival models for right-censored DLT and progression endpoints. In this competing risks framework, cause-specific hazards for DLT and progression-free of DLT were considered, with model parameters estimated using Bayesian inference. It aims to identify the optimal dose (OD), by minimizing the cumulative incidence of disease progression, given an acceptable toxicity threshold. In a simulation study, design operating characteristics were evaluated and compared to the TITE-BOIN-ET design and a nonparametric benchmark approach. The performance of the proposed method was consistent with the complexity of scenarios as assessed by the nonparametric benchmark. We found that the proposed design presents satisfying operating characteristics in selecting the OD and safety.
Collapse
Affiliation(s)
- Anaïs Andrillon
- ECSTRRA Team, UMR‐1153Université de Paris, INSERM, AP‐HP, Hôpital Saint LouisParisFrance,Department of BiostatisticsMailman School of Public Health, Columbia UniversityNew YorkNew YorkUSA
| | - Sylvie Chevret
- ECSTRRA Team, UMR‐1153Université de Paris, INSERM, AP‐HP, Hôpital Saint LouisParisFrance
| | - Shing M. Lee
- Department of BiostatisticsMailman School of Public Health, Columbia UniversityNew YorkNew YorkUSA
| | - Lucie Biard
- ECSTRRA Team, UMR‐1153Université de Paris, INSERM, AP‐HP, Hôpital Saint LouisParisFrance
| |
Collapse
|
33
|
Park Y. Interval design to identify the optimal biological dose for immunotherapy. Contemp Clin Trials Commun 2022; 30:101005. [PMID: 36186542 PMCID: PMC9520219 DOI: 10.1016/j.conctc.2022.101005] [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: 12/30/2021] [Revised: 07/31/2022] [Accepted: 09/16/2022] [Indexed: 11/04/2022] Open
Abstract
Immunotherapeutics have revolutionized the treatment of metastatic cancers and are expected to play an increasingly prominent role in the treatment of cancer patients. Recent advances in checkpoint inhibition show promising early results in a number of malignancies, and several treatments have been approved for use. However, the immunotherapeutic agents have been shown to have different mechanisms of antitumor activity from cytotoxic agents, and many limitations and challenges encountered in the traditional paradigm were recently pointed out for immunotherapy. I propose a desirability-based method to determine the optimal biological dose of immunotherapeutics by effectively using toxicity, immune response, and tumor response. Moreover, a new dose allocation algorithm of interval designs is proposed to incorporate immune response in addition to toxicity and tumor response. Simulation studies show that the proposed design has desirable operating characteristics compared to existing dose-finding designs. It also inherits the strengths of interval designs for dose-finding trials, yielding good performance with ease of implementation.
Collapse
Affiliation(s)
- Yeonhee Park
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, United States of America
| |
Collapse
|
34
|
Yuan Y, Zhao Y. Commentary on “Improving the performance of Bayesian logistic regression model with overdose control in oncology dose‐finding studies”. Stat Med 2022; 41:5484-5490. [DOI: 10.1002/sim.9496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Ying Yuan
- Department of Biostatistics The University of Texas MD Anderson Cancer Center Houston Texas USA
| | - Yixuan Zhao
- Department of Biostatistics and Data Science, School of Public Health The University of Texas Health Science Center at Houston Houston Texas USA
| |
Collapse
|
35
|
Hatayama T, Yasui S. CUSUMIN: A cumulative sum interval design for cancer phase I dose finding studies. Pharm Stat 2022; 21:1324-1341. [PMID: 35833753 PMCID: PMC9796866 DOI: 10.1002/pst.2247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 01/07/2023]
Abstract
Recently, model-assisted designs, including the Bayesian optimal interval (BOIN) design with optimal thresholds to determine the dose for the next cohort, have been proposed for cancer phase I studies. Model-assisted designs are useful because of their good performance as model-based designs in addition to their algorithm-based simplicity. In BOIN, escalation and de-escalation based on boundaries can be understood as a type of change point detection based on a sequential test procedure. Notably, the sequential test procedure is used in a wide range of fields and is known for its application to control charts, statistical monitoring methods used for detecting abnormalities in manufacturing processes. In control charts, abnormalities are detected if the control chart statistics are observed to be outside of the optimal boundaries. The cumulative sum (CUSUM) statistic, which is developed for control chart applications, derives higher power under the same erroneous judgment rate. Hence, it is expected that a more efficient model-assisted design can be achieved by the application of CUSUM statistics. In this study, a model-assisted design based on the CUSUM statistic is proposed. In the proposed design, the dose for the next cohort is decided by CUSUM statistics calculated from the counts of the dose-limiting toxicity and pre-defined boundaries, based on the CUSUM control chart scheme. Intensive simulation shows that our proposed method performs better than BOIN, and other representative model-assisted designs, including modified toxicity probability interval (mTPI) and Keyboard, in terms of controlling over-dosing rates while maintaining similar performance in the determination of maximum tolerated dose.
Collapse
Affiliation(s)
- Tomoyoshi Hatayama
- Statistics and Decision Sciences JapanJanssen Pharmaceutical K.K.TokyoJapan
| | - Seiichi Yasui
- Department of Industrial AdministrationTokyo University of ScienceTokyoJapan
| |
Collapse
|
36
|
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: 0.3] [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
|
37
|
Li Y, Izem R. Novel clinical trial design and analytic methods to tackle challenges in therapeutic development in rare diseases. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1034. [PMID: 36267797 PMCID: PMC9577738 DOI: 10.21037/atm-21-5496] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/28/2022] [Indexed: 12/03/2022]
Abstract
While only a fraction of the worldwide population may have a particular rare disorder, millions of people worldwide are affected across the over 6,000 rare disorders and do not have a safe and effective approved therapy to help them live or manage complications from the disorder. Challenges to clinical development of new therapies in rare disorders include difficulty in powering and recruiting into a study in small and often heterogenous population, scarcity of natural history data informing critical design elements such as endpoint selection and study duration, and ethical and recruitment challenges in randomizing patients to a placebo arm. In this review, we describe some existing and novel strategies to tackle these challenges, by efficient utilization of available resources. We discuss the role of natural history studies and endpoint selection as they remain critical features that apply across designs and disorders. We also review some novel clinical trial designs including incorporating external control and/or longitudinal measures, master protocol designs, and adaptive designs. Additionally, we review some analytic strategies that are often associated with these designs, such as the use of causal inference methods, and Bayesian methods. We hope this review will raise awareness of these novel approaches and encourage their use in studies of rare diseases.
Collapse
Affiliation(s)
- Yimei Li
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, PA, USA
| | - Rima Izem
- Statistical Methodology and Consulting, Novartis, Basel, Switzerland
| |
Collapse
|
38
|
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: 5] [Impact Index Per Article: 1.7] [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
|
39
|
Lin X, Lyu J, Yuan S, Bi D, Wang SJ, Ji Y. Bayesian Sample Size Planning Tool for Phase I Dose-Finding Trials. JCO Precis Oncol 2022; 6:e2200046. [PMID: 36001859 PMCID: PMC9489190 DOI: 10.1200/po.22.00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/10/2022] [Accepted: 06/24/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Through Bayesian inference, we propose a method called BayeSize as a reference tool for investigators to assess the sample size and its associated scientific property for phase I clinical trials. METHODS BayeSize applies the concept of effect size in dose finding, assuming that the maximum tolerated dose can be identified on the basis of an interval surrounding its true value because of statistical uncertainty. Leveraging a decision framework that involves composite hypotheses, BayeSize uses two types of priors, the fitting prior (for model fitting) and sampling prior (for data generation), to conduct sample size calculation under the constraints of statistical power and type I error. RESULTS Simulation results showed that BayeSize can provide reliable sample size estimation under the constraints of type I/II error rates. CONCLUSION BayeSize could facilitate phase I trial planning by providing appropriate sample size estimation. Look-up tables and R Shiny app are provided for practical applications.
Collapse
Affiliation(s)
- Xiaolei Lin
- School of Data Science, Fudan University, Shanghai, China
| | | | | | - Dehua Bi
- Laiya Consulting, Inc, Shanghai, China
| | | | - Yuan Ji
- Department of Public Health Sciences, The University of Chicago, Chicago, IL
| |
Collapse
|
40
|
Wages NA, Braun TM, Normolle DP, Schipper MJ. Adaptive Phase 1 Design in Radiation Therapy Trials. Int J Radiat Oncol Biol Phys 2022; 113:493-499. [PMID: 35777394 DOI: 10.1016/j.ijrobp.2022.02.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/20/2022] [Indexed: 10/17/2022]
Affiliation(s)
- Nolan A Wages
- Division of Translational Research & Applied Statistics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia.
| | - Thomas M Braun
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Daniel P Normolle
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania
| | - Matthew J Schipper
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan; Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
41
|
Chen X, Zhang J, Jiang Q, Yan F. Borrowing historical information to improve phase I clinical trials using meta-analytic-predictive priors. J Biopharm Stat 2022; 32:34-52. [PMID: 35594366 DOI: 10.1080/10543406.2022.2058526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Multiple phase I clinical trials may be performed to determine specific maximum tolerated doses (MTD) for specific races or cancer types. In these situations, borrowing historical information has potential to improve the accuracy of estimating toxicity rate and increase the probability of correctly targeting MTD. To utilize historical information in phase I clinical trials, we proposed using the Meta-Analytic-Predictive (MAP) priors to automatically estimate the heterogeneity between historical trials and give a relatively reasonable amount of borrowed information. We then applied MAP priors in some famous phase I trial designs, such as the continual reassessment method (CRM), Keyboard design and Bayesian optimal interval design (BOIN), to accomplish the process of dose finding. A clinical trial example and extended simulation studies show that our proposed methods have robust and efficient statistical performance, compared with those designs which do not consider borrowing information.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jingyi Zhang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qian Jiang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fangrong Yan
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
42
|
Zhou Y, Lin R, Lee JJ, Li D, Wang L, Li R, Yuan Y. TITE-BOIN12: A Bayesian phase I/II trial design to find the optimal biological dose with late-onset toxicity and efficacy. Stat Med 2022; 41:1918-1931. [PMID: 35098585 PMCID: PMC9199061 DOI: 10.1002/sim.9337] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 12/19/2021] [Accepted: 01/09/2022] [Indexed: 12/17/2022]
Abstract
In the era of immunotherapies and targeted therapies, the focus of early phase clinical trials has shifted from finding the maximum tolerated dose to identifying the optimal biological dose (OBD), which maximizes the toxicity-efficacy trade-off. One major impediment to using adaptive designs to find OBD is that efficacy or/and toxicity are often late-onset, hampering the designs' real-time decision rules for treating new patients. To address this issue, we propose the model-assisted TITE-BOIN12 design to find OBD with late-onset toxicity and efficacy. As an extension of the BOIN12 design, the TITE-BOIN12 design also uses utility to quantify the toxicity-efficacy trade-off. We consider two approaches, Bayesian data augmentation and an approximated likelihood method, to enable real-time decision making when some patients' toxicity and efficacy outcomes are pending. Extensive simulations show that, compared to some existing designs, TITE-BOIN12 significantly shortens the trial duration while having comparable or higher accuracy to identify OBD and a lower risk of overdosing patients. To facilitate the use of the TITE-BOIN12 design, we develop a user-friendly software freely available at http://www.trialdesign.org.
Collapse
Affiliation(s)
- Yanhong Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| | - J. Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Daniel Li
- Juno Therapeutics, a Bristol-Myers Squibb Company, WA, USA
| | - Li Wang
- Org Division, AbbVie Inc., IL, USA
| | - Ruobing Li
- The Center for Drug Evaluation, The National Medical Products Administration, Beijing, China
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| |
Collapse
|
43
|
Kojima M. Adaptive design for identifying maximum tolerated dose early to accelerate dose-finding trial. BMC Med Res Methodol 2022; 22:97. [PMID: 35382745 PMCID: PMC8985324 DOI: 10.1186/s12874-022-01584-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: 10/10/2021] [Accepted: 03/22/2022] [Indexed: 11/19/2022] Open
Abstract
Purpose The early identification of maximum tolerated dose (MTD) in phase I trial leads to faster progression to a phase II trial or an expansion cohort to confirm efficacy. Methods We propose a novel adaptive design for identifying MTD early to accelerate dose-finding trials. The early identification of MTD is determined adaptively by dose-retainment probability using a trial data via Bayesian analysis. We applied the early identification design to an actual trial. A simulation study evaluates the performance of the early identification design. Results In the actual study, we confirmed the MTD could be early identified and the study period was shortened. In the simulation study, the percentage of the correct MTD selection in the early identification Keyboard and early identification Bayesian optimal interval (BOIN) designs was almost same from the non-early identification version. The early identification Keyboard and BOIN designs reduced the study duration by about 50% from the model-assisted designs. In addition, the early identification Keyboard and BOIN designs reduced the study duration by about 20% from time-to-event model-assisted designs. Conclusion We proposed the early identification of MTD maintaining the accuracy to be able to short the study period. Supplementary Information The online version contains supplementary material available at 10.1186/s12874-022-01584-y.
Collapse
Affiliation(s)
- Masahiro Kojima
- Biometrics Department, Kyowa Kirin Co., Ltd, R&D Division, Tokyo, Japan. .,Department of Statistical Science, School of Multidisciplinary Sciences, The Graduate University for Advanced Studies, Tokyo, Japan.
| |
Collapse
|
44
|
Kojima M. Early Completion of Model-Assisted Designs for Dose-Finding Trials. JCO Precis Oncol 2022; 5:1449-1457. [PMID: 34994638 DOI: 10.1200/po.21.00192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE We propose a novel early completion method for phase I dose-finding trials using model-assisted designs. The trials can halt when a maximum tolerated dose (MTD) is estimated with sufficient accuracy. Early completion can reduce the average number of patients treated relative to the planned number, thereby allowing the trial to proceed to enrolling an expansion cohort for efficacy and enabling the trial to reach the next phase faster. METHODS Early completion is conducted on the basis of a dose-retainment probability using dose-assignment decisions. We evaluated early the completion for two actual trials. In addition, we performed a computer simulation to confirm the percentage of correctly selected MTDs, the early completion percentage, and the average number of patients treated. RESULTS In the evaluation of the two actual trials, we confirmed that the trials completed early. In the simulation results, we confirmed that the percentages of correct MTD selection were maintained relative to the original model-assisted designs. The early completion percentages ranged from 50% to 90%, and the number of patients treated reduced from 20%-60% relative to the planned number of patients. CONCLUSION We conclude that the early completion method can be applied unproblematically to the model-assisted design of phase I dose-finding trials.
Collapse
Affiliation(s)
- Masahiro Kojima
- Biometrics Department, R&D Division, Kyowa Kirin Co, Ltd, Tokyo, Japan.,Department of Statistical Science, School of Multidisciplinary Sciences, The Graduate University for Advanced Studies, Tokyo, Japan
| |
Collapse
|
45
|
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: 9] [Impact Index Per Article: 2.3] [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
|
46
|
Xu Z, Lin X. Probability-of-decision interval 3+3 (POD-i3+3) design for phase I dose finding trials with late-onset toxicity. Stat Methods Med Res 2021; 31:534-548. [PMID: 34806915 DOI: 10.1177/09622802211052746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Late-onset toxicities often occur in phase I trials investigating novel immunotherapy and molecular targeted therapies. For trials with cohort based designs (such as modified toxicity probability interval, Bayesian optimal interval, and i3+3), patients are often turned away since the current cohort are still being followed without definite dose-limiting toxicities, which results in prolonged trial duration and waste of patient resources. In this paper, we incorporate a probability-of-decision framework into the i3+3 design and allow real-time dosing inference when the next patient becomes available. Both follow-up time for the pending patients and time to dose-limiting toxicities for the observed patients are used in calculating the posterior probability of each possible dosing decision. An intensive simulation study is conducted to evaluate the operating characteristics of the newly proposed probability-of-decision-i3+3 design under various dosing scenarios and patient accrual settings. Results show that the probability-of-decision-i3+3 design achieves comparable safety and reliability performances but much shorter trial duration compared to the complete designs.
Collapse
Affiliation(s)
- Zichun Xu
- School of Life Sciences, 12478Fudan University, China
| | - Xiaolei Lin
- School of Data Science, 12478Fudan University, China
| |
Collapse
|
47
|
Yuan Y, Wu J, Gilbert MR. BOIN: a novel Bayesian design platform to accelerate early phase brain tumor clinical trials. Neurooncol Pract 2021; 8:627-638. [PMID: 34777832 DOI: 10.1093/nop/npab035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite decades of extensive research, the progress in developing effective treatments for primary brain tumors lags behind that of other cancers, largely due to the unique challenges of brain tumors (eg, the blood-brain barrier and high heterogeneity) that limit the delivery and efficacy of many therapeutic agents. One way to address this issue is to employ novel trial designs to better optimize the treatment regimen (eg, dose and schedule) in early phase trials to improve the success rate of subsequent phase III trials. The objective of this article is to introduce Bayesian optimal interval (BOIN) designs as a novel platform to design various types of early phase brain tumor trials, including single-agent and combination regimen trials, trials with late-onset toxicities, and trials aiming to find the optimal biological dose (OBD) based on both toxicity and efficacy. Unlike many novel Bayesian adaptive designs, which are difficult to understand and complicated to implement by clinical investigators, the BOIN designs are self-explanatory and user friendly, yet yield more robust and powerful operating characteristics than conventional designs. We illustrate the BOIN designs using a phase I clinical trial of brain tumor and provide software (freely available at www.trialdesign.org) to facilitate the application of the BOIN design.
Collapse
Affiliation(s)
- Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Wu
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark R Gilbert
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
48
|
Araujo DV, Oliva M, Li K, Fazelzad R, Liu ZA, Siu LL. Contemporary dose-escalation methods for early phase studies in the immunotherapeutics era. Eur J Cancer 2021; 158:85-98. [PMID: 34656816 DOI: 10.1016/j.ejca.2021.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 11/21/2022]
Abstract
Phase 1 dose-escalation trials are crucial to drug development by providing a framework to assess the toxicity of novel agents in a stepwise and monitored fashion. Despite widely adopted, rule-based dose-escalation methods (such as 3 + 3) are limited in finding the maximum tolerated dose (MTD) and tend to treat a significant number of patients at subtherapeutic doses. Newer methods of dose escalation, such as model-based and model-assisted designs, have emerged and are more accurate in finding MTD. However, these designs have not yet been broadly embraced by investigators. In this review, we summarise the advantages and disadvantages of contemporary dose-escalation methods, with emphasis on model-assisted designs, including time-to-event designs and hybrid methods involving optimal biological dose (OBD). The methods reviewed include mTPI, keyboard, BOIN, and their variations. In addition, the challenges of drug development (and dose-escalation) in the era of immunotherapeutics are discussed, where many of these agents typically have a wide therapeutic window. Fictional examples of how the dose-escalation method chosen can alter the outcomes of a phase 1 study are described, including the number of patients enrolled, the trial's timeframe, and the dose level chosen as MTD. Finally, the recent trends in dose-escalation methods applied in phase 1 trials in the immunotherapeutics era are reviewed. Among 856 phase I trials from 2014 to 2019, a trend towards the increased use of model-based and model-assisted designs over time (OR = 1.24) was detected. However, only 8% of the studies used non-rule-based dose-escalation methods. Increasing familiarity with such dose-escalation methods will likely facilitate their uptake in clinical trials.
Collapse
Affiliation(s)
- Daniel V Araujo
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Medical Oncology, Hospital de Base, São José Do Rio Preto, SP, Brazil
| | - Marc Oliva
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Medical Oncology, Institut Catala d' Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Kecheng Li
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Rouhi Fazelzad
- Library and Information Services, University Health Network, Toronto, ON, Canada
| | - Zhihui Amy Liu
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, ON, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
49
|
Kurzrock R, Lin CC, Wu TC, Hobbs BP, Pestana RC, Hong DS. Moving Beyond 3+3: The Future of Clinical Trial Design. Am Soc Clin Oncol Educ Book 2021; 41:e133-e144. [PMID: 34061563 DOI: 10.1200/edbk_319783] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Misgivings have been raised about the operating characteristics of the canonical 3+3 dose-escalation phase I clinical trial design. Yet, the traditional 3+3 design is still the most commonly used. Although it has been implied that adhering to this design is due to a stubborn reluctance to adopt change despite other designs performing better in hypothetical computer-generated simulation models, the continued adherence to 3+3 dose-escalation phase I strategies is more likely because these designs perform the best in the real world, pinpointing the correct dose and important side effects with an acceptable degree of precision. Beyond statistical simulations, there are little data to refute the supposed shortcomings ascribed to the 3+3 method. Even so, to address the unique nuances of gene- and immune-targeted compounds, a variety of inventive phase 1 trial designs have been suggested. Strategies for developing these therapies have launched first-in-human studies devised to acquire a breadth of patient data that far exceed the size of a typical phase I design and blur the distinction between dose selection and efficacy evaluation. Recent phase I trials of promising cancer therapies assessed objective tumor response and durability at various doses and schedules as well as incorporated multiple expansion cohorts spanning a variety of histology or biomarker-defined tumor subtypes, sometimes resulting in U.S. Food and Drug Administration approval after phase I. This article reviews recent innovations in phase I design from the perspective of multiple stakeholders and provides recommendations for future trials.
Collapse
Affiliation(s)
- Razelle Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsung-Che Wu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Brian P Hobbs
- Department of Population Health, Dell Medical School, University of Texas at Austin, Austin, TX
| | - Roberto Carmagnani Pestana
- Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
50
|
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
|