1
|
Gueguen L, Olgiati L, Brutti-Mairesse C, Sans A, Le Texier V, Verlingue L. A prospective pragmatic evaluation of automatic trial matching tools in a molecular tumor board. NPJ Precis Oncol 2025; 9:28. [PMID: 39870746 PMCID: PMC11772588 DOI: 10.1038/s41698-025-00806-y] [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: 10/22/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
Publicly available trial matching tools can improve the access to therapeutic innovations, but errors may expose to over-solicitation and disappointment. We performed a pragmatic non-interventional prospective evaluation on sequential patients at the Molecular Tumor Board of Centre Leon Berard. During 10 weeks in 2024, we analysed 157 patients with four clinical trial matching tools from the 19 screened: Klineo, ScreenAct, Trialing and DigitalECMT. Each patient had 2.19 trials proposed on average, and 38% had no trials suggested. The mean performances were precision = 0.33, recall = 0.32, AP@3 = 0.45, and NDCG@3 = 0.34. Using all the tools can increase to 26% the clinical trial options. The most frequent error concerned the type of gene variants required by the selection criteria. We showed that using a Large Language Model on the patients' molecular reports could improve the performance by up to 5%. We recommend that experts supervise the results and we advocate for improved technologies.
Collapse
Affiliation(s)
| | | | | | | | | | - Loic Verlingue
- CRCL, Centre Léon Bérard, Lyon, France.
- Phase 1 Clinical Unit, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
2
|
Guerra M, Alouani E, Hueso T, Ouali K, Danu A, Hollebecque A, Bahleda R, Willekens C, Gazzah A, Baldini C, Postel‐Vinay S, Micol J, Massard C, De Botton S, Ribrag V, Michot J. Relevance, Risks, and Benefits of Early-Phases Clinical Trials Participations for Patients With Hematological Malignancies From 2008 to 2023. Eur J Haematol 2025; 114:89-97. [PMID: 39305190 PMCID: PMC11613620 DOI: 10.1111/ejh.14307] [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: 05/11/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Early-phases clinical trials (Phases 1 and 2) have evolved from a traditional assessment of toxicity to an adaptive approach based on patients' medical needs and access to effective new therapies. The global risks, benefits, and relevance of early-phases clinical trials participation for patients with hematological malignancies remain poorly evaluated. PATIENTS AND METHODS All early-phases clinical trials participations for patients with hematological malignancies, from 2008 to 2023, in a tertiary academic center in Europe, were reviewed. Patient's demographics, tumor type categories, therapeutic responses, mortality, overall survival (OS), and investigational product (IP) were assessed. RESULTS Over the period 2008-2023, 736 patients participating in 92 different early-phases clinical trials, were analyzed. The most common tumor categories were diffuse large B-cell lymphoma (n = 253; 34.4%), acute myeloid leukemia/myelodysplastic syndrome (n = 164; 22.3%) and multiple myeloma (n = 100; 13.6%). The median OS was 14.8 (95% CI: 12.4-17.9) months and response rate 31.9%, including complete responses in 13.5% of patients. By tumor categories, the highest and lowest median duration of OS were observed for patients with Hodgkin lymphoma (99.8; [95% CI: 47.0-not reached] months) and peripheral T-cell lymphoma (8.9 [95% CI: 5.3-12.0] months), respectively. The on-protocol and treatment-related mortality rates were 5.43% and 0.54%, respectively. Overall response rate was 29.1% including 13.5% of complete response. Overall, 202 (27.5%) patients received an IP later approved by the health authorities, and those patients had better OS (18.2 months vs. 12.1 months HR: 1.160 [95% CI; 0.6977-1.391], p = 0.0283). CONCLUSION In conclusion, patients with hematologic malignancies who have participated in early-phases clinical trials over the past 15 years have achieved variable therapeutic response rates, acceptable risk/benefit ratio and potentially significant therapeutic advantages. This study provides framework material for hematologists to further discuss clinical trial participation with their patients.
Collapse
Affiliation(s)
- Matteo Guerra
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | - Emily Alouani
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | - Thomas Hueso
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | - Kaissa Ouali
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | - Alina Danu
- Département d'HématologieGustave RoussyVillejuifFrance
| | - Antoine Hollebecque
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | - Rastislav Bahleda
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | | | - Anas Gazzah
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | - Capucine Baldini
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
| | - Sophie Postel‐Vinay
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
- INSERM U981Gustave RoussyVillejuifFrance
| | | | - Christophe Massard
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
- Université Paris‐SaclayVillejuifFrance
| | - Stéphane De Botton
- Département d'HématologieGustave RoussyVillejuifFrance
- Université Paris‐SaclayVillejuifFrance
- INSERM U1170Gustave RoussyVillejuifFrance
| | - Vincent Ribrag
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
- Département d'HématologieGustave RoussyVillejuifFrance
- INSERM U1170Gustave RoussyVillejuifFrance
| | - Jean‐Marie Michot
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP)Gustave RoussyVillejuifFrance
- Département d'HématologieGustave RoussyVillejuifFrance
- INSERM U1170Gustave RoussyVillejuifFrance
| |
Collapse
|
3
|
Blanter J, Van Hyfte G, Ahmad M, Xu S, Hapanowicz O, Fazilov G, Lu A, Lucas N, Wu K, Shelton G, DeMerchant M, Lachowicz M, Kier M, Werner M, Eder JP, Galsky MD, Marron TU, Smith CB, LoRusso PM, Hofstatter E, Doroshow DB. Financial Toxicity Among Patients With Advanced Solid Tumors Participating in Early-Phase Clinical Trials. JCO Oncol Pract 2024:OP2400293. [PMID: 39661920 DOI: 10.1200/op.24.00293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/03/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
PURPOSE Financial toxicity (FT) adversely influences patient quality of life and is a barrier to clinical trial enrollment. Early-phase clinical trials (EPCTs) recruit patients who may have high baseline FT and require additional visits and procedures, potentially increasing FT. METHODS In this prospective survey study, we sought to assess FT at baseline and after 2 months among patients with advanced solid malignancies participating in EPCTs. Participants were age 18 years and older, were English-speaking, and were treated at the Yale Cancer Center (Yale) and the Tisch Cancer Institute at Mount Sinai (Mount Sinai). At the time of consent and 2 months later, patients completed a sociodemographic questionnaire as well as the 11-item validated Comprehensive Score for Financial Toxicity (COST) instrument. Primary outcomes were baseline COST score and change in COST score from baseline to 2 months. Lower score is associated with higher FT. RESULTS One hundred forty-six patients completed survey 1. Mean age was 61.5 years and 50.7% were male. The most common histologies were lung (17.8%), colorectal (16.4%), and breast (12.3%) cancers. Mean baseline COST score was 22.12 (standard deviation, 8.01). FT was associated with being a primary wage earner (P = .044) and inversely associated with age (P = .025). Seventy-one patients completed survey 2. Among 71 patients who completed both surveys, there was no significant difference in COST score between surveys 1 and 2 (P = .28). Race, ethnicity, education, and household income were not associated with baseline COST score or change in FT. CONCLUSION Moderate FT was reported at baseline and on trial among EPCT participants. No change in FT was observed between time points. Additional efforts should be made to decrease FT associated with EPCT participation to maximize access to novel therapies.
Collapse
Affiliation(s)
- Julia Blanter
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Maham Ahmad
- Yale University School of Medicine, New Haven, CT
| | - Suzanne Xu
- Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | - Ashley Lu
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Natalie Lucas
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kathy Wu
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gary Shelton
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | - Melanie Kier
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michael Werner
- Zucker School of Medicine at Hofstra/Northwell Health, New Hyde Park, NY
| | - Joseph P Eder
- Yale Cancer Center, New Haven, CT
- Parthenon Therapeutics
| | | | | | | | | | | | | |
Collapse
|
4
|
Cullum S, Vang H, Glover M, Alammarah H, Morton H, Pham N, Rahman M, Khan SA. Case report: Dramatic impact of DNA next generation sequencing results using specific targeted therapies- ALK and PIK3CA. Front Oncol 2024; 14:1462930. [PMID: 39659799 PMCID: PMC11628483 DOI: 10.3389/fonc.2024.1462930] [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: 07/10/2024] [Accepted: 10/21/2024] [Indexed: 12/12/2024] Open
Abstract
In the era of targeted therapies, the clinical importance and utility of next-generation sequencing (NGS) has expanded significantly. Owing to the relative ease and financial feasibility of NGS, the use of personalized treatment strategies has the potential to revolutionize cancer care. In this case report, we explored the use of NGS in salivary gland carcinoma (SGC) and spindle cell neoplasm of the scalp. In our patient with SGC, NGS revealed a GPHN-ALK variant that allowed off-label treatment with alectinib, with a remarkable response in primary and metastatic foci. Similarly, the use of NGS in a cutaneous neoplasm in which no definitive diagnosis could be reached by pathology and which had progressed through standard of care treatment elucidated a PIK3CA mutation in which alpelisib was added and ultimately halted POD. Here, we discuss the use of NGS, future projections, and our recommendations.
Collapse
Affiliation(s)
- Stav Cullum
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Hlu Vang
- Department of Otolaryngology/Head & Neck Surgery Divisions, Stanford University, Stanford, CA, United States
| | - Michael Glover
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Howra Alammarah
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Heather Morton
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| | - Nancy Pham
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Mobeen Rahman
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Saad A. Khan
- Division of Oncology, Department of Medicine, Stanford Cancer Institute and Stanford University, Stanford, CA, United States
| |
Collapse
|
5
|
Subbiah V, Kurzrock R. The best management for most patients with incurable cancer is on a clinical trial. Ann Oncol 2024:S0923-7534(24)04913-5. [PMID: 39550034 DOI: 10.1016/j.annonc.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Affiliation(s)
- V Subbiah
- Sarah Cannon Research Institute, Nashville.
| | - R Kurzrock
- Genomic Sciences and Precision Medicine Center, and Medical College of Wisconsin Cancer Center, Milwaukee, USA; WIN Consortium, Paris, France; University of Nebraska, Lincoln, USA
| |
Collapse
|
6
|
Ghosh S, Abushukair HM, Ganesan A, Pan C, Naqash AR, Lu K. Harnessing explainable artificial intelligence for patient-to-clinical-trial matching: A proof-of-concept pilot study using phase I oncology trials. PLoS One 2024; 19:e0311510. [PMID: 39446771 PMCID: PMC11500892 DOI: 10.1371/journal.pone.0311510] [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: 01/24/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
This study aims to develop explainable AI methods for matching patients with phase 1 oncology clinical trials using Natural Language Processing (NLP) techniques to address challenges in patient recruitment for improved efficiency in drug development. A prototype system based on modern NLP techniques has been developed to match patient records with phase 1 oncology clinical trial protocols. Four criteria are considered for the matching: cancer type, performance status, genetic mutation, and measurable disease. The system outputs a summary matching score along with explanations of the evidence. The outputs of the AI system were evaluated against the ground truth matching results provided by the domain expert on a dataset of twelve synthesized dummy patient records and six clinical trial protocols. The system achieved a precision of 73.68%, sensitivity/recall of 56%, accuracy of 77.78%, and specificity of 89.36%. Further investigation into the misclassified cases indicated that ambiguity of abbreviation and misunderstanding of context are significant contributors to errors. The system found evidence of no matching for all false positive cases. To the best of our knowledge, no system in the public domain currently deploys an explainable AI-based approach to identify optimal patients for phase 1 oncology trials. This initial attempt to develop an AI system for patients and clinical trial matching in the context of phase 1 oncology trials showed promising results that are set to increase efficiency without sacrificing quality in patient-trial matching.
Collapse
Affiliation(s)
- Satanu Ghosh
- Department of Computer Science, University of New Hampshire, Durham, New Hampshire, United States of America
| | | | - Arjun Ganesan
- School of Computer Science, University of Oklahoma Norman Campus, Norman, Oklahoma, United States of America
| | - Chongle Pan
- School of Computer Science, University of Oklahoma Norman Campus, Norman, Oklahoma, United States of America
| | - Abdul Rafeh Naqash
- Medical Oncology/ TSET Phase 1 Program, Stephenson Cancer Center, The University of Oklahoma Health Science Campus, Oklahoma City, Oklahoma, United States of America
| | - Kun Lu
- School of Library and Information Studies, University of Oklahoma Norman Campus, Norman, Oklahoma, United States of America
| |
Collapse
|
7
|
Iskander R, Magnan Robart A, Moyer H, Nipp R, Gupta A, Kimmelman J. Time Burdens for Participants With Advanced Cancer in Phase I Trials: A Cross-Sectional Study. JCO Oncol Pract 2024:OP2400334. [PMID: 39353144 DOI: 10.1200/op.24.00334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Participating in phase I cancer clinical trials often entails extra visits and procedures. We describe the planned time and procedures associated with phase I trial participation. METHODS We searched ClinicalTrials.gov for phase I cancer trials of new drugs with assessment schedules and results posted between 2020 and 2022. Trials were included if participants had advanced or metastatic disease. Our primary analysis measured the number of planned research days (PRDs; each day a clinic visit is required) per participant up to the first month of trial participation and for the entire trial duration. Secondarily, we estimated the number of research procedures. RESULTS Our sample included 71 phase I trials comprising 302 cohorts. These trials enrolled 3,904 participants; the median participation duration was 2.5 months. During screening and up to the first month of participation, the median PRDs per participant was 7 (IQR, 7-10). Across the entire trial, the median PRDs per participant was 4.5 days per month (IQR, 3.30-6.20). Participants spent 15% of trial days attending planned appointments. Per trial cohort, participants were given a median of 8 (IQR, 7-11) physical examinations, 6 (IQR, 3-10) infusions, 6 (IQR, 3-12) electrocardiograms, and 1 (IQR, 1-3) biopsy. CONCLUSION Participants commit a substantial amount of time to planned visits in phase I cancer trials, especially in the first month. Overall, they invest 15% of trial days attending planned research activities. These estimates provide a lower bound to the time participants in phase I trials donate to drug development, as our analysis excluded unplanned visits.
Collapse
Affiliation(s)
| | | | | | - Ryan Nipp
- University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Oklahoma City, OK
| | | | | |
Collapse
|
8
|
Ilié M, Heeke S, Horgan D, Hofman P. Navigating Change in Tumor Naming: Exploring the Complexities and Considerations of Shifting Toward Molecular Classifications. J Clin Oncol 2024; 42:3183-3186. [PMID: 38935877 DOI: 10.1200/jco.24.00323] [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: 02/15/2024] [Revised: 03/21/2024] [Accepted: 05/08/2024] [Indexed: 06/29/2024] Open
Abstract
Navigating change in tumor naming. Balance organ-based and molecular classifications for optimal treatment.
Collapse
Affiliation(s)
- Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Hospital-Integrated Biobank (BB-0033-00025), IHU RespirERA, FHU OncoAge, University Hospital Centre Nice, University Côte d'Azur, Nice, France
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Denis Horgan
- European Alliance for Personalised Medicine, Brussels, Belgium
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Hospital-Integrated Biobank (BB-0033-00025), IHU RespirERA, FHU OncoAge, University Hospital Centre Nice, University Côte d'Azur, Nice, France
| |
Collapse
|
9
|
Durbin SM, Lundquist DM, Pelletier A, Jimenez R, Petrillo L, Kim J, Lynch K, Healy M, Johnson A, Ollila N, Yalala V, Malowitz B, Kehlmann A, Chevalier N, Turbini V, Bame V, Heldreth H, Silva J, McIntyre C, Juric D, Nipp RD. Time Toxicity Experienced by Early-Phase Cancer Clinical Trial Participants. JCO Oncol Pract 2024; 20:1252-1262. [PMID: 38857457 DOI: 10.1200/op.23.00811] [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: 12/18/2023] [Revised: 03/12/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024] Open
Abstract
PURPOSE Early-phase clinical trials (EP-CTs) are designed to determine optimal dosing, tolerability, and preliminary activity of novel cancer therapeutics. Little is known about the time that patients spend interacting with the health care system (eg, time toxicity) while participating in these studies. METHODS We retrospectively reviewed the electronic health records of consecutive patients enrolled in EP-CTs from 2017 to 2019 to obtain baseline characteristics and number of health care-associated days, defined as all inpatient and outpatient visits while on trial. We used univariable and multivariable analyses to identify predictors of increased time toxicity, defined as the proportion of health care-associated days among total days on trial. For ease of interpretation, we created a dichotomous variable, with high time toxicity defined as ≥20% health care-associated days during time on trial and used regression models to evaluate relationships between time toxicity and clinical outcomes. RESULTS Among 408 EP-CT participants (mean age, 60.5 years [standard deviation, SD, 12.6]; 56.5% female; 88.2% White; 96.0% non-Hispanic), patients had an average of 22.5% health care-associated days while on trial (SD, 13.8%). Those with GI (B = 0.07; P = .002), head/neck (B = 0.09; P = .004), and breast (B = 0.06; P = .015) cancers and those with worse performance status (B = 0.04; P = .017) and those receiving targeted therapies (B = 0.04; P = .014) experienced higher time toxicity. High time toxicity was associated with decreased disease response rates (odds ratio, 0.07; P < .001), progression-free survival (hazard ratio [HR], 2.10; P < .001), and overall survival (HR, 2.16; P < .001). CONCLUSION In this cohort of EP-CT participants, patients spent more than one-fifth of days on trial with health care contact. We identified characteristics associated with higher time toxicity and found that high toxicity correlated with worse clinical outcomes. These data could help inform patient-clinician discussions about EP-CTs, guide future trial design, and identify at-risk patients.
Collapse
Affiliation(s)
- Sienna M Durbin
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA
| | - Debra M Lundquist
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | | | - Rachel Jimenez
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Laura Petrillo
- Division of Palliative Care and Geriatric Medicine, Massachusetts General Hospital, Boston, MA
| | - Janice Kim
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA
| | - Kaitlyn Lynch
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Megan Healy
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Andrew Johnson
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Nicholas Ollila
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Vaishnavi Yalala
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Benjamin Malowitz
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Allison Kehlmann
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Nicholas Chevalier
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Victoria Turbini
- Division of Palliative Care and Geriatric Medicine, Massachusetts General Hospital, Boston, MA
| | - Viola Bame
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Hope Heldreth
- Statistician, Brigham and Women's Hospital, Boston, MA
| | - Jenipher Silva
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Casandra McIntyre
- Department of Nursing & Patient Care Services, Massachusetts General Hospital, Boston, MA
| | - Dejan Juric
- Department of Medicine, Division of Hematology & Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA
| | - Ryan D Nipp
- University of Oklahoma Stephenson Cancer Center, Oklahoma City, OK
| |
Collapse
|
10
|
Graber N, Canova N, Bryant‐Lukosius D, Robert G, Navarro‐Rodrigo B, Trueb L, Coukos G, Eicher M, Corbière T, Colomer‐Lahiguera S. Reflections on the opportunities and challenges of applying experience-based co-design (EBCD) to phase 1 clinical trials in oncology. Health Expect 2024; 27:e14068. [PMID: 38937953 PMCID: PMC11211206 DOI: 10.1111/hex.14068] [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: 12/21/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Experience-Based Co-Design (EBCD) is a multi-stage participatory action research process which was developed originally to increase patient involvement in service improvement initiatives. This viewpoint article serves as a reflection on the researchers' experiences, focusing on the application and feasibility of participatory approaches, particularly co-design, in the specific context of early-phase clinical trials. METHODS We reflect on the opportunities and challenges of applying EBCD in a new context of early-phase clinical trials in oncology where experimental treatments are increasingly perceived as a therapeutic option and, in certain instances, their efficacy may lead to accelerated approval facilitating a swifter integration into standard care. RESULTS We propose that the opportunity of applying EBCD in such trials lies in improving the delivery of person-centered care, care coordination, and support during the transition from experimental to standard care. Three potential challenges when applying EBCD in early-phase clinical trials are discussed related to: the need for standardization in trial processes; planning EBCD in a context of high uncertainty; and vulnerability of patient populations. CONCLUSION Integrating EBCD into early-phase oncology trials presents an opportunity to enhance person-centered care and can lead to simultaneous improvements in care processes and therapeutic development. PATIENT OR PUBLIC CONTRIBUTION This article has been developed with the collaboration of a patient partner who serves on the advisory board of our ongoing EBCD study in early clinical trials.
Collapse
Affiliation(s)
- Nils Graber
- Institute of Higher Education and Research in Healthcare (IUFRS), Faculty of Biology and MedicineUniversity of Lausanne (UNIL)LausanneSwitzerland
| | - Nina Canova
- Institute of Higher Education and Research in Healthcare (IUFRS), Faculty of Biology and MedicineUniversity of Lausanne (UNIL)LausanneSwitzerland
| | | | - Glenn Robert
- Florence Nightingale Faculty of Nursing, Midwifery and Palliative CareKing's College LondonLondonUK
| | | | - Lionel Trueb
- Department of OncologyLausanne University Hospital (CHUV)LausanneSwitzerland
| | - George Coukos
- Department of OncologyLausanne University Hospital (CHUV)LausanneSwitzerland
| | - Manuela Eicher
- Institute of Higher Education and Research in Healthcare (IUFRS), Faculty of Biology and MedicineUniversity of Lausanne (UNIL)LausanneSwitzerland
- Department of OncologyLausanne University Hospital (CHUV)LausanneSwitzerland
| | - Tourane Corbière
- Institute of Higher Education and Research in Healthcare (IUFRS), Faculty of Biology and MedicineUniversity of Lausanne (UNIL)LausanneSwitzerland
| | - Sara Colomer‐Lahiguera
- Institute of Higher Education and Research in Healthcare (IUFRS), Faculty of Biology and MedicineUniversity of Lausanne (UNIL)LausanneSwitzerland
- Department of OncologyLausanne University Hospital (CHUV)LausanneSwitzerland
| |
Collapse
|
11
|
Alotaibi H, Anis AM, Alloghbi A, Alshammari K. Oncology Early-Phase Clinical Trials in the Middle East and North Africa: A Review of the Current Status, Challenges, Opportunities, and Future Directions. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:178-189. [PMID: 39219998 PMCID: PMC11361343 DOI: 10.36401/jipo-23-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 09/04/2024]
Abstract
Clinical trials, the empirical discipline of medical experimentation conducted on human subjects, have engendered a paradigm shift in medical research. The need for new clinical studies is paramount in the Middle East and North Africa (MENA) region, with its rising cancer incidence and demand for efficient oncology treatments. This paper comprehensively reviews the challenges, opportunities, and future directions of phase I oncology clinical trials in the MENA region. Early-phase trials are vital in determining drug dosage and assessing toxicity, bridging the gap between preclinical research and clinical practice. Considering the unique landscape of MENA, this review explores regulatory aspects, specific hurdles faced, potential advantages, and areas for improvement in conducting these trials. Various future directions can be pursued to maximize the potential of phase I oncology trials in MENA. While regulatory bodies like the Ministry of Health adhere to the International Conference on Harmonization-Good Clinical Practice guidelines, a unified system meeting high standards would yield better results. Strengthening research infrastructure, establishing research centers, incorporating clinical trial education into the curriculum, and improving access to medical facilities are crucial. Enhancing consumer understanding of research would facilitate increased participation and promote sustainability in trial recruitment. Navigating various funding sources would open the door for more funding opportunities. Collaborations between academia, industry, and regulatory bodies, both international and local, should be fostered to promote knowledge sharing, resource pooling, and harmonization of standards. Such collaborations would contribute to the sustainability of clinical trial activities by leveraging collective expertise, sharing research infrastructure, and distributing the burden of regulatory compliance. By adopting these strategies, the MENA region can advance its capacity to conduct early phases of oncology trials and contribute significantly to the global medical research landscape.
Collapse
Affiliation(s)
- Hawazin Alotaibi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Amna M. Anis
- Biomedical Engineering Department, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
- College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
| | - Abdurahman Alloghbi
- Medical Oncology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Kanan Alshammari
- Department of Oncology, Ministry of National Guard and Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Maki A, Narukawa M. Factors Associated with Inclusion of Japan in Phase I Multiregional Clinical Trials in Oncology. Ther Innov Regul Sci 2024; 58:766-772. [PMID: 38652349 DOI: 10.1007/s43441-024-00655-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Early inclusion of Japan in the global development program could be a key factor in reducing the drug lag, making participation in phase I multiregional clinical trials (Ph. I MRCTs) an important consideration for oncology drug development in Japan. We aimed to investigate the factors associated with the inclusion of Japan in Ph. I MRCTs in oncology. METHODS We compared the trial design, target population, type of primary tested drug, trial conduct profile, and sponsor profile for Ph. I MRCTs with or without Japan conducted by the top 20 companies in more than two countries and started between January 1, 2011, and December 31, 2020. RESULTS One hundred and ninety-seven Ph. I MRCTs included Japan, and 697 did not. Detailed features of the Ph. I MRCTs in oncology were summarized, and several factors (trial design, target population, trial conduct profile, and sponsor profile) associated with inclusion of Japan in the Ph. I MRCTs were identified. CONCLUSIONS It is important for Japanese subsidiaries within global pharmaceutical companies to closely communicate with the headquarters based on medical practice and unmet needs in Japan to join global development from an early stage. In addition, further efforts to attract emerging biopharmaceutical companies to Japan from the regulatory and/or political perspectives would be needed, thereby preventing drug lag in Japan.
Collapse
Affiliation(s)
- Akio Maki
- Department of Clinical Medicine (Pharmaceutical Medicine), Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, 108-8641, Tokyo, Japan.
| | - Mamoru Narukawa
- Department of Clinical Medicine (Pharmaceutical Medicine), Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, 108-8641, Tokyo, Japan
| |
Collapse
|
13
|
Spencer K, Butenschoen H, Alger E, Bachini M, Cook N. Amplifying the Patient's Voice in Oncology Early-Phase Clinical Trials: Solutions to Burdens and Barriers. Am Soc Clin Oncol Educ Book 2024; 44:e433648. [PMID: 38857456 DOI: 10.1200/edbk_433648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Dose-finding oncology trials (DFOTs) provide early access to novel compounds of potential therapeutic benefit in addition to providing critical safety and dosing information. While access to trials for which a patient is eligible remains the largest barrier to enrollment on clinical trials, additional direct and indirect barriers unique to enrollment on DFOTs are often overlooked but worthy of consideration. Direct barriers including financial costs of care, travel and time investments, and logical challenges including correlative study designs are important to bear in mind when developing strategies to facilitate the patient experience on DFOTs. Indirect barriers such as strict eligibility criteria, washout periods, and concomitant medication restrictions should be accounted for during DFOT design to maintain the fidelity of the trial without being overly exclusionary. Involving patients and advocates and incorporating patient-reported outcomes (PROs) throughout the process, from initial DFOT design, through patient recruitment and participation, is critical to informing strategies to minimize identified barriers to offer the benefit of DFOTs to all patients.
Collapse
Affiliation(s)
- Kristen Spencer
- Department of Medicine at NYU Grossman School of Medicine, NYU Langone Perlmutter Cancer Center, New York, NY
| | - Henry Butenschoen
- Department of Medicine at NYU Grossman School of Medicine, NYU Langone Perlmutter Cancer Center, New York, NY
| | - Emily Alger
- The Alan Turing Institute, London, United Kingdom
| | | | - Natalie Cook
- University of Manchester and the Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
14
|
Schattner E. Randomized trials of cancer drugs are for yesterday. Nature 2024; 629:S13. [PMID: 38811704 DOI: 10.1038/d41586-024-01430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
|
15
|
Sawyer CS, Taylor S, Carter L, Stanworth M, Davies M, Thistlethwaite F, Taylor J, Eastwood C, Yorke J. Development and validation of a patient reported experience measure for experimental cancer medicines (PREM-ECM) and their carers (PREM-ECM-Carer). BMC Cancer 2024; 24:500. [PMID: 38641809 PMCID: PMC11031988 DOI: 10.1186/s12885-024-11963-x] [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: 01/13/2023] [Accepted: 02/05/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Our aim was to develop a validated Patient Reported Experience Measure (PREM) to capture patient and carer experience during participation in experimental cancer medicine trials (ECM): called PREM-ECM. METHODS Mixed method design, consisting of four stages. Questionnaire items were produced for both patients and carers using interviews, focus groups, and cognitive interviews with patients and carers separately. For both patient and carer PREMs, a cross-sectional questionnaire study was conducted to identify final items for inclusion using hierarchical item reduction and Rasch analysis. Questionnaire validity and reliability were assessed, including administration feasibility. RESULTS Initial interview participants suggested the need for three PREMs, two specific to patients: (i) a 'prior' questionnaire that captured experiences of trial introduction, screening, consenting, and early trial experience (< 6 weeks post consent); and (ii) 'on-trial' that captured experiences of ongoing consent and trial participation; and (iii) a PREM specific for carers. The draft 25-item 'prior' questionnaire was completed by 162 patients and 162 patients completed the draft 35-item 'on-trial' questionnaire. Hierarchical and Rasch analysis produced a 14-item 'prior' list and a 15-item list for 'on-trial'. Both patient PREM's demonstrated a good fit to the Rasch model following Bonferroni correction (X2p = 0.008). The carer 34-draft item questionnaire was completed by 102 participants. Hierarchical and Rasch analysis produced a 13-item list for PREM-ECM-Carer, with good fit to the Rasch model ( X2p = 0.62). The pilot testing demonstrated the feasibility of all the PREMs in capturing patient and caregiver experiences in routine clinical settings. CONCLUSIONS The three PREM-ECM questionnaires will be the first validated experience measures for ECM trial patients and their carers. These questionnaires may be used to assess patients' and their carers' experiences of ECM and enable robust comparisons across cancer trial units highlighting areas for service improvement.
Collapse
Affiliation(s)
- Chelsea S Sawyer
- Division of Psychology and Mental Health, The University of Manchester, Manchester, UK
- Christie Patient Centred Research (CPCR), The Christie NHS Foundation Trust, Manchester, UK
| | - Sally Taylor
- Christie Patient Centred Research (CPCR), The Christie NHS Foundation Trust, Manchester, UK
- Division of Nursing Midwifery and Social Work, School of Health Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester, UK
| | - Louise Carter
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester, UK
| | - Melissa Stanworth
- Christie Patient Centred Research (CPCR), The Christie NHS Foundation Trust, Manchester, UK
| | - Michelle Davies
- NIHR Manchester Clinical Research Facility, The Christie NHS Foundation Trust, Manchester, UK
- The Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester, UK
| | - Fiona Thistlethwaite
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester, UK
- The Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester, UK
| | - Jo Taylor
- The Christie NHS Foundation Trust, Manchester, UK
| | - Charlotte Eastwood
- Christie Patient Centred Research (CPCR), The Christie NHS Foundation Trust, Manchester, UK
| | - Janelle Yorke
- Christie Patient Centred Research (CPCR), The Christie NHS Foundation Trust, Manchester, UK.
- Division of Nursing Midwifery and Social Work, School of Health Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester, UK.
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
16
|
Cannarile MA, Karanikas V, Reis B, Mancao C, Lagkadinou E, Rüttinger D, Rieder N, Ribeiro FR, Kao H, Dziadek S, Gomes B. Facts and Hopes on Biomarkers for Successful Early Clinical Immunotherapy Trials: Innovative Patient Enrichment Strategies. Clin Cancer Res 2024; 30:1448-1456. [PMID: 38100047 DOI: 10.1158/1078-0432.ccr-23-1530] [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: 05/20/2023] [Revised: 10/17/2023] [Accepted: 12/08/2023] [Indexed: 04/16/2024]
Abstract
Despite the clinical validation and unequivocal benefit to patients, the development of cancer immunotherapies is facing some key challenges and the attrition rate in early phases of development remains high. Identifying the appropriate patient population that would benefit most from the drug is on the critical path for successful clinical development. We believe that a systematic implementation of patient enrichment strategies early in the drug development process and trial design, is the basis for an innovative, more efficient, and leaner clinical development to achieve earlier a clear proof of concept or proof of failure. In this position article, we will describe and propose key considerations for the implementation of patient enrichment strategies as an opportunity to provide decision-enabling data earlier in the drug development process. We introduce an innovative multidimensional tool for immuno-oncology drug development that focuses on facilitating the identification and prioritization of enrichment-relevant biomarkers, based on the drug mechanism of action. To illustrate its utility, we discuss patient enrichment examples and use a case in the field of cancer immunotherapy, together with technical and regulatory considerations. Overall, we propose to implement fit for purpose enrichment strategies for all investigational drugs as early as possible in the development process. We believe that this will increase the success rate of immuno-oncology clinical trials, and eventually bring new and better medicines to patients faster.
Collapse
Affiliation(s)
- Michael A Cannarile
- Roche Diagnostics GmbH, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Munich, Munich, Germany
| | - Vaios Karanikas
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Bernhard Reis
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Christoph Mancao
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Eleni Lagkadinou
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Dominik Rüttinger
- Roche Diagnostics GmbH, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Munich, Munich, Germany
| | - Natascha Rieder
- Roche Diagnostics GmbH, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Munich, Munich, Germany
| | - Franclim R Ribeiro
- Roche Diagnostics GmbH, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Munich, Munich, Germany
| | - Henry Kao
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Sebastian Dziadek
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Bruno Gomes
- F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
17
|
Avery J, Bell JAH, Baryolay K, Rodin G, Nissim R, Balneaves LG. Decision-making and autonomy among participants in early-phase cancer immunotherapy trials: a qualitative study. BMC Cancer 2024; 24:373. [PMID: 38528488 DOI: 10.1186/s12885-024-12119-7] [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: 08/24/2023] [Accepted: 03/13/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Participants considering early-phase cancer clinical trials (CTs) need to understand the unique risks and benefits prior to providing informed consent. This qualitative study explored the factors that influence patients' decisions about participating in early-phase cancer immunotherapy CTs through the ethical lens of relational autonomy. METHODS Using an interpretive descriptive design, interviews were conducted with 21 adult patients with advanced cancer who had enrolled in an early-phase CT. Data was analyzed using relational autonomy ethical theory and constant comparative analysis. RESULTS The extent to which participants perceived themselves as having a choice to participate in early-phase cancer immunotherapy CTs was a central construct. Perceptions of choice varied according to whether participants characterized their experience as an act of desperation or as an opportunity to receive a novel treatment. Intersecting psychosocial and structural factors influenced participants' decision making about participating in early-phase cancer immunotherapy trials. These relational factors included: (1) being provided with hope; (2) having trust; (3) having the ability to withdraw; and (4) timing constraints. CONCLUSIONS Findings highlight the continuum of perceived choice that exists among patients with cancer when considering participation in early-phase cancer immunotherapy CTs. All participants were interpreted as exhibiting some degree of relational autonomy within the psychosocial and structural context of early-phase CT decision making. This study offers insights into the intersection of cancer care delivery, personal beliefs and values, and established CT processes and structures that can inform future practices and policies associated with early-phase cancer immunotherapy CTs to better support patients in making informed decisions.
Collapse
Affiliation(s)
- Jonathan Avery
- School of Nursing, University of British Columbia, Vancouver, BC, Canada
- Department of Supportive Care Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jennifer A H Bell
- Department of Supportive Care Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Clinical and Organizational Ethics, University Health Network, Toronto, ON, Canada.
- The Institute for Education Research, University Health Network, Toronto, ON, Canada.
- Department of Psychiatry and Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, Department of Supportive Care, Research Division, 700 Bay St., 23rd Floor, Toronto, ON, M5G 1Z6, Canada.
| | - Khotira Baryolay
- Department of Supportive Care Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Gary Rodin
- Department of Supportive Care Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Global Institute of Psychosocial, Palliative and End-of-Life Care (GIPPEC), Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Cancer Experience, University Health Network Cancer Program, University Health Network, Toronto, ON, Canada
- Princess Margaret Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Rinat Nissim
- Department of Supportive Care Research, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lynda G Balneaves
- Rady Faculty of Health Sciences, College of Nursing, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
18
|
Beninger P. Phase I Studies: Innovations and Issues. Clin Ther 2024; 46:85-89. [PMID: 38342708 DOI: 10.1016/j.clinthera.2024.01.002] [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: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/13/2024]
Abstract
INTRODUCTION Emeritus Editor-in-Chief, Richard Shader, published 2 editorials in 2014 to state that Clinical Therapeutics' would no longer consider simple innovator vs generic bioequivalence studies for publication and would require a rationale for the choice of agents when submitting drug-drug interaction studies for consideration. The intervening decade of developments in this field provides an opportunity to comment on these trends. Lewis Scheiner anchors the subsequent discussion in a "Learn and Confirm" super-structure of thinking about the goals of early development of pharmaceutical agents. Subsequent experience with newer agents that are focused on immunological targets has led to a shift from the simple No Observable Effect Level (NOEL) model to the Minimal Anticipated Biological Effect Level (MABEL) model for biologically focused effects to assess pre-clinical data in guiding the selection of a starting dose for First-in-Human studies. ELEMENTS OF PHASE I STUDIES The primary tasks of Phase I activities are to describe the pharmacokinetics (determination of absorption, distribution, metabolism, and excretion) and essential pharmacodynamics (the dose correlation with the physiological responses, plus any untoward effects, including idiosyncratic responses) keeping in mind reporting requirements. Other Phase I activities usually conducted later in the development cycle include evaluation of drug interactions with food and other pharmaceutical agents and thorough QT studies. INNOVATIONS Phase I studies have been evolving in response to the unrelenting pressures to improve access and efficiencies in time, cost, and effort. Changes have been occurring in the characteristics of the participating populations, the starting dose, and shifts in the enrollment schedule to a more flexible, data-driven, adaptive design. ISSUES Additional issues have gained attention in the recent past, including Phase 0/microdosing, use of Phase I studies explicitly for treatment in the case of oncological products, involvement of Data Safety Monitoring Committees especially for first-in-class molecules, and improved means of optimizing selection of candidate agents for advancement to subsequent stages of development. Of final importance is the need for greater transparency of the presently inaccessible, early development study data maintained in commercial corporate legacy databases. Taken together, these developments and innovations by a broad range of stakeholders point to continuing opportunities for clinical investigators to explore the potential of Phase I studies to contribute to their own specialties.
Collapse
Affiliation(s)
- Paul Beninger
- Public Health & Community Medicine, Tufts University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
19
|
Losurdo A, Dipasquale A, Giordano L, Persico P, Lorenzi E, Di Muzio A, Barigazzi C, Korolewicz J, Mehan A, Mohammed O, Scheiner B, Pinato DJ, Santoro A, Simonelli M. Refining patient selection for next-generation immunotherapeutic early-phase clinical trials with a novel and externally validated prognostic nomogram. Front Immunol 2024; 15:1323151. [PMID: 38298193 PMCID: PMC10828843 DOI: 10.3389/fimmu.2024.1323151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Introduction Identifying which patient may benefit from immunotherapeutic early-phase clinical trials is an unmet need in drug development. Among several proposed prognostic scores, none has been validated in patients receiving immunomodulating agents (IMAs)-based combinations. Patients and methods We retrospectively collected data of 208 patients enrolled in early-phase clinical trials investigating IMAs at our Institution, correlating clinical and blood-based variables with overall survival (OS). A retrospective cohort of 50 patients treated with IMAs at Imperial College (Hammersmith Hospital, London, UK) was used for validation. Results A total of 173 subjects were selected for analyses. Most frequent cancers included non-small cell lung cancer (26%), hepatocellular carcinoma (21.5%) and glioblastoma (13%). Multivariate analysis (MVA) revealed 3 factors to be independently associated with OS: line of treatment (second and third vs subsequent, HR 0.61, 95% CI 0.40-0.93, p 0.02), serum albumin as continuous variable (HR 0.57, 95% CI 0.36-0.91, p 0.02) and number of metastatic sites (<3 vs ≥3, HR 0.68, 95% CI 0.48-0.98, p 0.04). After splitting albumin value at the median (3.84 g/dL), a score system was capable of stratifying patients in 3 groups with significantly different OS (p<0.0001). Relationship with OS reproduced in the external cohort (p=0.008). Then, from these factors we built a nomogram. Conclusions Prior treatment, serum albumin and number of metastatic sites are readily available prognostic traits in patients with advanced malignancies participating into immunotherapy early-phase trials. Combination of these factors can optimize patient selection at study enrollment, maximizing therapeutic intent.
Collapse
Affiliation(s)
- Agnese Losurdo
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Angelo Dipasquale
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Laura Giordano
- Biostatistic Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Pasquale Persico
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Elena Lorenzi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Antonio Di Muzio
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Chiara Barigazzi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - James Korolewicz
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Aman Mehan
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Oreoluwa Mohammed
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Benhard Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - David J. Pinato
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Division of Oncology, Department of Translational Medicine (DIMET), Università del Piemonte Orientale A. Avogadro, Novara, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Milan, Italy
| |
Collapse
|
20
|
Locher C, Laporte S, Derambure P, Chassany O, Girault C, Avakiantz A, Bahans C, Deplanque D, Fustier P, Germe AF, Kassaï B, Lacoste L, Petitpain N, Roustit M, Simon T, Train C, Cucherat M. Data Monitoring Committees and clinical trials: From scientific justification to organisation. Therapie 2024; 79:111-121. [PMID: 38103949 DOI: 10.1016/j.therap.2023.12.002] [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: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Clinical trials often last several months or even several years. As the trial progresses, it can be tempting to find out whether the data obtained already answers the question posed at the start of the trial in order to stop inclusions or monitoring earlier. However, knowing and taking into account interim results can sometimes compromise the integrity of the results, which is counterproductive. To minimise this risk and ensure that the treatments are assessed reliably, safety and/or efficacy criteria are monitored during the study by a Data Monitoring Committee. After receiving the results confidentially, the Data Monitoring Committee assesses the benefit/risk ratio of the study treatment and recommends that the trial be continued, modified or terminated. Data Monitoring Committee members issuing these recommendations have an important responsibility: a hasty decision to end the trial may lead to inconclusive results unable to answer the initial question and, inversely, delaying the decision to end the trial may expose the subjects to potentially ineffective or even harmful interventions. The Data Monitoring Committee's task is therefore particularly complex. With this in mind, the round table discussion at the Giens workshops was a chance to review the scientific justification for creating Data Monitoring Committees and to recall the need for their members to receive comprehensive training on the complexities of multiple analyses, confidentiality requirements applying to the results and the need for them to be aware that recommendations to end a trial must be based on data that is robust enough to assess the benefit/risk ratio of the treatment studied.
Collapse
Affiliation(s)
- Clara Locher
- Inserm, UMR S 1085, service de pharmacologie clinique, Centre d'investigation clinique de Rennes (CIC1414), Institut de recherche en santé, environnement et travail (Irset), CHU de Rennes, 35000 Rennes, France.
| | - Silvy Laporte
- UMR 1059 Inserm, URC/pharmacologie clinique, université Jean-Monnet Saint-Étienne, CHU de Saint-Étienne, 42055 Saint-Étienne, France
| | | | - Olivier Chassany
- Unité de recherche clinique en économie de la santé (URC-ECO), hôpital Hôtel-Dieu, AP-HP, 75004 Paris, France
| | - Cécile Girault
- Fédération francophone de cancérologie digestive (FFCD), 21000 Dijon, France
| | | | - Claire Bahans
- Département de pédiatrie, CHU de Limoges, 87000 Limoges, France
| | - Dominique Deplanque
- Inserm, CIC 1403, Centre d'investigation clinique, University of Lille, CHU de Lille, 59000 Lille, France
| | - Pierre Fustier
- Département de recherche et développement clinique - hématologie - BeiGene, Switzerland GmbH, 4051 Basel, Switzerland
| | | | - Behrouz Kassaï
- Inserm, UMR 5558 CNRS, service de pharmacotoxicologie, Centre d'investigation clinique 1407, hospices civils de Lyon, université de Lyon, 69000 Lyon, France
| | - Louis Lacoste
- Pôle USSAR, anesthésie réanimation, CHU de Poitiers, 86021 Poitiers, France
| | - Nadine Petitpain
- Unité de vigilance des essais cliniques, DRCI CHRU de Nancy, 54500 Vandœuvre-lès-Nancy, France
| | - Matthieu Roustit
- Inserm, CIC1406, University Grenoble Alpes, CHU de Grenoble, 38000 Grenoble, France
| | - Tabassome Simon
- Service de pharmacologie clinique, plateforme de recherche de l'est parisien (URCEST-CRCEST-CRB), hôpital Saint-Antoine, Sorbonne université, Assistance publique-Hôpitaux de Paris (AP-HP), 75000 Paris, France
| | | | - Michel Cucherat
- metaEvidence.org, service de pharmacotoxicologie, hospices civils de Lyon, 69000 Lyon, France
| |
Collapse
|
21
|
Locher C, Laporte S, Derambure P, Chassany O, Girault C, Avakiantz A, Bahans C, Deplanque D, Fustier P, Germe AF, Kassaï B, Lacoste L, Petitpain N, Roustit M, Simon T, Train C, Cucherat M. Comité de surveillance indépendant dans les essais cliniques : de la justification scientifique à l’organisation. Therapie 2024; 79:99-110. [PMID: 37985309 DOI: 10.1016/j.therap.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Affiliation(s)
- Clara Locher
- Université de Rennes, CHU de Rennes, CIC 1414 (Centre d'investigation clinique de Rennes), 35000 Rennes, France.
| | - Silvy Laporte
- Université Jean Monnet Saint-Étienne, UMR 1059 Inserm, URC/Pharmacologie Clinique, CHU de Saint-Étienne, 42055 Saint-Étienne, France
| | | | - Olivier Chassany
- Unité de recherche clinique en économie de la santé (URC-ECO), hôpital Hôtel-Dieu, AP-HP, 75004 Paris, France
| | - Cécile Girault
- Fédération francophone de cancérologie digestive (FFCD), faculté de médecine, 21000 Dijon, France
| | | | - Claire Bahans
- Département de pédiatrie, CHU de Limoges, 87000 Limoges, France
| | - Dominique Deplanque
- Centre d'investigation clinique, Université Lille, Inserm, CHU de Lille, CIC 1403, 59000 Lille, France
| | - Pierre Fustier
- Département de recherche et développement clinique - Hématologie - BeiGene, Switzerland GmbH, 4051 Basel, Suisse
| | | | - Behrouz Kassaï
- Service de pharmacotoxicologie, centre d'investigation clinique 1407 Inserm - Hospices civils de Lyon, UMR 5558 CNRS université de Lyon, 69000 Lyon, France
| | - Louis Lacoste
- Pôle USSAR, anesthésie réanimation, CHU de Poitiers, 86021 Poitiers, France
| | - Nadine Petitpain
- Unité de vigilance des essais cliniques, DRCI CHRU de Nancy, 54500 Vandoeuvre lès Nancy, France
| | - Matthieu Roustit
- Université Grenoble Alpes, Inserm, CHU de Grenoble, CIC1406, 38000 Grenoble, France
| | - Tabassome Simon
- Sorbonne Université, service de pharmacologie clinique, plateforme de recherche de l'est parisien (URCEST-CRCEST-CRB), Assistance publique-Hôpitaux de Paris (AP-HP), Hôpital St Antoine, 75000 Paris, France
| | | | - Michel Cucherat
- Service de pharmacotoxicologie Hospices civils de Lyon, metaEvidence.org, 69000 Lyon, France
| |
Collapse
|
22
|
Lalonde CS, Switchenko JM, Behera M, Bilen MA, Owonikoko TK, Kaufman JL, Nooka AK, Lewis CM, Hitron E, Collins H, Judson EC, Alese OB, Donald Harvey R, Carlisle JW. Shifting Sociodemographic Characteristics of a Phase I Clinical Trial Population at an NCI-Designated Comprehensive Cancer Center in the Southeast. Oncologist 2023; 28:1055-1063. [PMID: 37418599 PMCID: PMC10712723 DOI: 10.1093/oncolo/oyad181] [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: 02/27/2023] [Accepted: 05/30/2023] [Indexed: 07/09/2023] Open
Abstract
Racial and ethnic minority populations are consistently under-represented in oncology clinical trials despite comprising a disproportionate share of a cancer burden. Phase I oncology clinical trials pose a unique challenge and opportunity for minority inclusion. Here we compared the sociodemographic characteristics of patients participating in phase 1 clinical trials a National Cancer Institute ( NCI)-designated comprehensive center to all patients at the center, patients with new cancer diagnosis in metropolitan Atlanta and patients with new cancer diagnoses in the state of Georgia. From 2015 to 2020, 2325 patients (43.4% female, 56.6% male) consented to participate in a phase I trial. Grouped self-reported race distribution was 70.3% White, 26.2% Black, and 3.5% other. Of new patient registrations at Winship Cancer Institute (N = 107 497) (50% F, 50% M), grouped race distribution was 63.3% White, 32.0% Black, and 4.7% other. Patients with new cancer diagnoses in metro Atlanta from 2015 to 2016 (N = 31101) were 58.4% White, 37.2% Black, and 4.3% other. Race and sex distribution of phase I patients was significantly different than Winship patients (P < .001). Over time, percent of White patients decreased in both phase I and Winship groups (P = .009 and P < .001, respectively); percentage of females did not change in either group (P = .54 phase I, P = .063 Winship). Although phase I patients were more likely to be White, male, and privately ensured than the Winship cohort, from 2015 to 2020 the percentage of White patients in phase I trials and among all new patients treated at Winship decreased. The intent of characterizing existing disparities is to improve the representation of patients from racial and ethnic minority backgrounds in phase I clinical trials.
Collapse
Affiliation(s)
- Chloe S Lalonde
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeffrey M Switchenko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health of Emory University, Atlanta, GA, USA
| | - Madhusmita Behera
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehmet A Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan L Kaufman
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ajay K Nooka
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Elise Hitron
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Hannah Collins
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Emma C Judson
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Olatunji B Alese
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - R Donald Harvey
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jennifer W Carlisle
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
23
|
Shyr CR, Liu LC, Chien HS, Huang CP. Immunotherapeutic Agents for Intratumoral Immunotherapy. Vaccines (Basel) 2023; 11:1717. [PMID: 38006049 PMCID: PMC10674963 DOI: 10.3390/vaccines11111717] [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: 09/21/2023] [Revised: 10/22/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Immunotherapy using systemic immune checkpoint inhibitors (ICI) and chimeric antigen receptor (CAR) T cells has revolutionized cancer treatment, but it only benefits a subset of patients. Systemic immunotherapies cause severe autoimmune toxicities and cytokine storms. Immune-related adverse events (irAEs) plus the immunosuppressive tumor microenvironment (TME) have been linked to the inefficacy of systemic immunotherapy. Intratumoral immunotherapy that increases immunotherapeutic agent bioavailability inside tumors could enhance the efficacy of immunotherapies and reduce systemic toxicities. In preclinical and clinical studies, intratumoral administration of immunostimulatory agents from small molecules to xenogeneic cells has demonstrated antitumor effects not only on the injected tumors but also against noninjected lesions. Herein, we review and discuss the results of these approaches in preclinical models and clinical trials to build the landscape of intratumoral immunotherapeutic agents and we describe how they stimulate the body's immune system to trigger antitumor immunity as well as the challenges in clinical practice. Systemic and intratumoral combination immunotherapy would make the best use of the body's immune system to treat cancers. Combining precision medicine and immunotherapy in cancer treatment would treat both the mutated targets in tumors and the weakened body's immune system simultaneously, exerting maximum effects of the medical intervention.
Collapse
Affiliation(s)
- Chih-Rong Shyr
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404328, Taiwan; (C.-R.S.); (H.-S.C.)
- eXCELL Biotherapeutics Inc., Taichung 404328, Taiwan
| | - Lang-Chi Liu
- Department of Medicine, Department of Surgery, College of Medicine, China Medical University and Hospital, Taichung 404328, Taiwan;
| | - Hui-Shan Chien
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404328, Taiwan; (C.-R.S.); (H.-S.C.)
| | - Chi-Ping Huang
- Department of Medicine, Urology Division, China Medical University and Hospital, Taichung 404328, Taiwan
| |
Collapse
|
24
|
Turner JH. Cancer Care by Committee to be Superseded by Personal Physician-Patient Partnership Informed by Artificial Intelligence. Cancer Biother Radiopharm 2023; 38:497-505. [PMID: 37366774 DOI: 10.1089/cbr.2023.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Multidisciplinary tumor boards (MTBs) have become the reference standard of cancer management, founded upon randomized controlled trial (RCT) evidence-based guidelines. The inordinate delays inherent in awaiting formal regulatory agency approvals of novel therapeutic agents, and the rigidities and nongeneralizability of this regimented approach, often deny cancer patients timely access to effective innovative treatment. Reluctance of MTBs to accept theranostic care of patients with advanced neuroendocrine tumors (NETs) and metastatic castrate-resistant prostate cancer resulted in decades of delay in the incorporation of 177Lu-octreotate and 177Lu-prostate-specific membrane antigen (PSMA) into routine clinical oncology practice. Recent developments in immunotherapy and molecular targeted precision therapy, based on N-of-One individual multifactorial genome analyses, have greatly increased the complexity of decision-making. Burgeoning specialist workload and tight time frames now threaten to overwhelm the logistically, and emotionally, demanding MTB system. It is hypothesized that the advent of advanced artificial intelligence technology and Chatbot natural language algorithms will shift the cancer care paradigm from a MTB management model toward a personal physician-patient shared-care partnership for real-world practice of precision individualized holistic oncology.
Collapse
Affiliation(s)
- J Harvey Turner
- Department of Nuclear Medicine, Fiona Stanley Fremantle Hospitals Group, The University of Western Australia, Murdoch, Australia
| |
Collapse
|
25
|
Gnanasakthy A, Norcross L, Fitzgerald K. Assessment of Patient-Reported Outcomes in Industry-Sponsored Phase I Oncology Studies: Considerations for Translating Theory Into Practice. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1440-1443. [PMID: 37353056 DOI: 10.1016/j.jval.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/04/2023] [Accepted: 06/09/2023] [Indexed: 06/25/2023]
Abstract
An increasing interest in the identification of optimal dosage for oncology therapies has prompted key opinion leaders and regulators to encourage the integration of patient-reported outcome (PRO) assessments in phase I oncology clinical trials. Although the potential benefits of assessing PROs in early-phase studies have been acknowledged, the difficulties that arise from such a radical shift have been largely overlooked in the public discussion. In this commentary, the authors provide insight into the challenges that industry sponsors face in integrating PRO assessments into phase I oncology trials, with the ultimate goal of facilitating conversations that may help to resolve some of these issues.
Collapse
|
26
|
Uehara Y, Koyama T, Katsuya Y, Sato J, Sudo K, Kondo S, Yoshida T, Shoji H, Shimoi T, Yonemori K, Yamamoto N. Travel Time and Distance and Participation in Precision Oncology Trials at the National Cancer Center Hospital. JAMA Netw Open 2023; 6:e2333188. [PMID: 37713200 PMCID: PMC10504617 DOI: 10.1001/jamanetworkopen.2023.33188] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/01/2023] [Indexed: 09/16/2023] Open
Abstract
Importance Genotype-matched trials, which are becoming increasingly important in the precision oncology era, require referrals from institutions providing comprehensive genomic profiling (CGP) testing to those conducting these trials, and the travel burden for trial participation is significant. However, it remains unknown whether travel time or distance are associated with genotype-matched trial participation. Objective To assess whether travel time or distance are associated with disparities in genotype-matched trial participation following CGP testing. Design, Setting, and Participants This retrospective cohort study from June 2020 to June 2022 included patients with advanced or metastatic solid tumors referred to the National Cancer Center Hospital for participation in genotype-matched trials following CGP testing and discussion by molecular tumor boards. Data were analyzed from June to October 2022. Exposures Travel time and distance. Main Outcomes and Measures The primary and secondary outcomes were enrollment in genotype-matched trials and all-cancer clinical trials, respectively. Results Of 1127 patients (mean [range] age, 62 [16-85] years; 584 women [52%]; all residents of Japan), 127 (11%) and 241 (21%) were enrolled in genotype-matched trials and all-cancer clinical trials, respectively. The overall median (IQR) travel distance and time were 38 (21-107) km and 55 (35-110) minutes, respectively. On multivariable regression with 23 covariates, travel distance (≥100 km vs <100 km) was not associated with the likelihood of genotype-matched trial participation (26 of 310 patients [8%] vs 101 of 807 patients [12%]; odds ratio [OR], 0.64; 95% CI, 0.40-1.02), whereas in patients with travel time of 120 minutes or more, the likelihood of genotype-matched trial participation was significantly lower than those with travel time less than 120 minutes (19 of 276 patients [7%] vs 108 of 851 patients [13%]; OR, 0.51; 95% CI, 0.29-0.84). The likelihood of genotype-matched trial participation decreased as travel time increased from less than 40 (38 of 283 patients [13%]) to 40 to 120 (70 of 568 patients [12%]) and 120 or more (19 of 276 patients [7%]) minutes (OR, 0.74; 95% CI, 0.48-1.17; OR, 0.41; 95% CI, 0.22-0.74, respectively). Neither travel time nor distance were associated with the likelihood of all-cancer clinical trial participation. Conclusions and Relevance In this cohort study of patients undergoing CGP testing, an increased travel time was associated with a decreased likelihood of genotype-matched trial participation. This warrants further research on interventions, such as decentralization of clinical trials to mitigate travel burden.
Collapse
Affiliation(s)
- Yuji Uehara
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Katsuya
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Jun Sato
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuki Sudo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hirokazu Shoji
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
27
|
Maki A, Narukawa M. Exploratory Analysis of Drug Lag in New Oncology Drugs Between Japan and the US. Ther Innov Regul Sci 2023; 57:671-677. [PMID: 36966205 DOI: 10.1007/s43441-023-00512-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/07/2023] [Indexed: 03/27/2023]
Abstract
BACKGROUND Drug lag in Japan has greatly decreased over the past decades; however, new instances of drug lag have appeared along with changes in the circumstances of oncology drug development. We aimed to investigate the factors associated with the approval lag for new oncology drugs between Japan and the United States (US) over the past decade by comparing approval dates and modalities, lead indications, approval types, and phase I strategies for earlier approval in Japan. METHOD We descriptively evaluated the characteristics of 117 new oncology drugs approved in either Japan or the US from January 1, 2011, to December 31, 2020. RESULTS Seventy-one drugs were approved in Japan, 112 in the US, five only in Japan, and 46 only in the US. Interestingly, new oncology drugs were predominantly developed by the top 20 pharmaceutical companies in Japan; however, the opposite was true for drugs that were not yet approved in Japan. However, no clear trend was observed in the relationship between drug lag and the studied factors, except for the phase I strategy. There was a numerical but clear trend in which a higher percentage of phase I multiregional clinical trials (MRCTs) in the drug development strategy was observed for drugs with earlier approval in Japan. CONCLUSION Participation in global drug development during the early stages, such as during phase I MRCTs, is one of the keys to successfully minimizing this new instance of drug lag in Japan.
Collapse
Affiliation(s)
- Akio Maki
- Department of Clinical Medicine (Pharmaceutical Medicine), Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-Ku, Tokyo, 108-8641, Japan.
| | - Mamoru Narukawa
- Department of Clinical Medicine (Pharmaceutical Medicine), Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-Ku, Tokyo, 108-8641, Japan
| |
Collapse
|
28
|
Frankel PH, Groshen S, Beumer JH, Cleveland L, Kim ES, Karp JE. Ethics and Clinical Research: Improving Transparency and Informed Consent in Phase I Oncology Trials. J Clin Oncol 2023; 41:2155-2158. [PMID: 36724409 PMCID: PMC10448934 DOI: 10.1200/jco.22.01736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/15/2022] [Accepted: 11/18/2022] [Indexed: 02/03/2023] Open
Affiliation(s)
- Paul H. Frankel
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA
| | - Susan Groshen
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA
| | - Jan H. Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | - Judith E. Karp
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
29
|
Adashek JJ, Sapkota S, de Castro Luna R, Seiwert TY. Complete response to alectinib in ALK-fusion metastatic salivary ductal carcinoma. NPJ Precis Oncol 2023; 7:36. [PMID: 37041305 PMCID: PMC10090142 DOI: 10.1038/s41698-023-00378-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/30/2023] [Indexed: 04/13/2023] Open
Abstract
The advent of next-generation sequencing (NGS) has allowed for the identification of novel therapeutic targets for patients with uncommon cancers. It is well known that fusion translocations are potent driver of cancer pathogenesis and can render tumors exquisitely sensitive to matching targeted therapies. Here we describe a patient with ALK-fusion positive widely metastatic salivary ductal carcinoma, who achieved a durable complete response from alectinib, a potent and specific ALK tyrosine kinase inhibitor. This case serves as another reminder that ALK-fusions can be targeted regardless of histology and can afford patients dramatic and durable benefit. It also emphasizes the need for insurance coverage for such beneficial therapies. While ALK fusions are exceedingly rare in salivary ductal carcinoma, the presence of multiple other targetable aberrations supports the recommendation for universal NGS testing for such tumors.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Surendra Sapkota
- Department of Internal Medicine, Saint Agnes Hospital, Baltimore, MD, USA
| | - Rodrigo de Castro Luna
- Department of Radiology and Radiological Science, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Tanguy Y Seiwert
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
30
|
Lundquist DM, Jimenez R, Durbin S, Horick N, Healy M, Johnson A, Bame V, Capasso V, McIntyre C, Cashavelly B, Juric D, Nipp RD. Identifying Early-Phase Clinical Trial Participants at Risk for Experiencing Worse Clinical Outcomes. JCO Oncol Pract 2023:OP2200742. [PMID: 36791343 DOI: 10.1200/op.22.00742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
PURPOSE To identify early-phase clinical trial (EP-CT) participants at risk for experiencing worse clinical outcomes and describe receipt of supportive care services. METHODS A retrospective review of the electronic health records of consecutive patients enrolled in EP-CTs from 2017 to 2019 examined baseline characteristics, clinical outcomes, and receipt of supportive care services. The validated Royal Marsden Hospital (RMH) prognosis score was calculated using data at the time of EP-CT enrollment (scores range from 0 to 3; scores ≥ 2 indicate poor prognosis). Differences in patient characteristics, clinical outcomes, and receipt of supportive care services were compared on the basis of RMH scores. RESULTS Among 350 patients (median age = 63.2 years [range, 23.0-84.3 years], 57.1% female, 98.0% metastatic cancer), 31.7% had an RMH score indicating a poor prognosis. Those with poor prognosis RMH scores had worse overall survival (hazard ratio [HR], 2.00; P < .001), shorter time on trial (HR, 1.53; P < .001), and lower likelihood of completing the dose-limiting toxicity period (odds ratio, 0.42; P = .006) versus those with good prognosis scores. Patients with poor prognosis scores had greater risk of emergency room visits (HR, 1.66; P = .037) and hospitalizations (HR, 1.69; P = .016) while on trial, and earlier hospice enrollment (HR, 2.22; P = .006). Patients with poor prognosis scores were significantly more likely to receive palliative care consultation (46.8% v 27.6%; P < .001), but not other supportive care services. CONCLUSION This study found that RMH prognosis score could identify patients at risk for decreased survival, shorter time on trial, and greater use of health care services. The findings underscore the need to develop supportive care interventions targeting EP-CT participants' distinct needs.
Collapse
Affiliation(s)
- Debra M Lundquist
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Rachel Jimenez
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Sienna Durbin
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Nora Horick
- Biostats Center, Massachusetts General Hospital, Boston, MA
| | - Megan Healy
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Andrew Johnson
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Viola Bame
- Cancer Center Protocol Office, Massachusetts General Hospital, Boston, MA
| | - Virginia Capasso
- Department of Nursing & Patient Care Services, Massachusetts General Hospital, Boston, MA
| | - Casandra McIntyre
- Department of Nursing & Patient Care Services, Massachusetts General Hospital, Boston, MA
| | - Barbara Cashavelly
- Department of Nursing & Patient Care Services, Massachusetts General Hospital, Boston, MA
| | - Dejan Juric
- Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ryan D Nipp
- University of Oklahoma Stephenson Cancer Center, Oklahoma City, OK
| |
Collapse
|
31
|
Yang GM, Ong WY, Tan J, Ding J, Ho S, Tan D, Neo P. Motivations and experiences of patients with advanced cancer participating in Phase 1 clinical trials: A qualitative study. Palliat Med 2023; 37:257-264. [PMID: 36476098 DOI: 10.1177/02692163221137105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Persons with advanced cancer may participate in Phase 1 clinical trials - first-in-human trials that are conducted with the main objectives of safety and dosing. The motivations for participation are not well understood and may include hope for cure. AIM To explore the perspectives of persons with advanced cancer in order to understand the motivations for participating in Phase 1 clinical trials, experiences while being on trial and views on palliative care provision. DESIGN Qualitative study with a constructivist stance, using thematic analysis based upon the grounded theory approach. SETTING/PARTICIPANTS 20 persons with advanced cancer who were participating in a Phase 1 clinical trial. RESULTS Many participants described how Phase 1 clinical trial participation was their last hope, as they were cognisant of their advanced disease. Information-seeking needs differed - some needed comprehensive information while others relied on the doctor's recommendation. Participants experienced varied negative and positive physical and psycho-emotional concerns, and needed to draw on multiple sources of support such as family, friends and healthcare professionals. Some could list potential benefits of palliative care but felt they did not require it yet. The overarching theme was hope and positive thinking as a way of coping. CONCLUSIONS The concepts of hope as a way of coping and the supportive presence of healthcare professionals could be weaved into a future model of palliative care to improve the illness journey for patients considering Phase 1 clinical trial participation and other persons with advanced cancer.
Collapse
Affiliation(s)
- Grace Meijuan Yang
- Division of Supportive and Palliative care, National Cancer Centre Singapore, Singapore.,Lien Centre for Palliative Care, Duke-NUS Medical School, Singapore
| | - Wah Ying Ong
- Division of Supportive and Palliative care, National Cancer Centre Singapore, Singapore
| | - Jasmine Tan
- Division of Supportive and Palliative care, National Cancer Centre Singapore, Singapore
| | | | - Shirlynn Ho
- Division of Supportive and Palliative care, National Cancer Centre Singapore, Singapore
| | - Daniel Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Patricia Neo
- Division of Supportive and Palliative care, National Cancer Centre Singapore, Singapore
| |
Collapse
|
32
|
Wendler D, Schupmann W, Li X. Views of IRB members regarding phase 1 pediatric oncology trials. Pediatr Hematol Oncol 2023; 40:14-25. [PMID: 35502918 PMCID: PMC9630167 DOI: 10.1080/08880018.2022.2069894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/24/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023]
Abstract
There is significant debate over whether phase 1 pediatric oncology trials are ethical and approvable. We thus surveyed IRB members to answer four questions. First, do IRB members think the potential medical benefits of average phase 1 pediatric oncology trials justify the risks? Second, do they think these trials are ethically appropriate? Third, do they think these trials are approvable? Fourth, how do the views of IRB members on the first two questions compare to the views of the US public? Of the 80 respondents who answered the test questions correctly, 18.8% stated that the potential medical benefits of average phase 1 pediatric oncology trials outweigh the risks, 32.5% stated that the potential medical benefits and risks are about equal, and 48.8% stated that the risks outweigh the potential medical benefits. Compared to the general public, IRB members were significantly more likely to think the risks outweigh the potential medical benefits (p = 0.01). Finally, 68.8% of IRB members indicated that average phase 1 pediatric oncology trials are approvable, and 56.3% indicated that these trials are appropriate in children. These findings suggest two-thirds of IRB members believe average phase 1 pediatric oncology trials are approvable. Yet, almost half regard the risks as outweighing the potential medical benefits and almost half think these trials are inappropriate. These findings raise important questions regarding why IRB members and the general public evaluate the same risk/benefit profile differently, and whether it is possible to reconcile the two perspectives.
Collapse
Affiliation(s)
- David Wendler
- Department of Bioethics, NIH Clinical Center, Bethesda, Maryland, USA
| | - Will Schupmann
- Department of Bioethics, NIH Clinical Center, Bethesda, Maryland, USA
| | - Xiaobai Li
- Biostatistics and Clinical Epidemiology Service, NIH Clinical Center, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Lim J, Akbar Ali S, Prawira A, Sim HW. Impact of travel distance on outcomes for clinical trial patients: the Kinghorn Cancer Centre experience. Intern Med J 2023; 53:242-249. [PMID: 34613656 DOI: 10.1111/imj.15561] [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: 07/01/2021] [Revised: 09/18/2021] [Accepted: 10/01/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Geographic isolation and travel distance to specialist care is a known social determinant of health and contributes to poorer oncology survival outcomes. AIMS To compare survival and toxicity outcomes for patients travelling long distances (>50 km) for treatment on clinical trials with local patients (<10 km and 10-50 km). METHODS We performed a retrospective cohort study based at the Kinghorn Cancer Centre, a comprehensive cancer care centre in metropolitan Sydney. We included adult patients with advanced solid-organ malignancies who were enrolled on therapeutic clinical trials between July 2015 and December 2017. Outcome measures included overall survival, progression-free survival, rates of grade 3-4 toxicity and unplanned hospital admissions for the duration of the clinical trial. RESULTS We included 173 patients, of whom 27% lived within 10 km, 29% lived between 10 and 50 km and 44% lived further than 50 km. We did not identify significant differences between survival or toxicity outcomes between patients travelling long distances and local patients. CONCLUSIONS All patients should be considered for clinical trial referral based on clinical parameters and preference, regardless of geographic proximity. In the meantime, improving access to clinical trials for rural and regional patients continues to be a priority.
Collapse
Affiliation(s)
- Jennifer Lim
- Department of Medical Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Syafiq Akbar Ali
- Department of Medical Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Amy Prawira
- Department of Medical Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Hao-Wen Sim
- Department of Medical Oncology, The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia.,NHMRC Clinical Trials Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
34
|
Agrawal S, Arora S, Amiri-Kordestani L, de Claro RA, Fashoyin-Aje L, Gormley N, Kim T, Lemery S, Mehta GU, Scott EC, Singh H, Tang S, Theoret MR, Pazdur R, Kluetz PG, Beaver JA. Use of Single-Arm Trials for US Food and Drug Administration Drug Approval in Oncology, 2002-2021. JAMA Oncol 2023; 9:266-272. [PMID: 36580315 DOI: 10.1001/jamaoncol.2022.5985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Importance Single-arm trials have allowed for transformative therapies to be made available to patients expeditiously. However, using single-arm trials to support drug approval presents several challenges that must be carefully considered. Observations Between January 1, 2002, and December 31, 2021, the US Food and Drug Administration granted 176 new malignant hematology and oncology indications based on single-arm trials, including 116 accelerated approvals (AAs) and 60 traditional approvals. Overall, 87 approvals (49%) were for new molecular entities or original biologics and 89 (51%) were supplemental indications. Response rate (RR) was the most common end point used to support approval in these single-arm trials (173 of 176 [98%]). Of the 116 AAs based on single-arm trials, 45 (38%) fulfilled their postmarketing requirement to verify clinical benefit, 61 (52%) are pending verification of benefit, and 10 (9%) were withdrawn from the market as of December 31, 2021. Most (56 of 61 [92%]) AAs based on single-arm trials pending verification of benefit occurred during the previous 5 years and have ongoing confirmatory trials as of December 2021. Conclusions and Relevance Single-arm trials have been a common development strategy to support regulatory approval as early-stage expansion cohorts with promising durable RRs have become more prevalent. In the appropriate context, single-arm trials using durable RRs can allow patients expedited access to novel therapies and will continue to serve a role in advancing drug development in oncology. However, single-arm trials have a smaller noncomparative safety data set, inability to use time-to-event end points, and other limitations that require careful consideration within the context of the disease and available therapies. The randomized clinical trial remains the preferred approach in clinical investigation.
Collapse
Affiliation(s)
- Sundeep Agrawal
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Shaily Arora
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - R Angelo de Claro
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Lola Fashoyin-Aje
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Nicole Gormley
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Tamy Kim
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| | - Steven Lemery
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Gautam U Mehta
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Emma C Scott
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Harpreet Singh
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Shenghui Tang
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Marc R Theoret
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland.,Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| | - Richard Pazdur
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland.,Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| | - Paul G Kluetz
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland.,Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| | - Julia A Beaver
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland.,Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
35
|
Debien V, Vignot S, Massard C, Malouf G, Hollebecque A, Scoazec JY, Michiels S, Verlingue L. Molecular analysis for refractory rare cancers: Sequencing battle continues - learnings for the MOSCATO-01 study. Crit Rev Oncol Hematol 2023; 181:103888. [PMID: 36460264 DOI: 10.1016/j.critrevonc.2022.103888] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND For patients with metastatic rare cancers, treatments are limited. How systematic tumor sequencing can improve therapeutic possibilities in this population? PATIENTS AND METHODS Patients with rare cancer were identified in the MOSCATO-01 trial. Patients' outcome was measured by progression-free survival (PFS) and overall survival (OS). RESULTS The most frequently identified histologic subypes were ovarian adenocarcinoma (N = 13), carcinoma of unknown primary (N = 11), and leiomyosarcoma (N = 10). Ninety-nine (39%) of them had at least one targetable cancer molecular alteration Forty-nine patients (50%) received the therapy proposed by the molecular tumor board, and 13 patients (26%, 95%CI 15-41%) achieved a PFS2/PFS1 > 1.3. The median PFS2 on matched treatment subgroup was 2.3 months (95% CI 1.8-3.6) and the median OS was 11.4 months (95% CI 9-15.5). CONCLUSIONS The molecular screening of patients with refractory, metastatic rare cancers might increase the therapeutic options. Facilitating access strategy to molecular-driven clinical trials or agnostic-approved treatment is crucial.
Collapse
Affiliation(s)
- Véronique Debien
- Department of oncology, Institut de Cancérologie Strasbourg Europe, ICANS, Strasbourg, France; Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; Institut Jules Bordet, Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | | | - Christophe Massard
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; Department of Oncology, Institut Eugène Marquis, Rennes, France
| | - Gabriel Malouf
- Department of oncology, Institut de Cancérologie Strasbourg Europe, ICANS, Strasbourg, France
| | - Antoine Hollebecque
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Jean-Yves Scoazec
- Pathology Department, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; AMMICa, CNRS UAR3655 INSERM US23, Université Paris Saclay, Villejuif, France
| | - Stefan Michiels
- Service de Biostatistique et d'Epidémiologie, Oncostat, CESP, Inserm U1018, Université Paris-Saclay, Equipe labellisée Ligue Contre le Cancer, Institut Gustave Roussy, Villejuif, France
| | - Loïc Verlingue
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France; Unité de Phase 1, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
36
|
Molecular oncology: what is needed to speed access to innovative therapies in clinical research? Curr Opin Oncol 2022; 34:575-578. [PMID: 35855513 DOI: 10.1097/cco.0000000000000880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW A better understanding of the biology of cancer cells has led in the past 20 years to more and more molecular and immunological driven treatment strategies impacting both clinical trials and day-to-day practice. The aim of this review is to describe new approaches to conduct clinical trials in this area to speed up drug development and increase access to innovation for cancer patients. RECENT FINDINGS The design of an early phase trial has an impact on its clinical benefit. Trials deriving from a specific biomarker or histologic characteristic (also known as enrichment design) are more likely to demonstrate benefit than trials based on a more conventional design. However, the increase of low incidence cancer molecular subtypes poses a major hurdle in the clinical management and drug development research for cancer patients. SUMMARY With the identification of news targets and the subsequent introduction of precision medicine, new strategies and tools are needed to provide access to biomarker identification and target-oriented clinical trials to all cancer patients. We propose to set up a new patient-centered model to conduct clinical trials allowing simply to 'bring the trial to the patient'.
Collapse
|
37
|
Chihara D, Lin R, Flowers CR, Finnigan SR, Cordes LM, Fukuda Y, Huang EP, Rubinstein LV, Nastoupil LJ, Ivy SP, Doroshow JH, Takebe N. Early drug development in solid tumours: analysis of National Cancer Institute-sponsored phase 1 trials. Lancet 2022; 400:512-521. [PMID: 35964611 PMCID: PMC9477645 DOI: 10.1016/s0140-6736(22)01390-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/23/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND The low expectation of clinical benefit from phase 1 cancer therapeutics trials might negatively affect patient and physician participation, study reimbursement, and slow the progress of oncology research. Advances in cancer drug development, meanwhile, might have favourably improved treatment responses; however, little comprehensive data exist describing the response and toxicity associated with phase 1 trials across solid tumours. The aim of the study is to evaluate the trend of toxicity and response in phase 1 trials for solid tumours over time. METHODS We analysed patient-level data from the Cancer Therapy Evaluation Program of the National Cancer Institute-sponsored investigator-initiated phase 1 trials for solid tumours, from Jan 1, 2000, to May 31, 2019. We assessed risks of treatment-related death (grade 5 toxicity ratings possibly, probably, or definitely attributable to treatment), all on-treatment deaths (deaths during protocol treatment regardless of attribution), grade 3-4 toxicity, and proportion of overall response (complete response and partial response) and complete response rate in the study periods of 2000-05, 2006-12, and 2013-2019, and evaluated their trends over time. We also analysed cancer type-specific and investigational agent-specific response, and analysed the trend of response in each cancer type over time. Univariate associations of overall response rates with patients' baseline characteristics (age, sex, performance status, BMI, albumin concentration, and haemoglobin concentration), enrolment period, investigational agents, and trial design were assessed using risk ratio based on the modified Poisson regression model. FINDINGS We analysed 465 protocols that enrolled 13 847 patients using 261 agents. 144 (31%) trials used a monotherapy and 321 (69%) used combination therapies. The overall treatment-related death rate was 0·7% (95% CI 0·5-0·8) across all periods. Risks of treatment-related deaths did not change over time (p=0·52). All on-treatment death risk during the study period was 8·0% (95% CI 7·6-8·5). The most common grade 3-4 adverse events were haematological; grade 3-4 neutropenia occurred in 2336 (16·9%) of 13 847 patients, lymphopenia in 1230 (8·9%), anaemia in 894 (6·5%), and thrombocytopenia in 979 (7·1%). The overall response rate for all trials during the study period was 12·2% (95% CI 11·5-12·8; 1133 of 9325 patients) and complete response rate was 2·7% (2·4-3·0; 249 of 9325). Overall response increased from 9·6% (95% CI 8·7-10·6) in 2000-05 to 18·0% (15·7-20·5) in 2013-19, and complete response rates from 2·5% (2·0-3·0) to 4·3% (3·2-5·7). Overall response rates for combination therapy were substantially higher than for monotherapy (15·8% [15·0-16·8] vs 3·5% [2·8-4·2]). The overall response by class of agents differed across diseases. Anti-angiogenesis agents were associated with higher overall response rate for bladder, colon, kidney and ovarian cancer. DNA repair inhibitors were associated with higher overall response rate in ovarian and pancreatic cancer. The rates of overall response over time differed markedly by disease; there were notable improvements in bladder, breast, and kidney cancer and melanoma, but no change in the low response of pancreatic and colon cancer. INTERPRETATION During the past 20 years, the response rate in phase 1 trials nearly doubled without an increase in the treatment-related death rate. However, there is significant heterogeneity in overall response by various factors such as cancer type, investigational agent, and trial design. Therefore, informed decision making is crucial for patients before participating in phase 1 trials. This study provides updated encouraging outcomes of modern phase 1 trials in solid tumours. FUNDING National Cancer Institute.
Collapse
Affiliation(s)
- Dai Chihara
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher R Flowers
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shanda R Finnigan
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa M Cordes
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yoko Fukuda
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erich P Huang
- Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Larry V Rubinstein
- Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Loretta J Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S Percy Ivy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
38
|
Tini G, Trapani D, Duso BA, Beria P, Curigliano G, Pelicci PG, Mazzarella L. Quantifying geographical accessibility to cancer clinical trials in different income landscapes. ESMO Open 2022; 7:100515. [PMID: 35738201 PMCID: PMC9271515 DOI: 10.1016/j.esmoop.2022.100515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background Clinical trials are increasingly perceived as a therapeutic opportunity for cancer patients. Favoring their concentration in few high-expertise academic centers maximizes quality of data collection but poses an issue of access equality. Analytical tools to quantify trial accessibility are needed to rationalize resources. Materials and methods We constructed a distance-based accessibility index (dAI) using publicly available data on demographics, cancer incidence and trials. Multiple strategies were applied to mitigate or quantify clear sources of bias: reporting biases by text mining multiple registries; reliability of simple geographical distance by comparison with high-quality travel cost data for Italy; index inflation due to highly heterogeneous cancer incidence by log-transformation. We studied inequalities by Gini index and time trend significance by Mann–Kendall test. We simulated different resource allocation models in representative countries and identified locations where new studies would maximally improve the national index. Results The dAI approximated well a more realistic but not widely applicable travel cost-based index. Accessibility was unevenly distributed across and within countries (Gini index ∼0.75), with maximal inequalities in high- and upper-middle-income countries (China, United States, Russian Federation). Over time, accessibility increased but less than the total number of trials, most evidently in upper-middle-income countries. Simulations in representative countries (Italy and Serbia) identified ideal locations able to maximally raise the national index. Conclusions Access to clinical trials is highly uneven across and within countries and is not mitigated by simple increase in the number of trials; a rational algorithmic approach can be used to mitigate inequalities. Accessibility to cancer clinical trials grew less than total number of trials over time in upper-middle-income countries. Accessibility is unevenly distributed, with maximal inequalities in high- and upper-middle-income countries. Simulation of resource allocation can identify ideal locations able to raise the national accessibility index.
Collapse
Affiliation(s)
- G Tini
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milano, Italy
| | - D Trapani
- Division of Early Drug Development, IEO European Institute of Oncology, IRCCS, Milano, Italy
| | - B A Duso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milano, Italy
| | - P Beria
- Department of Architecture and Urban Studies (DAStU), Politecnico of Milano, Milano, Italy
| | - G Curigliano
- Division of Early Drug Development, IEO European Institute of Oncology, IRCCS, Milano, Italy; Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy
| | - P G Pelicci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milano, Italy; Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy
| | - L Mazzarella
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milano, Italy; Division of Early Drug Development, IEO European Institute of Oncology, IRCCS, Milano, Italy.
| |
Collapse
|
39
|
Escritt K, Mann M, Nelson A, Harrop E. Hope and meaning-making in phase 1 oncology trials: a systematic review and thematic synthesis of qualitative evidence on patient-participant experiences. Trials 2022; 23:409. [PMID: 35578308 PMCID: PMC9112562 DOI: 10.1186/s13063-022-06306-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background Phase 1 drug trials are popular treatment options for patients with advanced disease, despite the greater levels of uncertainty associated with them. However, their meaning and consequences for patient-participants remains under-explored. This review synthesises the qualitative evidence of patients’ experiences of participating in phase 1 oncology trials, exploring their decisions to take part and the impacts of these trials on patient wellbeing. Methods A comprehensive literature search involving medical subject headings (MeSH) and keywords was undertaken in the following databases: MEDLINE, EMBASE, PsycINFO, Scopus, CINAHL, and Cochrane CENTRAL, with supplementary searches also conducted. Studies were independently screened for inclusion by two researchers. Included studies were critically appraised and data extracted using standardised forms. Qualitative results were analysed using thematic synthesis. Results Three main themes were identified across 13 studies: decision-making and joining the trial; experiences of taking part in the trial and hope and coping. Patients primarily joined trials hoping for therapeutic benefits, sentiments which prevailed and shaped their experiences across their trial journey. Rather than indicate therapeutic misconception based on poor understanding, patient perspectives more commonly pointed to differences between hope and expectation and cultural narratives of staying positive, trying everything and trusting in experts. Conclusions These findings challenge information-based models of consent, favouring coping frameworks which account for the role of hope and meaning-making during serious illness. Personalised consideration of existential and quality-of-life matters before and during trials is recommended, including palliative and supportive care alternatives to active treatment. Review Registration The review was registered with PROSPERO international prospective register of systematic reviews (CRD 42020163250). Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06306-9.
Collapse
|
40
|
Nassif EF, Blay JY, Massard C, Dufresne A, Brahmi M, Cassier P, Ray-Coquard I, Pautier P, Leary A, Sunyach MP, Bahleda R, Levy A, Le Pechoux C, Honoré C, Mir O, Le Cesne A. Early phase trials in soft-tissue sarcomas: clinical benefit of inclusion in early lines of treatment, molecular screening, and histology-driven trials. ESMO Open 2022; 7:100425. [PMID: 35255445 PMCID: PMC9058915 DOI: 10.1016/j.esmoop.2022.100425] [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: 01/07/2022] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The prognosis of patients with advanced soft-tissue sarcomas (STS) remains dismal, and systemic therapeutic options are limited. Early phase trials are becoming increasingly safe and effective. This study aimed to identify the prognostic factors for progression-free survival (PFS). PATIENTS AND METHODS This retrospective analysis included all STS patients participating in early phase trials at Gustave Roussy and Léon Bérard between 1 January 2012 and 31 December 2020. RESULTS Overall, 199 patients accounted for 214 inclusions in advanced STS. The most frequent histotypes were well-differentiated/dedifferentiated liposarcomas (n = 55), leiomyosarcomas (n = 53), synovial sarcomas (n = 22), undifferentiated pleomorphic sarcomas (n = 15), angiosarcomas (n = 12), and myxoid liposarcomas (n = 10). The median PFS was 2.8 months (95% confidence interval 2.7-4.1 months). The median PFS in the first, second, and later lines was 8.3, 5.4, and 2.6 months, respectively (P = 0.00015). The median PFS was 2.8 months in case of molecular screening, 4.1 months in case of histology-driven screening, and 1.6 months (P = 0.00014) in the absence of either screening modalities. In univariate analysis, histotype (P = 0.026), complex genomics (P = 0.008), number of prior lines (P < 0.001), prior anthracyclines (P < 0.001), number of metastatic sites (P = 0.003), liver metastasis (P < 0.001), lung metastasis (P < 0.001), absence of molecular or histology-driven screening (P < 0.001), first-in-human trials (P < 0.001), dose-escalation cohorts (P = 0.011), and Royal Marsden Hospital (RMH) score >1 (P < 0.001) were significantly associated with shorter PFS. In multivariate analysis, independent prognostic factors for shorter PFS were myxoid liposarcoma (P = 0.031), ≥2 prior lines of treatment (P = 0.033), liver metastasis (P = 0.007), and RMH score >2 (P = 0.006). Factors associated with improved PFS were leiomyosarcomas (P = 0.010), molecular screening (P = 0.025), and histology-driven screening (P = 0.010). The median overall survival rates were 36.3, 12.6, and 9.2 months in the first, second, and later lines, respectively (P = 0.0067). The grade 3-4 toxicity rate was 36%. CONCLUSIONS Early phase trials provide an active therapeutic option for STS, even in first-line settings. Molecular screening and histology-driven trials further improve the clinical benefit.
Collapse
Affiliation(s)
- E F Nassif
- Cancer Medicine Department, Centre Léon Bérard, Lyon, France. https://twitter.com/NassifElise
| | - J-Y Blay
- Cancer Medicine Department, Centre Léon Bérard, Lyon, France. https://twitter.com/jeanyvesblay
| | - C Massard
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France. https://twitter.com/drcmassard
| | - A Dufresne
- Cancer Medicine Department, Centre Léon Bérard, Lyon, France
| | - M Brahmi
- Cancer Medicine Department, Centre Léon Bérard, Lyon, France
| | - P Cassier
- Early Phase Trial Unit, Centre Léon Bérard, Lyon, France
| | - I Ray-Coquard
- Cancer Medicine Department, Centre Léon Bérard, Lyon, France. https://twitter.com/CoquardRay
| | - P Pautier
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - A Leary
- Cancer Medicine Department, Gustave Roussy, Villejuif, France
| | - M-P Sunyach
- Radiation Oncology Department, Centre Léon Bérard, Lyon, France
| | - R Bahleda
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - A Levy
- Radiation Oncology Department, Gustave Roussy, Villejuif, France
| | - C Le Pechoux
- Radiation Oncology Department, Gustave Roussy, Villejuif, France
| | - C Honoré
- Surgical Oncology Department, Gustave Roussy, Villejuif, France
| | - O Mir
- Ambulatory Cancer Care Department, Gustave Roussy, Villejuif, France
| | - A Le Cesne
- International Department, Gustave Roussy, Villejuif, France.
| |
Collapse
|
41
|
Chihara D, Huang EP, Finnigan SR, Cordes LM, Skorupan N, Fukuda Y, Rubinstein LV, Ivy SP, Doroshow JH, Nastoupil LJ, Flowers CR, Takebe N. Trends in Grade 5 Toxicity and Response in Phase I Trials in Hematologic Malignancy: 20-Year Experience From the Cancer Therapy Evaluation Program at the National Cancer Institute. J Clin Oncol 2022; 40:1949-1957. [PMID: 35263120 DOI: 10.1200/jco.21.02190] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Cancer drug development has largely shifted from cytotoxic chemotherapy to targeted treatment in the past two decades. Although previous studies have highlighted improvement in response rates in recent phase I trials, disease-focused reporting is limited. METHODS We integrated patient-level data for patients with hematologic malignancies who participated in phase I trials sponsored by the National Cancer Institute Cancer Therapy Evaluation Program between January 2000 and May 2019 and estimated the trend of grade 5 toxicity and response by disease subtype over time. RESULTS We analyzed 161 trials involving 3,308 patients, all of whom were assessed for toxicity and 2,404 of whom were evaluable for response to therapy. The overall rate of grade 5 toxicities was 1.81% (95% CI, 1.36 to 2.27), with no significant change in the rate over time. Baseline characteristics associated with higher risk of grade 5 toxicity were age and performance status ≥ 2 at enrollment. Overall response rate (ORR) and complete response (CR) rate for all trials during the study period were 25.1% and 14.7%, respectively. A significant increase in both ORR and CR rate was observed over time (ORR: 18.5% in 2000-2005, 25.9% in 2006-2012, and 50.6% in 2013-2019, P < .001). ORR in phase I trials varied across disease subtypes: 20.2% in acute myeloid leukemia, 9.1% in myelodysplastic syndrome, 43.2% in lymphoma, 42.9% in chronic lymphocytic leukemia, 15.1% in acute lymphoblastic leukemia, and 16.5% in myeloma. CONCLUSION Over time, the ORR and CR rates in phase I trials for hematologic malignancy have improved meaningfully, whereas the rate of toxicity-related death remains stable. This study provides broad experience that physicians can use when discussing the potential outcomes for patients with hematologic malignancy considering participation in phase I trials.
Collapse
Affiliation(s)
- Dai Chihara
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX.,Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Erich P Huang
- Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Shanda R Finnigan
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa M Cordes
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Nebojsa Skorupan
- Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yoko Fukuda
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Larry V Rubinstein
- Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - S Percy Ivy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Loretta J Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christopher R Flowers
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
42
|
Gad KT, Lassen U, Duun-Henriksen AK, Dalton SO, Mau-Sørensen M, Bidstrup PE, Høeg BL, Rohrberg KS, Spanggard I, von Heymann A, Johansen C. Patients in phase 1 cancer trials: psychological distress and understanding of trial information. Acta Oncol 2022; 61:341-348. [PMID: 34709958 DOI: 10.1080/0284186x.2021.1993331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Psychological distress may be present among patients who are considering enrollment in phase 1 cancer trials, as they have advanced cancer and no documented treatment options remain. However, the prevalence of psychological distress has not been previously investigated in larger cohorts. In complex phase 1 cancer trials, it is important to ensure adequate understanding of the study premises, such as the undocumented effects and the risk of adverse events. MATERIALS AND METHODS In a prospective study, patients completed questionnaires at two time points. We investigated psychological distress, measured as stress, anxiety, and depression, among patients at their first visit to the phase 1 unit (N = 229). Further, we investigated the understanding of trial information among patients who were enrolled in a phase 1 cancer trial (N = 57). RESULTS We enrolled 75% of 307 eligible patients. We found a lower mean score of stress in our population compared to population norms, while the mean scores of anxiety and depression were higher. A total of 9% showed moderate to severe symptoms of anxiety and 11% showed moderate to severe symptoms of depression, which indicates higher levels than cancer patients in general. A total of 46 (81% of enrolled patients) completed questionnaires on trial information and consent. The understanding of the information on phase 1 cancer trials in these patients was slightly lower than the level reported for cancer trials in general. Some aspects relating to purpose, benefit, and additional risks were understood by fewer than half of the patients. CONCLUSION Our results suggest that distress is not as prevalent in the population of patients referred to phase 1 cancer trials as in the general cancer population. Although patients' understanding of trial information was reasonable, some aspects of complex phase 1 cancer trials were not easily understood by enrolled patients.
Collapse
Affiliation(s)
- Katrine T. Gad
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Lassen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Anne K. Duun-Henriksen
- Statistics and Data analysis, The Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Susanne O. Dalton
- Survivorship & Inequality in Cancer, The Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Clinical Oncology and Palliative Services, Zealand University Hospital, Naestved, Denmark
| | - Morten Mau-Sørensen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pernille E. Bidstrup
- Survivorship & Inequality in Cancer, The Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Beverley. L. Høeg
- Statistics and Data analysis, The Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Kristoffer S. Rohrberg
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Iben Spanggard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Annika von Heymann
- Late Effect Research Unit (CASTLE), Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christoffer Johansen
- Late Effect Research Unit (CASTLE), Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
43
|
Tsang M, DeBoer RJ, Garrett SB, Dohan D. Decision-making about clinical trial options among older patients with metastatic cancer who have exhausted standard therapies. J Geriatr Oncol 2022; 13:594-599. [PMID: 35125334 PMCID: PMC9232893 DOI: 10.1016/j.jgo.2022.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Mazie Tsang
- Division of Hematology/Oncology, University of California San Francisco, 505 Parnassus Avenue, Room M1286, Mailbox 1270, San Francisco, CA 94143, United States.
| | - Rebecca J DeBoer
- Division of Hematology/Oncology, University of California San Francisco, 505 Parnassus Avenue, Room M1286, Mailbox 1270, San Francisco, CA 94143, United States.
| | - Sarah B Garrett
- Institute for Health Policy Studies, University of California San Francisco, 490 Illinois Street, San Francisco, CA 94158, United States.
| | - Daniel Dohan
- Institute for Health Policy Studies, University of California San Francisco, 490 Illinois Street, San Francisco, CA 94158, United States.
| |
Collapse
|
44
|
Simonelli M, Persico P, Capucetti A, Carenza C, Franzese S, Lorenzi E, Dipasquale A, Losurdo A, Giordano L, Pessina F, Navarria P, Politi LS, Mavilio D, Locati M, Della Bella S, Santoro A, Bonecchi R. Immunotherapeutic early-phase clinical trials and malignant gliomas: A single-center experience and comprehensive immunophenotyping of circulating leukocytes. Neurooncol Adv 2021; 3:vdab160. [PMID: 34901858 PMCID: PMC8661084 DOI: 10.1093/noajnl/vdab160] [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] [Indexed: 02/05/2023] Open
Abstract
Background Immunotherapeutic early-phase clinical trials (ieCTs) increasingly adopt large expansion cohorts exploring novel agents across different tumor types. High-grade glioma (HGG) patients are usually excluded from these trials. Methods Data of patients with recurrent HGGs treated within multicohort ieCTs between February 2014 and August 2019 (experimental group, EG) at our Phase I Unit were retrospectively reviewed and compared to a matched control group (CG) of patients treated with standard therapies. We retrospectively evaluated clinical, laboratory, and molecular parameters through univariate and multivariate analysis. A prospective characterization of circulating leukocyte subpopulations was performed in the latest twenty patients enrolled in the EG, with a statistical significance cutoff of P < .1. Results Thirty HGG patients were treated into six ieCTs. Fifteen patients received monotherapies (anti-PD-1, anti-CSF-1R, anti-TGFβ, anti-cereblon), fifteen patients combination regimens (anti-PD-L1 + anti-CD38, anti-PD-1 + anti-CSF-1R). In the EG, median progression-free survival and overall survival (OS) from treatment initiation were 1.8 and 8.6 months; twelve patients survived more than 12 months, and two of them more than 6 years. Univariate analysis identified O6-methylguanine DNA methyltransferase (MGMT) promoter methylation and total protein value at six weeks as significantly correlated with a better outcome. Decreased circulating neutrophils and increased conventional dendritic cells levels lead to significantly better OS. Conclusions A subgroup of EG patients achieved remarkably durable disease control. MGMT promoter methylation identifies patients who benefit more from immunotherapy. Monitoring dynamic changes of innate immune cell populations may help to predict clinical outcomes.
Collapse
Affiliation(s)
- Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Pasquale Persico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Arianna Capucetti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Claudia Carenza
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Sara Franzese
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Lorenzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Angelo Dipasquale
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Agnese Losurdo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Laura Giordano
- IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Federico Pessina
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | | | - Letterio S Politi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Massimo Locati
- Unit of Leukocyte Biology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Silvia Della Bella
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Rozzano, Milan, Italy
| |
Collapse
|
45
|
Sabnis HS, Shulman DS, Mizukawa B, Bouvier N, Zehir A, Fangusaro J, Fabrizio VA, Whitlow C, Winchester M, Agresta L, Turpin B, Wechsler DS, DuBois SG, Glade-Bender J, Castellino SM, Shukla N. Multicenter Analysis of Genomically Targeted Single Patient Use Requests for Pediatric Neoplasms. J Clin Oncol 2021; 39:3822-3828. [PMID: 34591650 PMCID: PMC9851705 DOI: 10.1200/jco.21.01213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE The US Food and Drug Administration-expanded access program (EAP) uses a single patient use (SPU) mechanism to provide patient access to investigational agents in situations where no satisfactory or comparable therapy is available. Genomic profiling of de novo and relapsed or refractory childhood cancer has led to increased identification of new drug targets in the last decade. The aim of this study is to examine the SPU experience for genomically targeted therapies in patients with pediatric cancer. PATIENTS AND METHODS All genomically targeted therapeutic SPUs obtained over a 5-year period were evaluated at four large pediatric cancer programs. Data were collected on the type of neoplasm, agents requested, corresponding molecularly informed targets, and clinical outcomes. RESULTS A total of 45 SPUs in 44 patients were identified. Requests were predominantly made for CNS and solid tumors (84.4%) compared with hematologic malignancies (15.6%). Lack of an available clinical trial was the main reason for SPU initiation (64.4%). The median time from US Food and Drug Administration submission to approval was 3 days (range, 0-12 days) and from Institutional Review Board submission to approval was 5 days (range, 0-50 days). Objective tumor response was seen in 39.5% (15 of 38) of all evaluable SPUs. Disease progression was the primary reason for discontinuation of drug (66.7%) followed by toxicity (13.3%). CONCLUSION SPU requests remain an important mechanism for pediatric access to genomically targeted agents given the limited availability of targeted clinical trials for children with high-risk neoplasms. Furthermore, this subset of SPUs resulted in a substantial number of objective tumor responses. The development of a multi-institutional data registry of SPUs may enable systematic review of toxicity and clinical outcomes and provide evidence-based access to new drugs in rare pediatric cancers.
Collapse
Affiliation(s)
- Himalee S. Sabnis
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA,Emory University School of Medicine, Department of Pediatrics, Atlanta, GA,Himalee S. Sabnis, MD, MSc, The Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, 426J Emory Children's Center, 2015 Uppergate Dr, Atlanta, GA 30322; e-mail:
| | - David S. Shulman
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Benjamin Mizukawa
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH,University of Cincinnati College of Medicine, Cincinnati OH
| | - Nancy Bouvier
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jason Fangusaro
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA,Emory University School of Medicine, Department of Pediatrics, Atlanta, GA
| | - Vanessa A. Fabrizio
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Chanta Whitlow
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA
| | - Marilyn Winchester
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Laura Agresta
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH,University of Cincinnati College of Medicine, Cincinnati OH
| | - Brian Turpin
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH,University of Cincinnati College of Medicine, Cincinnati OH
| | - Daniel S. Wechsler
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA,Emory University School of Medicine, Department of Pediatrics, Atlanta, GA
| | - Steven G. DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Julia Glade-Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sharon M. Castellino
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA,Emory University School of Medicine, Department of Pediatrics, Atlanta, GA
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
46
|
Lee J, Gillam L, Visvanathan K, Hansford JR, McCarthy MC. Clinical Utility of Precision Medicine in Pediatric Oncology: A Systematic Review. JCO Precis Oncol 2021; 5:1088-1102. [DOI: 10.1200/po.20.00405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Precision medicine uses advanced molecular techniques to guide the use of targeted therapeutic drugs and is an emerging paradigm in pediatric oncology. Clinical evidence related to the efficacy of many novel targeted drugs, however, is currently very limited given the rarity of pediatric cancer and the lack of published evidence for the use of these drugs in children. This systematic review aimed to evaluate the existing evidence for the feasibility and clinical efficacy of precision medicine in pediatric oncology. METHODS A systematic review was conducted using the PubMed, Medline, and Embase databases. Clinical trials and observational studies, which used molecular assays such as whole-exome sequencing to identify molecular targets that guided the allocation of targeted cancer drugs and reported clinical outcomes, were included in this review. RESULTS Twenty-one clinical trials and observational studies were identified, collectively enrolling 1,408 pediatric patients across nine countries. Therapeutic targets were found in 647 patients (46.0%); however, only 175 of these patients (27.0%) received a targeted drug. Objective responses were recorded for 73 (41.7%) of these 175 patients, only 5.2% of the total sample. Inconsistent outcome reporting and limited comparison with conventional treatment hindered evaluation of the clinical utility of precision medicine. CONCLUSION Precision medicine can feasibly identify molecular targets in a clinical setting. However, the inaccessibility of targeted drugs is a significant barrier, restricting the exploration of its therapeutic potential in pediatric oncology. Future clinical trials should endeavor to link the molecular testing results with access to targeted drugs and standardize outcome reporting to advance understanding of the benefits of this novel paradigm in improving patient outcomes.
Collapse
Affiliation(s)
- Justin Lee
- Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
| | - Lynn Gillam
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
- Department of Human Bioethics, University of Melbourne, Melbourne, VIC, Australia
| | - Keshini Visvanathan
- Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
| | - Jordan R. Hansford
- Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Maria C. McCarthy
- Children's Cancer Centre, Royal Children's Hospital, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
47
|
Van Norman GA. Data Safety and Monitoring Boards Should Be Required for Both Early- and Late-Phase Clinical Trials. JACC Basic Transl Sci 2021; 6:887-896. [PMID: 34869954 PMCID: PMC8617574 DOI: 10.1016/j.jacbts.2021.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Gail A. Van Norman
- Address for correspondence: Dr Gail A. Van Norman, Department of Anesthesia and Pain Medicine, University of Washington, 2601 West Boston Street, Seattle, Washington 98199, USA.
| |
Collapse
|
48
|
Schupmann W, Li X, Wendler D. Do the Potential Medical Benefits of Phase 1 Pediatric Oncology Trials Justify the Risks? Views of the United States Public. J Pediatr 2021; 238:249-258.e3. [PMID: 34144034 PMCID: PMC8551010 DOI: 10.1016/j.jpeds.2021.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To assess the US public's views on whether the potential medical benefits of phase 1 pediatric oncology trials justify the risks. STUDY DESIGN Online survey of a nationally representative sample of US adults. Participants were presented with a hypothetical scenario in which they have a 10-year-old child with advanced cancer. They were then offered the option of giving their child supportive care or trying one more potential treatment, in the research or clinical care setting, which has the same risks and potential medical benefits as the average phase 1 pediatric oncology trial. We assessed what percentage of respondents thought the potential medical benefits justify the risks. RESULTS In total, 1658 of the 2508 individuals who were sent the survey participated (response rate = 66.1%). Of those who passed all 3 test questions indicating understanding, 67.1% in the research scenario and 58.5% in the clinical care scenario regarded the potential medical benefits of an average phase 1 pediatric oncology trial as equal to or greater than the risks. In addition, 53.4% of respondents in the research scenario thought it was appropriate for researchers to conduct a study in children with these risks and potential medical benefits, and 46.9% stated they would enroll their own child in such a trial. CONCLUSIONS A majority of the US public regards the potential medical benefits of average phase 1 pediatric oncology trials as justifying the risks. This finding suggests that these trials are ethically appropriate and approvable in patients who have no more effective treatment options. At the same time, a significant minority thought the potential medical benefits do not justify the risks, suggesting these trials should be approved only when they have significant social value. Moreover, approximately one-half of respondents regarded the trials as inappropriate and would not enroll their own child, underscoring the need for a rigorous informed consent process that accurately educates parents regarding the risks, potential medical benefits, and alternatives, so they can decide whether to enroll their child based on their own preferences and goals.
Collapse
Affiliation(s)
- Will Schupmann
- Department of Bioethics, NIH Clinical Center, Bethesda, MD
| | - Xiaobai Li
- Biostatistics and Clinical Epidemiology Service, NIH Clinical Center, Bethesda, MD
| | - David Wendler
- Department of Bioethics, NIH Clinical Center, Bethesda, MD.
| |
Collapse
|
49
|
Mackley MP, Fernandez NR, Fletcher B, Woolcott CG, Fernandez CV. Revisiting Risk and Benefit in Early Oncology Trials in the Era of Precision Medicine: A Systematic Review and Meta-Analysis of Phase I Trials of Targeted Single-Agent Anticancer Therapies. JCO Precis Oncol 2021; 5:17-26. [PMID: 34994588 DOI: 10.1200/po.20.00214] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Phase I trials are a crucial step in the evaluation of new cancer therapies. Historically, low rates of response (5%) and comparably high rates of death from toxicities (0.5%) have contributed to debates on the ethics and orientation of these trials. With the introduction of novel targeted therapies, a contemporary estimate is needed. METHODS We systematically searched PubMed, Embase, and ClinicalTrials.gov for reports of phase I oncology trials of single-agent targeted immunomodulators, molecularly targeted therapies, and antiangiogenic agents, published between January 2015 and July 2018. Adult and pediatric trials of solid and hematological malignancies were eligible. Treatment-related adverse events (grades 3, 4, and 5) and response rates (objective, complete, and partial) were extracted and analyzed. RESULTS One hundred and fifty-eight trial reports, covering 6,707 patients, were included. The rate of treatment-related deaths was 0.0% (95% CI, 0.0 to 0.1), while 13.2% of patients (9.5 to 17.3) experienced a grade 3 or 4 treatment-related toxicity. The combined objective response rate was 6.4% (4.6 to 8.5). Among trials using tumor biomarkers as eligibility criteria, the objective response rate was higher (12.0% [7.3 to 17.6] compared to 4.9% [2.5 to 5.7], P value < .01). The same was true of trials focusing on a single tumor type (13.4% [8.2 to 19.4]) compared to multiple tumor types (3.8% [2.5 to 5.3], P value < .01). CONCLUSION Reduced grade 5 risk and improved benefit appears to exist in modern phase I oncology trials, particularly in trials that target single tumor types and integrate biomarkers as eligibility criteria. These findings provide information to support informed consent discussions, highlight the need for improved reporting of phase I oncology trials, and provide direction for optimizing their design.
Collapse
Affiliation(s)
| | - Nicholas R Fernandez
- Department of Chemistry and Biochemistry, Mount Allison University, Sackville, NB, Canada
| | - Benjamin Fletcher
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom.,Centre for Patient Reported Outcome Research (CPROR), Institute of Applied Health Research (IAHR), University of Birmingham, Birmingham, United Kingdom
| | - Christy G Woolcott
- IWK Health Centre, Halifax, NS, Canada.,Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.,Department of Obstetrics and Gynaecology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Conrad V Fernandez
- IWK Health Centre, Halifax, NS, Canada.,Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.,Department of Bioethics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
50
|
Kazmi F, Shrestha N, Booth S, Dodwell D, Aroldi F, Foord T, Nicholson BD, Heesen P, Lord S, Yeoh KW, Blagden S. Next-generation sequencing for guiding matched targeted therapies in people with relapsed or metastatic cancer. Hippokratia 2021. [DOI: 10.1002/14651858.cd014872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Farasat Kazmi
- Department of Oncology; University of Oxford; Oxford UK
| | - Nipun Shrestha
- Department of Primary Care and Mental Health; University of Liverpool; Liverpool UK
| | - Stephen Booth
- Department of Oncology; University of Oxford; Oxford UK
| | - David Dodwell
- Nuffield Department of Population Health; University of Oxford; Oxford UK
| | | | | | - Brian D Nicholson
- Nuffield Department of Primary Care Health Sciences; University of Oxford; Oxford UK
| | | | - Simon Lord
- Department of Oncology; University of Oxford; Oxford UK
| | - Kheng-Wei Yeoh
- Radiation Oncology; National Cancer Centre; Singapore Singapore
| | - Sarah Blagden
- Department of Oncology; University of Oxford; Oxford UK
| |
Collapse
|