1
|
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
|
2
|
Pelizzola M, Tanderup K, Chopra S, Jürgenliemk-Schulz IM, Nout R, Kirchheiner K, Spampinato S. Co-occurrence of symptoms after radiochemotherapy in locally advanced cervix cancer patients: a cluster analysis. Acta Oncol 2023; 62:1479-1487. [PMID: 37906286 DOI: 10.1080/0284186x.2023.2271252] [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: 06/26/2023] [Accepted: 10/06/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND State of the art combined radiochemotherapy and image-guided brachytherapy for locally advanced cervical cancer (LACC) has shown improved disease control and survival as well as a significant reduction of organ related morbidity. However, LACC cancer survivors are still experiencing a spectrum of symptoms. The aim of this study was to identify co-occurring symptoms in cervix cancer survivors by using patient-reported outcome and physician assessed morbidity. MATERIALS AND METHOD EMBRACE I is a multicenter prospective observational study with 1416 LACC patients (2008-2015). Information on physician-assessed morbidity and patient-reported outcome was assessed at baseline and at regular follow-ups up with the CTCAE v.3 and EORTC-C30/CX24, respectively. Patients with at least 2 years of follow-up were included and data from 3 months to 2 years was used in the analysis. Factor analysis was used on both EORTC and CTCAE data with symptoms and follow-ups as observations. The extracted factors represent clusters of symptoms. Subsequently, regression models were built to investigate associations between the symptom clusters and QOL. RESULTS The analysis included 742 patients. Despite the differences in the definition of physician-assessed and patient-reported symptoms, similar clusters are identified by the two assessment methods. Three main organ-related clusters are recognized for urinary, gastro-intestinal and vaginal morbidity. Furthermore, a general symptoms cluster where fatigue, pain, insomnia, neuropathy, and hot flashes have large weights is found. Lastly, a cluster with nausea, vomit and lack of appetite is also identified. The general, gastrointestinal and nausea clusters show significant associations with general QOL. CONCLUSIONS This analysis on both PRO and physician-assessed morbidity found a cluster associated with general symptoms and organ-related symptom clusters (urinary, gastrointestinal, vaginal). This shows that LACC survivors experience a variety of co-occurring symptoms. Our analysis also shows that the cluster of general symptoms is associated with a decrease in QOL.
Collapse
Affiliation(s)
- Marta Pelizzola
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Kari Tanderup
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Supriya Chopra
- Department of Radiation Oncology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Navi Mumbai, India
| | - Ina M Jürgenliemk-Schulz
- Department of Radiation Oncology, University Medical Centre Utrecht, Utrecht, CX, The Netherlands
| | - Remi Nout
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Kathrin Kirchheiner
- Department of Radiation Oncology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sofia Spampinato
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
3
|
Choi JI, Simone CB, Lozano A, Frank SJ. Advances and Challenges in Conducting Clinical Trials With Proton Beam Therapy. Semin Radiat Oncol 2023; 33:407-415. [PMID: 37684070 PMCID: PMC10503212 DOI: 10.1016/j.semradonc.2023.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Advances in proton therapy have garnered much attention and speculation in recent years as the indications for proton therapy have grown beyond pediatric, prostate, spine, and ocular tumors. To achieve and maintain consistent access to this cancer treatment and to ensure the future viability and availability of proton centers in the United States, a call for evidence has been heard and answered by proton radiation oncologists. Answers provided in this review include the evolution of proton therapy research, rationale for proton clinical trial design, challenges in and barriers to the conduct of proton therapy research, and other unique considerations for the study of proton therapy.
Collapse
Affiliation(s)
- J Isabelle Choi
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY.; New York Proton Center, New York, NY..
| | - Charles B Simone
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY.; New York Proton Center, New York, NY
| | - Alicia Lozano
- Center for Biostatistics and Health Data Science, Department of Statistics, Virginia Tech, Roanoke, VA
| | - Steven J Frank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
4
|
O'Connell NS, Wages NA, Garrett-Mayer E. Quasi-partial order continual reassessment method: Applying toxicity scores to cancer dose-finding drug combination trials. Contemp Clin Trials 2023; 125:107050. [PMID: 36529437 DOI: 10.1016/j.cct.2022.107050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
The primary endpoint of most dose-finding cancer trials is patient toxicity, and the primary goal is to identify the maximum tolerated dose (MTD), that is, the highest dose that falls below or within a pre-specified toxicity tolerability threshold. Conventionally, dose-finding methods have utilized a binary toxicity endpoint based on whether or not a patient experiences a dose limiting-toxicity (DLT). Improving upon this, in recent years several methods have been developed for modeling toxicity scores, a novel continuous endpoint designed to more precisely estimate patient toxicity burden. Separately, drug-combination trials have become increasingly prevalent, and due to added complexities regarding estimating 'true' dose ordering and potential for more complex patient toxicity profiles, provide an ideal setting which may benefit from the improved precision of toxicity scores. In this paper, we merge two frameworks based on the Continual Reassessment Method (CRM) - the Quasi-CRM and the Partial Order CRM (POCRM) - to propose a novel approach for modeling toxicity scores in a combination-trial setting. We demonstrate that utilizing toxicity scores has the potential to greatly improve correct dose-selection over a variety of trial scenarios. We further present a simple adaptation to the toxicity-score model to control for potential over-dosing issues such that it adheres to the conventional DLT definition and will, at worst, perform equivalently to that of the traditional binary DLT framework. We demonstrate that extending toxicity scores to the combination-trial setting offers potential for improvement over the conventional binary endpoint models.
Collapse
Affiliation(s)
- Nathaniel S O'Connell
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston Salem, NC, USA.
| | - Nolan A Wages
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Elizabeth Garrett-Mayer
- Center for Research and Analytics, American Society for Clinical Oncology, Alexandria, VA, USA
| |
Collapse
|
5
|
Doersch KM, Tabayoyong WB, Bandari J. Evaluation of toxicities for intravesical drugs in phase 1 bladder cancer trials. Cancer 2023; 129:39-48. [PMID: 36262086 DOI: 10.1002/cncr.34508] [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: 07/06/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Improving clinical trial design is important for optimizing approval of safe and effective drugs. Phase 1 clinical trials seek to determine phase 2 doses by investigating predefined dose-limiting toxicities. Traditional definitions of dose-limiting toxicity may not be applicable to intravesical therapies for bladder cancer. This study compared the frequency of dose-limiting toxicities and serious adverse events in bladder cancer trials for intravesical therapies to other routes of administration. METHODS Studies were abstracted from ClinicalTrials.gov and reconciled with a PubMed search. Primary and secondary end points were predefined before data abstraction, and the primary end point was subject-level dose-limiting toxicity rate. Fisher exact tests were performed with p < .05 designated as significant. RESULTS Eighteen intravesical studies and 24 studies with other routes of administration (the per os/intravenous/intramuscular [PO/IV/IM] group) were identified. Dose-limiting toxicities were reported in 38.9% of intravesical studies, affecting 3.29% of subjects, compared with 30.0% of PO/IV/IM studies representing 4.19% of subjects (p = .52 for study-level and p = .60 for subject-level comparisons). Serious adverse events occurred in 53.9% of intravesical studies in 10.3% of subjects versus 91.0% of studies reporting serious adverse events affecting 41.4% of subjects in the PO/IV/IM group (p = .03 for subject-level and p < .0001 for study-level comparisons). CONCLUSIONS There was no difference in subject-level dose-limiting toxicity rate between intravesical and PO/IV/IM bladder cancer trials. The serious adverse event rate was lower in the intravesical group. Heterogeneity of dose-limiting toxicity definition may affect interpretation of toxicity in phase 1 bladder cancer clinical trials studying different routes of administration. LAY SUMMARY Bladder cancer is a common cancer type that may be treated with therapies that are instilled into the bladder and act locally, called intravesical therapies. This study used publicly available regulatory data from early phase clinical trials to determine whether measures of tolerability used in clinical trials are applicable to intravesical therapies for bladder cancer.
Collapse
Affiliation(s)
- Karen M Doersch
- Department of Urology, University of Rochester, Rochester, New York, USA
| | | | - Jathin Bandari
- Department of Urology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
6
|
Hobbs BP, Pestana RC, Zabor EC, Kaizer AM, Hong DS. Basket Trials: Review of Current Practice and Innovations for Future Trials. J Clin Oncol 2022; 40:3520-3528. [PMID: 35537102 PMCID: PMC10476732 DOI: 10.1200/jco.21.02285] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/06/2021] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
Advances in biology and immunology have elucidated genetic and immunologic origins of cancer. Innovations in sequencing technologies revealed that distinct cancer histologies shared common genetic and immune phenotypic traits. Pharmacologic developments made it possible to target these alterations, yielding novel classes of targeted agents whose therapeutic potential span multiple tumor types. Basket trials, one type of master protocol, emerged as a tool for evaluating biomarker-targeted therapies among multiple tumor histologies. Conventionally conducted within the phase II setting and designed to estimate high and durable objective responses, basket trials pose challenges to statistical design and interpretation of results. This article reviews basket trials implemented in oncology studies and discusses issues related to their statistical design and analysis.
Collapse
Affiliation(s)
- Brian P. Hobbs
- Dell Medical School, The 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
| | - Emily C. Zabor
- Quantitative Health Sciences & Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Alexander M. Kaizer
- Biostatistics and Informatics, University of Colorado-Anschutz Medical Campus, Aurora, CO
| | - David S. Hong
- Investigational Cancer Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
7
|
Polan DF, Epelman MA, Wu VW, Sun Y, Varsta M, Owen DR, Jarema D, Matrosic CK, Jolly S, Schonewolf CA, Schipper MJ, Matuszak MM. Direct incorporation of patient-specific efficacy and toxicity estimates in radiation therapy plan optimization. Med Phys 2022; 49:6279-6292. [PMID: 35994026 PMCID: PMC9826508 DOI: 10.1002/mp.15940] [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: 02/01/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Current radiation therapy (RT) treatment planning relies mainly on pre-defined dose-based objectives and constraints to develop plans that aim to control disease while limiting damage to normal tissues during treatment. These objectives and constraints are generally population-based, in that they are developed from the aggregate response of a broad patient population to radiation. However, correlations of new biologic markers and patient-specific factors to treatment efficacy and toxicity provide the opportunity to further stratify patient populations and develop a more individualized approach to RT planning. We introduce a novel intensity-modulated radiation therapy (IMRT) optimization strategy that directly incorporates patient-specific dose response models into the planning process. In this strategy, we integrate the concept of utility-based planning where the optimization objective is to maximize the predicted value of overall treatment utility, defined by the probability of efficacy (e.g., local control) minus the weighted sum of toxicity probabilities. To demonstrate the feasibility of the approach, we apply the strategy to treatment planning for non-small cell lung cancer (NSCLC) patients. METHODS AND MATERIALS We developed a prioritized approach to patient-specific IMRT planning. Using a commercial treatment planning system (TPS), we calculate dose based on an influence matrix of beamlet-dose contributions to regions-of-interest. Then, outside of the TPS, we hierarchically solve two optimization problems to generate optimal beamlet weights that can then be imported back to the TPS. The first optimization problem maximizes a patient's overall plan utility subject to typical clinical dose constraints. In this process, we facilitate direct optimization of efficacy and toxicity trade-off based on individualized dose-response models. After optimal utility is determined, we solve a secondary optimization problem that minimizes a conventional dose-based objective subject to the same clinical dose constraints as the first stage but with the addition of a constraint to maintain the optimal utility from the first optimization solution. We tested this method by retrospectively generating plans for five previously treated NSCLC patients and comparing the prioritized utility plans to conventional plans optimized with only dose metric objectives. To define a plan utility function for each patient, we utilized previously published correlations of dose to local control and grade 3-5 toxicities that include patient age, stage, microRNA levels, and cytokine levels, among other clinical factors. RESULTS The proposed optimization approach successfully generated RT plans for five NSCLC patients that improve overall plan utility based on personalized efficacy and toxicity models while accounting for clinical dose constraints. Prioritized utility plans demonstrated the largest average improvement in local control (16.6%) when compared to plans generated with conventional planning objectives. However, for some patients, the utility-based plans resulted in similar local control estimates with decreased estimated toxicity. CONCLUSION The proposed optimization approach, where the maximization of a patient's RT plan utility is prioritized over the minimization of standardized dose metrics, has the potential to improve treatment outcomes by directly accounting for variability within a patient population. The implementation of the utility-based objective function offers an intuitive, humanized approach to biological optimization in which planning trade-offs are explicitly optimized.
Collapse
Affiliation(s)
- Daniel F Polan
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Marina A Epelman
- Department of Industrial and Operations EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Victor W Wu
- Department of Industrial and Operations EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Yilun Sun
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA,Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | | | - Daniel R Owen
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - David Jarema
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Charles K Matrosic
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Shruti Jolly
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - Matthew J Schipper
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA,Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Martha M Matuszak
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
8
|
Thomas KE, Karamanis A, Dauchy E, Chapple AG, Loch MM. Assessing the effect of blood type on death and a novel scoring system to assess clinical course in patients with COVID-19. Am J Med Sci 2022; 364:7-15. [PMID: 34986364 PMCID: PMC8720494 DOI: 10.1016/j.amjms.2021.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023]
Abstract
Background Coronavirus disease (COVID-19) continues to lead to worldwide morbidity and mortality. This study examined the association between blood type and clinical outcomes in patients with COVID-19 measured by a calculated morbidity score and mortality rates. The secondary aim was to investigate the relationship between patient characteristics and COVID-19 associated clinical outcomes and mortality. Methods Logistic regression was used to determine what factors were associated with death. A total morbidity score was constructed based on overall patient's COVID-19 clinical course. This score was modeled using Quasi-Poisson regression. Bayesian variable selection was used for the logistic regression to obtain a posterior probability that blood type is important in predicting worsened clinical outcomes and death. Results Neither blood type nor Rh+ status was a significant moderator of death or morbidity score in regression analyses. Increased age (adjusted Odds Ratio=3.37, 95% CI=2.44–4.67), male gender (aOR=1.35, 95% CI=1.08-1.69), and number of comorbid conditions (aOR=1.28, 95% CI=1.01-1.63) were significantly associated with death. Significant factors in predicting total morbidity score were age (adjusted Multiplicative Effect=1.45; 95% CI=1.349-1.555) and gender (aME=1.17; 95% CI=1.109-1.243). The posterior probability that blood type influenced death was only 10%. Conclusions There is strong evidence that blood type was not a significant predictor of clinical course or death in patients hospitalized with COVID-19. Older age and male gender led to worse clinical outcomes and higher rates of death; older age, male gender, and comorbidities predicted a worse clinical course and higher morbidity score. Race was not a significant predictor of death in our population and was associated with an increased, albeit not significant, morbidity score.
Collapse
Affiliation(s)
- Katharine E Thomas
- Louisiana State University Health Science Center, Department of Hematology/Oncology, New Orleans, LA
| | - Amber Karamanis
- Louisiana State University School of Medicine, New Orleans, LA
| | - Erin Dauchy
- Louisiana State University Health Science Center, Department of Hematology/Oncology, New Orleans, LA
| | - Andrew G Chapple
- Biostatistics Program, School of Public Health, LSU Health Sciences Center, New Orleans, LA
| | - Michelle M Loch
- Louisiana State University Health Science Center, Department of Hematology/Oncology, New Orleans, LA.
| |
Collapse
|
9
|
Wang X, Hobbs B, Gandhi SJ, Muijs CT, Langendijk JA, Lin SH. Current status and application of proton therapy for esophageal cancer. Radiother Oncol 2021; 164:27-36. [PMID: 34534613 DOI: 10.1016/j.radonc.2021.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 08/11/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022]
Abstract
Esophageal cancer remains one of the leading causes of death from cancer across the world despite advances in multimodality therapy. Although early-stage disease can often be treated surgically, the current state of the art for locally advanced disease is concurrent chemoradiation, followed by surgery whenever possible. The uniform midline tumor location puts a strong importance on the need for precise delivery of radiation that would minimize dose to the heart and lungs, and the biophysical properties of proton beam makes this modality potential ideal for esophageal cancer treatment. This review covers the current state of knowledge of proton therapy for esophageal cancer, focusing on published retrospective single- and multi-institutional clinical studies, and emerging data from prospective clinical trials, that support the benefit of protons vs photon-based radiation in reducing postoperative complications, cardiac toxicity, and severe radiation induced immune suppression, which may improve survival outcomes for patients. In addition, we discuss the incorporation of immunotherapy to the curative management of esophageal cancers in the not-too-distant future. However, there is still a lack of high-level evidence to support proton therapy in the treatment of esophageal cancer, and proton therapy has its limitations in clinical application. It is expected to see the results of future large-scale randomized clinical trials and the continuous improvement of proton radiotherapy technology.
Collapse
Affiliation(s)
- Xin Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, China
| | - Brian Hobbs
- Department of Population Health, University of Texas, Austin, USA
| | - Saumil J Gandhi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Christina T Muijs
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Johannes A Langendijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
10
|
Thaker NG, Boyce-Fappiano D, Ning MS, Pasalic D, Guzman A, Smith G, Holliday EB, Incalcaterra J, Garden AS, Shaitelman SF, Gunn GB, Fuller CD, Blanchard P, Feeley TW, Kaplan RS, Frank SJ. Activity-Based Costing of Intensity-Modulated Proton versus Photon Therapy for Oropharyngeal Cancer. Int J Part Ther 2021; 8:374-382. [PMID: 34285963 PMCID: PMC8270081 DOI: 10.14338/ijpt-20-00042.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 01/11/2021] [Indexed: 11/29/2022] Open
Abstract
PURPOSE In value-based health care delivery, radiation oncologists need to compare empiric costs of care delivery with advanced technologies, such as intensity-modulated proton therapy (IMPT) and intensity-modulated radiation therapy (IMRT). We used time-driven activity-based costing (TDABC) to compare the costs of delivering IMPT and IMRT in a case-matched pilot study of patients with newly diagnosed oropharyngeal (OPC) cancer. MATERIALS AND METHODS We used clinicopathologic factors to match 25 patients with OPC who received IMPT in 2011-12 with 25 patients with OPC treated with IMRT in 2000-09. Process maps were created for each multidisciplinary clinical activity (including chemotherapy and ancillary services) from initial consultation through 1 month of follow-up. Resource costs and times were determined for each activity. Each patient-specific activity was linked with a process map and TDABC over the full cycle of care. All calculated costs were normalized to the lowest-cost IMRT patient. RESULTS TDABC costs for IMRT were 1.00 to 3.33 times that of the lowest-cost IMRT patient (mean ± SD: 1.65 ± 0.56), while costs for IMPT were 1.88 to 4.32 times that of the lowest-cost IMRT patient (2.58 ± 0.39) (P < .05). Although single-fraction costs were 2.79 times higher for IMPT than for IMRT (owing to higher equipment costs), average full cycle cost of IMPT was 1.53 times higher than IMRT, suggesting that the initial cost increase is partly mitigated by reductions in costs for other, non-RT supportive health care services. CONCLUSIONS In this matched sample, although IMPT was on average more costly than IMRT primarily owing to higher equipment costs, a subset of IMRT patients had similar costs to IMPT patients, owing to greater use of supportive care resources. Multidimensional patient outcomes and TDABC provide vital methodology for defining the value of radiation therapy modalities.
Collapse
Affiliation(s)
- Nikhil G. Thaker
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Arizona Oncology, The US Oncology Network, Tucson, AZ, USA
| | - David Boyce-Fappiano
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew S. Ning
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dario Pasalic
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexis Guzman
- The Institute for Cancer Care Innovation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grace Smith
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emma B. Holliday
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James Incalcaterra
- The Institute for Cancer Care Innovation, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adam S. Garden
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simona F. Shaitelman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G. Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C. David Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pierre Blanchard
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Steven J. Frank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Making Patient-Specific Treatment Decisions Using Prognostic Variables and Utilities of Clinical Outcomes. Cancers (Basel) 2021; 13:cancers13112741. [PMID: 34205968 PMCID: PMC8198909 DOI: 10.3390/cancers13112741] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/18/2021] [Accepted: 05/30/2021] [Indexed: 12/19/2022] Open
Abstract
We argue that well-informed patient-specific decision-making may be carried out as three consecutive tasks: (1) estimating key parameters of a statistical model, (2) using prognostic information to convert these parameters into clinically interpretable values, and (3) specifying joint utility functions to quantify risk-benefit trade-offs between clinical outcomes. Using the management of metastatic clear cell renal cell carcinoma as our motivating example, we explain the role of prognostic covariates that characterize between-patient heterogeneity in clinical outcomes. We show that explicitly specifying the joint utility of clinical outcomes provides a coherent basis for patient-specific decision-making.
Collapse
|
12
|
Nicholas O, Prosser S, Mortensen HR, Radhakrishna G, Hawkins MA, Gwynne SH. The Promise of Proton Beam Therapy for Oesophageal Cancer: A Systematic Review of Dosimetric and Clinical Outcomes. Clin Oncol (R Coll Radiol) 2021; 33:e339-e358. [PMID: 33931290 DOI: 10.1016/j.clon.2021.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/08/2021] [Accepted: 04/13/2021] [Indexed: 12/25/2022]
Abstract
AIMS Due to its physical advantages over photon radiotherapy, proton beam therapy (PBT) has the potential to improve outcomes from oesophageal cancer. However, for many tumour sites, high-quality evidence supporting PBT use is limited. We carried out a systematic review of published literature of PBT in oesophageal cancer to ascertain potential benefits of this technology and to gauge the current state-of-the-art. We considered if further evaluation of this technology in oesophageal cancer is desirable. MATERIALS AND METHODS A systematic literature search of Medline, Embase, Cochrane Library and Web of Science using structured search terms was carried out. Inclusion criteria included non-metastatic cancer, full articles and English language studies only. Articles deliberating technical aspects of PBT planning or delivery were excluded to maintain a clinical focus. Studies were divided into two sections: dosimetric and clinical studies; qualitatively synthesised. RESULTS In total, 467 records were screened, with 32 included for final qualitative synthesis. This included two prospective studies with the rest based on retrospective data. There was heterogeneity in treatment protocols, including treatment intent (neoadjuvant or definitive), dose, fractionation and chemotherapy used. Compared with photon radiotherapy, PBT seemed to reduce dose to organs at risk, especially lung and heart, although not for all reported parameters. Toxicity outcomes, including postoperative complications, were reduced compared with photon radiotherapy. Survival outcomes were reported to be at least comparable with photon radiotherapy. CONCLUSION There is a paucity of high-quality evidence supporting PBT use in oesophageal cancer. Wide variation in intent and treatment protocols means that the role and 'gold-standard' treatment protocol are yet to be defined. Current literature suggests significant benefit in terms of toxicity reduction, especially in the postoperative period, with comparable survival outcomes. PBT in oesophageal cancer holds significant promise for improving patient outcomes but requires robust systematic evaluation in prospective studies.
Collapse
Affiliation(s)
- O Nicholas
- South West Wales Cancer Centre, Swansea, UK; Swansea University Medical School, Swansea, UK.
| | - S Prosser
- South West Wales Cancer Centre, Swansea, UK
| | - H R Mortensen
- The Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | | | - M A Hawkins
- University College Hospital NHS Foundation Trust, London, UK
| | - S H Gwynne
- South West Wales Cancer Centre, Swansea, UK; Swansea University Medical School, Swansea, UK
| |
Collapse
|
13
|
Adverse Event Burden Score-A Versatile Summary Measure for Cancer Clinical Trials. Cancers (Basel) 2020; 12:cancers12113251. [PMID: 33158080 PMCID: PMC7694214 DOI: 10.3390/cancers12113251] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022] Open
Abstract
Simple Summary In cancer clinical trials, adverse event data are collected after every treatment cycle, using the Common Terminology Criteria for adverse events, which includes 837 terms. The vast number of potentially reportable adverse events over multiple treatment cycles makes summarizing and analyzing adverse event data challenging. The current standard reporting of adverse event data includes the frequency of the maximum (worst) grade of commonly occurring adverse events. In this article, we propose a single quantitative summary measure that incorporates both the frequency and the severity of multiple adverse events over time; the adverse event burden score. This score is a well-defined measure that enables statistical comparisons analogous to other quantitative endpoints in clinical trials. The adverse event burden score can readily accommodate different trial settings, diseases, and treatments, with diverse safety profiles. Abstract This article introduces the adverse event (AE) burden score. The AE burden by treatment cycle is a weighted sum of all grades and AEs that the patient experienced in a cycle. The overall AE burden score is the total AE burden the patient experienced across all treatment cycles. AE data from two completed Alliance multi-center randomized double-blind placebo-controlled trials, with different AE profiles (NCCTG 97-24-51: 176 patients, and A091105: 83 patients), were utilized for illustration. Results of the AE burden score analyses corroborated the trials’ primary results. In 97-24-51, the overall AE burden for patients on the treatment arm was 2.2 points higher than those on the placebo arm, with a higher AE burden for patients who went off treatment early due to AE. Similarly, in A091105, the overall AE burden was 1.6 points higher on the treatment arm. On the placebo arms, the AE burden in 97-24-51 remained constant over time; and increased in later cycles in A091105, likely attributable to the increase in disease morbidity. The AE burden score enables statistical comparisons analogous to other quantitative endpoints in clinical trials, and can readily accommodate different trial settings, diseases, and treatments, with diverse AE profiles.
Collapse
|
14
|
Milton DR, Lin SH, Hobbs BP. Comparing Radiation Modalities with Trimodality Therapy Using Total Toxicity Burden. Int J Radiat Oncol Biol Phys 2020; 107:1001-1005. [PMID: 32698967 DOI: 10.1016/j.ijrobp.2020.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/10/2020] [Accepted: 04/17/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Denái R Milton
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Brian P Hobbs
- Department of Quantitative Health Sciences, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
15
|
Simone CB. First Randomized Trial Supporting the Use of Proton Over Photon Chemoradiotherapy in Esophageal Cancer. J Clin Oncol 2020; 38:2952-2955. [PMID: 32706638 DOI: 10.1200/jco.20.01405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Charles B Simone
- Department of Radiation Oncology, New York Proton Center and Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
16
|
Zabor EC, Kaizer AM, Hobbs BP. Randomized Controlled Trials. Chest 2020; 158:S79-S87. [PMID: 32658656 PMCID: PMC8176647 DOI: 10.1016/j.chest.2020.03.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/19/2019] [Accepted: 03/09/2020] [Indexed: 12/18/2022] Open
Abstract
Randomized controlled trials (RCTs) are considered the highest level of evidence to establish causal associations in clinical research. There are many RCT designs and features that can be selected to address a research hypothesis. Designs of RCTs have become increasingly diverse as new methods have been proposed to evaluate increasingly complex scientific hypotheses. This article reviews the principles and general concepts behind many common RCT designs and introduces newer designs that have been proposed, such as adaptive and cluster randomized trials. A focus on the many choices for randomization within an RCT is described, along with their potential tradeoffs. To illustrate their diversity, examples of RCTs from the literature are provided. Statistical considerations, such as power and type I error rates, are discussed with the intention of providing practical guidance about how to specify study hypotheses that address the scientific question while being statistically appropriate. Finally, the freely available Consolidated Standards of Reporting Trials guidelines and US Food and Drug Administration guidance documents are introduced, along with a set of guidelines one should consider when planning an RCT or reviewing RCTs submitted for publication in peer-reviewed academic journals.
Collapse
Affiliation(s)
- Emily C Zabor
- Department of Quantitative Health Sciences & Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH.
| | - Alexander M Kaizer
- Department of Quantitative Health Sciences & Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Brian P Hobbs
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
17
|
Sisk BA, Dubois J, Hobbs BP, Kodish E. Reprioritizing Risk and Benefit: The Future of Study Design in Early-Phase Cancer Research. Ethics Hum Res 2020; 41:2-11. [PMID: 31743629 DOI: 10.1002/eahr.500033] [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] [Indexed: 02/04/2023]
Abstract
The scientific purpose of phase I trials is to determine the maximum tolerated dose and/or optimal biological dose of experimental agents. Yet most participants in phase I oncology trials enroll hoping for direct medical benefit. The most common phase I trial designs use low starting doses and escalate cautiously in a "risk-escalation" model focused on minimizing risk for each participant. This approach ensures that a proportion of subjects will likely not receive any benefit, even if the intervention proves to be successful at appropriate doses. In this article, we propose that trial designs should employ dosing strategies that increase chances of providing benefit if the investigational agent should prove to be successful while limiting risk to reasonable levels. We then describe how adaptive trial designs can facilitate refined dose optimization based on both therapeutic benefit and toxicity, which can simultaneously decrease the risk of harm while increasing the chances of benefit.
Collapse
Affiliation(s)
- Bryan Anthony Sisk
- Clinical fellow in pediatric hematology/oncology in the Department of Pediatrics at Washington University School of Medicine
| | - James Dubois
- Professor in the Department of Medicine at Washington University School of Medicine
| | - Brian P Hobbs
- Associate staff member in the Department of Quantitative Health Sciences in the Lerner Research Institute at the Cleveland Clinic
| | - Eric Kodish
- Professor of pediatrics, oncology, and bioethics at Case Western Reserve and Cleveland Clinic Lerner College of Medicine
| |
Collapse
|
18
|
Lin SH, Hobbs BP, Verma V, Tidwell RS, Smith GL, Lei X, Corsini EM, Mok I, Wei X, Yao L, Wang X, Komaki RU, Chang JY, Chun SG, Jeter MD, Swisher SG, Ajani JA, Blum-Murphy M, Vaporciyan AA, Mehran RJ, Koong AC, Gandhi SJ, Hofstetter WL, Hong TS, Delaney TF, Liao Z, Mohan R. Randomized Phase IIB Trial of Proton Beam Therapy Versus Intensity-Modulated Radiation Therapy for Locally Advanced Esophageal Cancer. J Clin Oncol 2020; 38:1569-1579. [PMID: 32160096 DOI: 10.1200/jco.19.02503] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Whether dosimetric advantages of proton beam therapy (PBT) translate to improved clinical outcomes compared with intensity-modulated radiation therapy (IMRT) remains unclear. This randomized trial compared total toxicity burden (TTB) and progression-free survival (PFS) between these modalities for esophageal cancer. METHODS This phase IIB trial randomly assigned patients to PBT or IMRT (50.4 Gy), stratified for histology, resectability, induction chemotherapy, and stage. The prespecified coprimary end points were TTB and PFS. TTB, a composite score of 11 distinct adverse events (AEs), including common toxicities as well as postoperative complications (POCs) in operated patients, quantified the extent of AE severity experienced over the duration of 1 year following treatment. The trial was conducted using Bayesian group sequential design with three planned interim analyses at 33%, 50%, and 67% of expected accrual (adjusted for follow-up). RESULTS This trial (commenced April 2012) was approved for closure and analysis upon activation of NRG-GI006 in March 2019, which occurred immediately prior to the planned 67% interim analysis. Altogether, 145 patients were randomly assigned (72 IMRT, 73 PBT), and 107 patients (61 IMRT, 46 PBT) were evaluable. Median follow-up was 44.1 months. Fifty-one patients (30 IMRT, 21 PBT) underwent esophagectomy; 80% of PBT was passive scattering. The posterior mean TTB was 2.3 times higher for IMRT (39.9; 95% highest posterior density interval, 26.2-54.9) than PBT (17.4; 10.5-25.0). The mean POC score was 7.6 times higher for IMRT (19.1; 7.3-32.3) versus PBT (2.5; 0.3-5.2). The posterior probability that mean TTB was lower for PBT compared with IMRT was 0.9989, which exceeded the trial's stopping boundary of 0.9942 at the 67% interim analysis. The 3-year PFS rate (50.8% v 51.2%) and 3-year overall survival rates (44.5% v 44.5%) were similar. CONCLUSION For locally advanced esophageal cancer, PBT reduced the risk and severity of AEs compared with IMRT while maintaining similar PFS.
Collapse
Affiliation(s)
- Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brian P Hobbs
- Quantitative Health Sciences, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, PA
| | - Rebecca S Tidwell
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Grace L Smith
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiudong Lei
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Erin M Corsini
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Isabel Mok
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiong Wei
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luyang Yao
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xin Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Ritsuko U Komaki
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Joe Y Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen G Chun
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Melenda D Jeter
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen G Swisher
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mariela Blum-Murphy
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ara A Vaporciyan
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Reza J Mehran
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Albert C Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Saumil J Gandhi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wayne L Hofstetter
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Thomas F Delaney
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Zhongxing Liao
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Radhe Mohan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
19
|
Thall PF. Bayesian Utility-Based Designs for Subgroup-Specific Treatment Comparison and Early-Phase Dose Optimization in Oncology Clinical Trials. JCO Precis Oncol 2019; 3:1800379. [PMID: 33015521 DOI: 10.1200/po.18.00379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2019] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Despite the fact that almost any sample of patients with a particular disease is heterogeneous, most clinical trial designs ignore the possibility that treatment or dose effects may differ between prognostic or biologically defined subgroups. This article reviews two clinical trial designs that make subgroup-specific decisions and compares each to a simpler design that ignores patient heterogeneity. The purpose is to illustrate the benefits of accounting prospectively for treatment-subgroup interactions and how utilities may be used to quantify risk-benefit trade-offs. METHODS Two Bayesian clinical trial designs that perform subgroup-specific decision making and inference based on elicited utilities of patient outcomes are reviewed. The first is a randomized comparative trial of nutritional prehabilitation for patients undergoing esophageal resection that has two prognostic subgroups and is based on postoperative morbidity score. The second is a sequentially adaptive trial of natural killer cells for treating hematologic malignancies that is based on five time-to-event outcomes and that performs safety monitoring and optimizes cell dose within six disease subgroups. Computer simulations under a range of different scenarios are presented for each design to establish its operating characteristics and compare it to a more conventional design that ignores patient heterogeneity. RESULTS Each design has attractive operating characteristics, is greatly superior to a simplified design that ignores patient subgroups, is robust to deviations from its assumed statistical model, and is feasible to use for conducting trials. CONCLUSION Bayesian designs that make subgroup-specific decisions in randomized comparative trials or sequentially adaptive early-phase dose-finding trials are superior to designs that ignore patient heterogeneity. Using elicited utilities of complex patient outcomes to quantify risk-benefit trade-offs provides a practical and ethical basis for decision making and treatment evaluation in clinical trials.
Collapse
Affiliation(s)
- Peter F Thall
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
20
|
Ma J, Stingo FC, Hobbs BP. Bayesian personalized treatment selection strategies that integrate predictive with prognostic determinants. Biom J 2019; 61:902-917. [PMID: 30786040 PMCID: PMC7341533 DOI: 10.1002/bimj.201700323] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 09/28/2018] [Accepted: 12/04/2018] [Indexed: 01/13/2023]
Abstract
The evolution of "informatics" technologies has the potential to generate massive databases, but the extent to which personalized medicine may be effectuated depends on the extent to which these rich databases may be utilized to advance understanding of the disease molecular profiles and ultimately integrated for treatment selection, necessitating robust methodology for dimension reduction. Yet, statistical methods proposed to address challenges arising with the high-dimensionality of omics-type data predominately rely on linear models and emphasize associations deriving from prognostic biomarkers. Existing methods are often limited for discovering predictive biomarkers that interact with treatment and fail to elucidate the predictive power of their resultant selection rules. In this article, we present a Bayesian predictive method for personalized treatment selection that is devised to integrate both the treatment predictive and disease prognostic characteristics of a particular patient's disease. The method appropriately characterizes the structural constraints inherent to prognostic and predictive biomarkers, and hence properly utilizes these complementary sources of information for treatment selection. The methodology is illustrated through a case study of lower grade glioma. Theoretical considerations are explored to demonstrate the manner in which treatment selection is impacted by prognostic features. Additionally, simulations based on an actual leukemia study are provided to ascertain the method's performance with respect to selection rules derived from competing methods.
Collapse
Affiliation(s)
- Junsheng Ma
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center Houston, Texas 77030
| | - Francesco C. Stingo
- Department of Statistica, Informatica, Applicazioni “G.Parenti”, University of Florence, Florence, 50134, Italy
| | - Brian P. Hobbs
- Quantitative Health Sciences and The Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
21
|
Badiyan SN, Hallemeier CL, Lin SH, Hall MD, Chuong MD. Proton beam therapy for gastrointestinal cancers: past, present, and future. J Gastrointest Oncol 2018; 9:962-971. [PMID: 30505599 DOI: 10.21037/jgo.2017.11.07] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite the conformality of modern X-ray therapy limiting high dose received by normal tissues the physical properties of X-rays make it impossible to avoid dose being delivered distal to the target. This "exit dose" is likely clinically significant especially for patients with gastrointestinal (GI) cancers when considering that even low dose received by the heart, lungs, bowel, and other radiosensitive structures can lead to morbidity and even may affect long-term tumor control. In contrast, proton beam therapy (PBT) delivers no "exit dose" and a growing body of literature suggests that this may improve clinical outcomes by reducing toxicity and even allowing for safe dose intensification to enhance tumor control. While there are not yet robust prospective data demonstrating the role of PBT for GI cancers, emerging retrospective data provide a strong rationale for continued study of how PBT may improve the therapeutic ratio for these patients. Here we review these data as well as discuss ongoing clinical trials of PBT for GI cancers.
Collapse
Affiliation(s)
- Shahed N Badiyan
- Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, MD, USA
| | | | - Steven H Lin
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew D Hall
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Michael D Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| |
Collapse
|
22
|
Blanchard P, Gunn GB, Lin A, Foote RL, Lee NY, Frank SJ. Proton Therapy for Head and Neck Cancers. Semin Radiat Oncol 2018; 28:53-63. [PMID: 29173756 DOI: 10.1016/j.semradonc.2017.08.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Because of its sharp lateral penumbra and steep distal fall-off, proton therapy offers dosimetric advantages over photon therapy. In head and neck cancer, proton therapy has been used for decades in the treatment of skull-base tumors. In recent years the use of proton therapy has been extended to numerous other disease sites, including nasopharynx, oropharynx, nasal cavity and paranasal sinuses, periorbital tumors, skin, and salivary gland, or to reirradiation. The aim of this review is to present the physical properties and dosimetric benefit of proton therapy over advanced photon therapy; to summarize the clinical benefit described for each disease site; and to discuss issues of patient selection and cost-effectiveness.
Collapse
Affiliation(s)
- Pierre Blanchard
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Radiation Oncology, Institut Gustave Roussy, Villejuif, France
| | - Gary Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexander Lin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA
| | - Robert L Foote
- Departments of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steven J Frank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
23
|
Shohoudi A, Stephens DA, Khairy P. Bayesian adaptive trials for rare cardiovascular conditions. Future Cardiol 2018; 14:143-150. [PMID: 29405070 DOI: 10.2217/fca-2017-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Escalating costs of cardiovascular trials are limiting medical innovations, prompting the development of more efficient and flexible study designs. The Bayesian paradigm offers a framework conducive to adaptive trial methodologies and is well suited for the study of small populations. Bayesian adaptive trials provide a statistical structure for combining prior information with accumulating data to compute probabilities of unknown quantities of interest. Adaptive design features are useful in modifying randomization schemes, adjusting sample sizes and providing continuous surveillance to guide decisions on dropping study arms or premature trial interruption. Advantages include greater efficiency, minimization of risks, inclusion of knowledge as it is generated, cost savings and more intuitive interpretability. Extensive high-level computations are facilitated by an expanding armamentarium of available tools.
Collapse
Affiliation(s)
- Azadeh Shohoudi
- Montreal Health Innovations Coordinating Center (MHICC), Montreal, Canada
| | - David A Stephens
- Department of Mathematics & Statistics, McGill University, Montreal, Canada
| | - Paul Khairy
- Montreal Health Innovations Coordinating Center (MHICC), Montreal, Canada.,Department of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| |
Collapse
|
24
|
Murray TA, Thall PF, Yuan Y, McAvoy S, Gomez DR. Robust treatment comparison based on utilities of semi-competing risks in non-small-cell lung cancer. J Am Stat Assoc 2017; 112:11-23. [PMID: 28943681 PMCID: PMC5607962 DOI: 10.1080/01621459.2016.1176926] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 01/01/2016] [Indexed: 12/25/2022]
Abstract
A design is presented for a randomized clinical trial comparing two second-line treatments, chemotherapy versus chemotherapy plus reirradiation, for treatment of recurrent non-small-cell lung cancer. The central research question is whether the potential efficacy benefit that adding reirradiation to chemotherapy may provide justifies its potential for increasing the risk of toxicity. The design uses two co-primary outcomes: time to disease progression or death, and time to severe toxicity. Because patients may be given an active third-line treatment at disease progression that confounds second-line treatment effects on toxicity and survival following disease progression, for the purpose of this comparative study follow-up ends at disease progression or death. In contrast, follow-up for disease progression or death continues after severe toxicity, so these are semi-competing risks. A conditionally conjugate Bayesian model that is robust to misspecification is formulated using piecewise exponential distributions. A numerical utility function is elicited from the physicians that characterizes desirabilities of the possible co-primary outcome realizations. A comparative test based on posterior mean utilities is proposed. A simulation study is presented to evaluate test performance for a variety of treatment differences, and a sensitivity assessment to the elicited utility function is performed. General guidelines are given for constructing a design in similar settings, and a computer program for simulation and trial conduct is provided.
Collapse
Affiliation(s)
| | - Peter F Thall
- Department of Biostatistics, MD Anderson Cancer Center
| | - Ying Yuan
- Department of Biostatistics, MD Anderson Cancer Center
| | - Sarah McAvoy
- Department of Radiation Oncology, MD Anderson Cancer Center
| | - Daniel R Gomez
- Department of Radiation Oncology, MD Anderson Cancer Center
| |
Collapse
|
25
|
Zhu H, Yu Q, Mercante DE. A Bayesian sequential design with binary outcome. Pharm Stat 2017; 16:192-200. [PMID: 28251815 DOI: 10.1002/pst.1805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/22/2016] [Accepted: 02/03/2017] [Indexed: 11/09/2022]
Abstract
Several researchers have proposed solutions to control type I error rate in sequential designs. The use of Bayesian sequential design becomes more common; however, these designs are subject to inflation of the type I error rate. We propose a Bayesian sequential design for binary outcome using an alpha-spending function to control the overall type I error rate. Algorithms are presented for calculating critical values and power for the proposed designs. We also propose a new stopping rule for futility. Sensitivity analysis is implemented for assessing the effects of varying the parameters of the prior distribution and maximum total sample size on critical values. Alpha-spending functions are compared using power and actual sample size through simulations. Further simulations show that, when total sample size is fixed, the proposed design has greater power than the traditional Bayesian sequential design, which sets equal stopping bounds at all interim analyses. We also find that the proposed design with the new stopping for futility rule results in greater power and can stop earlier with a smaller actual sample size, compared with the traditional stopping rule for futility when all other conditions are held constant. Finally, we apply the proposed method to a real data set and compare the results with traditional designs.
Collapse
Affiliation(s)
- Han Zhu
- Pharmaceutical Product Development, LLC., Austin, TX, USA
| | - Qingzhao Yu
- Biostatistics Program, School of Public Health, LSUHSC, New Orleans, LA, USA
| | - Donald E Mercante
- Biostatistics Program, School of Public Health, LSUHSC, New Orleans, LA, USA
| |
Collapse
|
26
|
Ehrmann S, Quartin A, Hobbs BP, Robert-Edan V, Cely C, Bell C, Lyons G, Pham T, Schein R, Geng Y, Lakhal K, Ng CS. Contrast-associated acute kidney injury in the critically ill: systematic review and Bayesian meta-analysis. Intensive Care Med 2017; 43:785-794. [PMID: 28197679 DOI: 10.1007/s00134-017-4700-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/27/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE Critically ill patients, among whom acute kidney injury is common, are often considered particularly vulnerable to iodinated contrast medium nephrotoxicity. However, the attributable incidence remains uncertain given the paucity of observational studies including a control group. This study assessed acute kidney injury incidence attributable to iodinated contrast media in critically ill patients based on new data accounting for sample and effect size and including a control group. METHODS Systematic review of studies measuring incidence of acute kidney injury in critically ill patients following contrast medium exposure compared to matched unexposed patients. Patient-level meta-analysis implementing a Bayesian nested mixed effects multiple logistic regression model. RESULTS Ten studies were identified; only four took into account the baseline acute kidney injury risk, three by patient matching (560 patients). Objective meta-analysis of these three studies (vague and impartial a priori hypothesis concerning attributable acute kidney injury risk) did not find that iodinated contrast media increased the incidence of acute kidney injury (odds ratio 0.95, 95% highest posterior density interval 0.45-1.62). Bayesian analysis demonstrated that, to conclude in favor of a statistically significant incidence of acute kidney injury attributable to contrast media despite this observed lack of association, one's a priori belief would have to be very strongly biased, assigning to previous uncontrolled reports 3-12 times the weight of evidence strength provided by the matched studies including a control group. CONCLUSIONS Meta-analysis of matched cohort studies of iodinated contrast medium exposure does not support a significant incidence of acute kidney injury attributable to iodinated contrast media in critically ill patients.
Collapse
Affiliation(s)
- Stephan Ehrmann
- Médecine Intensive Réanimation, Centre Hospitalier Régional et Universitaire de Tours, 37044, Tours, France. .,Faculté de Médecine, Université François Rabelais, Tours, France.
| | - Andrew Quartin
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Brian P Hobbs
- Department of Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vincent Robert-Edan
- Réanimation Chirurgicale Polyvalente, Service d'Anesthésie-Réanimation, Hôpital Laënnec, Centre Hospitalier Universitaire, Nantes, France
| | - Cynthia Cely
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Cynthia Bell
- Division of Pediatric Nephrology and Hypertension, University of Texas Health Science Center-Houston, Houston, TX, USA
| | - Genevieve Lyons
- Department of Biostatistics, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tai Pham
- Réanimation et USC Médico-chirurgicale, Hôpital Tenon, Assistance Publique, Hôpitaux de Paris, 75970, Paris, France.,INSERM UMR 1153, ECSTRA Team, Paris, France.,Saint Michael's Hospital, Interdepartmental Division of Critical Care, University of Toronto, Toronto, Canada
| | - Roland Schein
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Yimin Geng
- Research Medical Library, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Karim Lakhal
- Réanimation Chirurgicale Polyvalente, Service d'Anesthésie-Réanimation, Hôpital Laënnec, Centre Hospitalier Universitaire, Nantes, France
| | - Chaan S Ng
- Department of Radiology, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
27
|
Xu Y, Müller P, Wahed AS, Thall P. Rejoinder. J Am Stat Assoc 2016. [DOI: 10.1080/01621459.2016.1200917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Yanxun Xu
- Division of Statistics and Scientific Computing, The University of Texas at Austin, Austin, TX, USA
| | - Peter Müller
- Department of Mathematics, The University of Texas at Austin, Austin, TX, USA
| | - Abdus S. Wahed
- Epidemiology Data Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter Thall
- M. D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|