1
|
Eisenhauer EA, Abdihamid O, Booth CM, Cherny N, Fojo AT, Gyawali B, Marini BL, Mohyuddin GR, Pe M, Pond GR, Soto-Perez-de-Celis E, Tannock IF, Trapani D, Tregear M, van der Graaf WTA, Wilson BE. Guidance for discussants of randomized cancer trials at major meetings. Eur J Cancer 2025; 220:115357. [PMID: 40117861 DOI: 10.1016/j.ejca.2025.115357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Discussants of potentially practice-changing randomized clinical trials (RCTs) at major cancer meetings have an important responsibility to place new research in the context of current cancer care, to assess the generalizability of the data, to evaluate whether the outcomes are meaningful to patients, and to convey this information effectively and objectively to a diverse audience. Without a standard approach to critiquing clinical trial design or results discussants may overlook key weaknesses in their commentary. COMMON SENSE ONCOLOGY (CSO) The CSO initiative was launched in 2023 and is now comprised of an international collective of > 1000 clinicians, academics, policymakers, and patients. Its primary vision is that patients should have access to cancer treatments that provide meaningful improvements in outcomes, irrespective of where they live. To do this, one focus is to try to improve evidence generation and reporting. GUIDANCE FOR DISCUSSANTS As part of this work, the CSO RCT Working Group has identified key elements for use in the development of discussant presentations to facilitate a balanced high-quality examination of RCTs. Elements include assessment of: a) Study design: evaluation of the study question, selection of population and control arm, use of blinding, choice of primary and secondary endpoints; b) Study results: treatment delivery, use of crossover, impact of censoring, unplanned analyses, patient reported outcomes, adverse effects; and c) Conclusions: Appraise the value and generalizability of trial results and, when positive results are claimed, assess if they offer meaningful benefits over current standard(s) of care in outcomes of importance to patients.
Collapse
Affiliation(s)
| | | | | | | | - Antonio T Fojo
- Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | - Madeline Pe
- European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | | | - Enrique Soto-Perez-de-Celis
- University of Colorado, Denver, CO, USA; Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Ian F Tannock
- Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
2
|
Sanford NN, Shi Q, Hein DM, Hall WA. Benchmarks of success in radiotherapy vs systemic therapy: National Clinical Trials Network (NCTN) randomized controlled trials sponsored by the National Cancer Institute (NCI). J Natl Cancer Inst 2025; 117:879-889. [PMID: 39656956 DOI: 10.1093/jnci/djae313] [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: 08/28/2024] [Revised: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND The National Clinical Trials Network (NCTN) is the largest government-sponsored organization in the United States; it is tasked with funding randomized controlled trials in oncology. It is unknown whether there are differences in study design by treatment modality. We evaluated differences in methodology between trials testing radiation therapy (RT) vs systemic therapy. METHODS The Clinical Trials Support Unit website was used to identify active randomized controlled trials of systemic therapy or RT across NCTN cooperative groups through December 31, 2023. Studies in disease sites with more than 5 RT trials were included. Each trial's protocol was reviewed to obtain key design information that was descriptively compared: primary endpoint, hypothesis testing type (superiority vs noninferiority), noninferiority margin, hypothesized effect size, power, and statistical significance level. RESULTS A total of 186 randomized controlled trials (142 systemic therapy, 44 RT) were examined. Comparing primary endpoints, 59.1% vs 26.8% of RT vs systemic therapy trials, respectively, had a primary endpoint of overall survival. Nearly one-third (31.2%) of RT trials were noninferiority vs 6.3% of systemic therapy trials. Among breast cancer trials, 75% of RT studies were noninferiority vs 11.1% systemic therapy. Target effect size, power, and statistical significance level were similar by treatment modality within tumor types and disease settings. CONCLUSION Among NCTN cooperative group randomized controlled trials, there were marked differences in study design between RT vs systemic therapy trials. A higher benchmark for defining success for RT interventions was observed, with greater emphasis on overall survival as the primary endpoint. This finding may reflect differences in therapeutic mechanism by modality and types of study questions posed.
Collapse
Affiliation(s)
- Nina N Sanford
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, United States
| | - David M Hein
- Lyda Hill Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - William A Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| |
Collapse
|
3
|
Cardoso Borges F, van der Graaf WTA, Saesen R, Aebi S, Amariutei AE, Bekelman J, Gorlia T, Hulstaert F, Huys I, Kluetz P, Morris MJ, Patil V, Prindiville SA, Schilsky RL, Thomson A, Treweek S, Weller M, Zuidgeest M, Retel V, Lacombe D. Defining the role of pragmatic clinical trials in cancer clinical research: outcomes of a collaborative workshop hosted by the European Organisation for Research and Treatment of Cancer. Lancet Oncol 2025; 26:e253-e263. [PMID: 40318657 DOI: 10.1016/s1470-2045(24)00756-3] [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: 10/23/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 05/07/2025]
Abstract
Explanatory clinical trials, which focus on evaluating therapeutic efficacy under ideal circumstances, are crucial for learning about new therapeutic interventions; however, they also exhibit shortcomings. These include non-representative populations and frequent use of intermediate endpoints, leading to uncertainty about the applicability of study results to patients in the real-world. Moreover, these trials often do not address all clinically meaningful questions, highlighting the need for optimisation within the oncology research framework. Refinements can be partly achieved by incorporating more pragmatic elements into cancer clinical trials. At a virtual European Organisation for Research and Treatment of Cancer workshop, key stakeholders convened to discuss the methodological characteristics and value of pragmatic trials, which focus on evaluating effectiveness in routine clinical practice, and their capacity to address the efficacy-effectiveness gap. This Policy Review outlines and discusses some of the views and perspectives expressed on the role of pragmatic trials in the current framework and their ability to inform decision making, and the recommended priorities for enhancing pragmatism in cancer clinical research.
Collapse
Affiliation(s)
- Fábio Cardoso Borges
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium; Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| | - Winette T A van der Graaf
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium; Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Medical Oncology, ErasmusMC Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Robbe Saesen
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium; Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Stefan Aebi
- Faculty of Medicine, University of Berne, Berne, Switzerland; Cancer Center, Lucerne Cantonal Hospital, Lucerne, Switzerland
| | - Ana E Amariutei
- European Patient Advocacy Institute, Riemerling, Munich, Germany
| | - Justin Bekelman
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thierry Gorlia
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Frank Hulstaert
- Belgian Health Care Knowledge Centre (KCE), Brussels, Belgium
| | - Isabelle Huys
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Paul Kluetz
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, MD, USA
| | - Michael J Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vijay Patil
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sheila A Prindiville
- Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Shaun Treweek
- Aberdeen Centre for Evaluation, University of Aberdeen, Aberdeen, UK
| | - Michael Weller
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium; Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland; Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Mira Zuidgeest
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Valesca Retel
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands; Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Denis Lacombe
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| |
Collapse
|
4
|
Zuellig JA, Adam R, Udry F, Tibau A, Šeruga B, Ocaña A, Amir E, Templeton AJ. The Effect of Staging Intervals on Progression-Free Survival in Registration Studies of Oncologic Drugs: A Meta-Analysis. Cancers (Basel) 2025; 17:1359. [PMID: 40282533 PMCID: PMC12025954 DOI: 10.3390/cancers17081359] [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: 02/10/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES To study whether shorter restaging intervals are associated with lower hazard ratios (HRs) for progression-free survival (PFS), as suggested in breast cancer. METHODS Studies supporting the registration of oncologic drugs in Switzerland from 2010 to 2022 were analyzed. HRs and 95% confidence intervals (CIs) for PFS were pooled in a meta-analysis using the generic inverse-variance method and a random-effects model in RevMan v5.4. The HRs were stratified by restaging intervals ( RESULTS A total of 112 studies comprising 69,579 patients were included. The median restaging interval was 8 weeks, with a range of 4 to 18 weeks. Longer restaging intervals (≥8 weeks) were associated with lower HRs compared to shorter intervals (<8 weeks), with pooled HRs of 0.48 (95% CI: 0.44-0.52) and 0.58 (95% CI: 0.53-0.63), respectively. The difference between the groups was statistically significant (p = 0.005), with a substantial heterogeneity (Cochran's Q p < 0.001; I2 = 90%). Subgroup analyses based on treatment type, including immunotherapy, monoclonal antibodies, and tyrosine kinase inhibitors, did not show any statistically significant differences in HRs. Studies of melanoma with shorter staging intervals were associated with lower HRs (0.44 vs. 0.58, p = 0.02), whereas shorter interval studies of kidney cancer had higher HRs (0.67 vs. 0.44, p = 0.01). Sensitivity analyses with other cut-offs and a meta-regression yielded similar results. CONCLUSIONS Studies leading to the authorization of drugs to treat incurable solid tumors applying restaging intervals ≥ 8 weeks were associated with lower HRs for PFS. The potential impact of restaging intervals on the results for PFS warrants further investigation.
Collapse
Affiliation(s)
- Jonas A. Zuellig
- Faculty of Medicine, University of Basel, 4001 Basel, Switzerland; (J.A.Z.); (R.A.); (F.U.)
| | - Roman Adam
- Faculty of Medicine, University of Basel, 4001 Basel, Switzerland; (J.A.Z.); (R.A.); (F.U.)
| | - Filomena Udry
- Faculty of Medicine, University of Basel, 4001 Basel, Switzerland; (J.A.Z.); (R.A.); (F.U.)
| | - Ariadna Tibau
- Oncology Department, Hospital de la Santa Creu i Sant Pau, Institut d’Investigació Biomèdica Sant Pau, 08041 Barcelona, Spain;
- Departament de Medicina, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Bostjan Šeruga
- Department of Medical Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Alberto Ocaña
- Experimental Therapeutics Unit, Medical Oncology Department, Hospital Clínico Universitario San Carlos IdISSC, 28040 Madrid, Spain;
- Instituto de Investiagción Sanitaria del Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Center, Toronto, ON M5S 1A1, Canada;
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Arnoud J. Templeton
- Faculty of Medicine, University of Basel, 4001 Basel, Switzerland; (J.A.Z.); (R.A.); (F.U.)
- Department of Medical Oncology, St. Claraspital, 4058 Basel, Switzerland
- St. Clara Research, 4058 Basel, Switzerland
| |
Collapse
|
5
|
Eldridge L, Goodman NR, Chtourou A, Galassi A, Monge C, Cira MK, Pearlman PC, Loehrer PJ, Gopal S, Ginsburg O. Barriers and Opportunities for Cancer Clinical Trials in Low- and Middle-Income Countries. JAMA Netw Open 2025; 8:e257733. [PMID: 40293747 PMCID: PMC12038506 DOI: 10.1001/jamanetworkopen.2025.7733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/18/2025] [Indexed: 04/30/2025] Open
Abstract
Importance Clinical trials represent the gold standard to test the safety and efficacy of new or updated approaches to treatments that will inform quality cancer care. However, cancer trials enroll few patients in low- and middle-income countries (LMICs), are often led by investigators from high-income countries, and do not adequately reflect global disease burden or population diversity. Objective To identify key challenges and strategies to advance contextually relevant, quality cancer trials in LMICs. Design, Setting, and Participants The survey used in this survey study was available in English, Arabic, French, Portuguese, and Spanish and was conducted by the US National Cancer Institute from October 18 to December 22, 2023. Clinicians with experience in cancer therapeutic clinical trials in LMICs were eligible. The survey covered their professional background and views on challenges and strategies for improving clinical trial opportunities in LMICs. Analysis was performed from April 2 to August 26, 2024. Main Outcomes and Measures Respondents were asked to rate 34 challenges by impact on their ability to conduct cancer trials using a 4-point Likert scale and 8 strategies by importance using a 5-point Likert scale. Descriptive statistics summarized participants' backgrounds, challenges, and priorities. Results Of 453 respondents who began the survey, a total of 223 (49%) were eligible for inclusion, and 131 of those (59%) completed the survey in full. Among the 133 respondents who provided gender data, 81 (61%) were male. In all, 107 of 130 respondents (82%) were affiliated with LMIC institutions, 65 of 223 (29%) were medical oncologists, and 52 of 133 (39%) were midcareer. Financial challenges were rated as the most impactful, with 133 of 170 respondents (78%) rating difficulty obtaining funding for investigator-initiated trials as having a large impact on ability to carry out a trial. Human capacity issues followed, with 105 of 192 respondents (55%) rating lack of dedicated research time as having a large impact. Increasing opportunities for funding and improving human capacity were reported as key strategies to advance capacity to conduct clinical trials in LMICs. Conclusions and Relevance This survey study of clinicians with clinical trial experience in LMICs suggests that adequate funding and a well-trained research workforce are 2 predominant challenges to advancing cancer therapeutic clinical trials in LMICs. Understanding these obstacles can inform efforts to support cancer clinical trials that better reflect worldwide needs and diversity by prioritizing and sustaining research led by LMIC investigators.
Collapse
Affiliation(s)
- Linsey Eldridge
- Center for Global Health, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Nina R. Goodman
- Office of Communications and Public Liaison, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Amina Chtourou
- Center for Global Health, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | | | - Cecilia Monge
- Center for Global Health, National Cancer Institute, National Institutes of Health, Rockville, Maryland
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mishka Kohli Cira
- Center for Global Health, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Paul C. Pearlman
- Center for Global Health, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Patrick J. Loehrer
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis
| | - Satish Gopal
- Center for Global Health, National Cancer Institute, National Institutes of Health, Rockville, Maryland
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ophira Ginsburg
- Center for Global Health, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
6
|
Antonarelli G, Pérez-García JM, Gion M, Rugo H, Schmid P, Bardia A, Hurvitz S, Harbeck N, Tolaney SM, Curigliano G, Llombart-Cussac A, Cortés J. Redefining clinical trial strategic design to support drug approval in medical oncology. Ann Oncol 2025:S0923-7534(25)00111-5. [PMID: 40086733 DOI: 10.1016/j.annonc.2025.03.005] [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: 01/27/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Randomized clinical trials represent the gold standard for the introduction of innovative therapies in medical oncology, and they provide the highest level of evidence to ascertain the clinical activity of new drugs or novel combinations. However, the current infrastructure of clinical trials supporting innovative drug approvals is challenged by an increased body of knowledge concerning tumor biology and therapy resistance, a fast-growing armamentarium of novel anticancer compounds, an impressively upscaled data analysis capacity, as well as increasing costs related to clinical trials management. In this scenario, modern clinical trial designs need to evolve to expedite new drug approvals by tailoring patients' treatment strategies according to their medical needs. Balanced, patient-oriented clinical trial designs are eagerly warranted to increase their efficiency, to include the fast pace of technological innovations and scientific discoveries, and, ultimately, to face the challenges of the modern medical oncology field.
Collapse
Affiliation(s)
- G Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - J M Pérez-García
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, USA; International Breast Cancer Center (IBCC), Pangaea Oncology, Quirón Group, Barcelona
| | - M Gion
- Hospital Universitario Ramón y Cajal, Madrid, Spain; IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid
| | - H Rugo
- Department of Medicine, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, USA
| | - P Schmid
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - A Bardia
- University of California Los Angeles (UCLA), Los Angeles
| | - S Hurvitz
- Fred Hutchinson Cancer Center, University of Washington School of Medicine, Seattle, USA
| | - N Harbeck
- Breast Center, Department of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, Germany
| | - S M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - G Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - A Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Arnau de Vilanova Hospital, Universidad Católica de Valencia, Valencia
| | - J Cortés
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, USA; IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain; Oncology Department, Hospital Universitario Torrejón, Ribera Group, Madrid, Spain.
| |
Collapse
|
7
|
Alkhudair N, Howaidi J, Alnuhait M, Alshamrani M, Khan M, Alharbi A, Alnajjar F, Bajnaid E, Almodaheem H, Alhowimel M, Alzahrani A, Khardaly A, Alnahedh M, Elsoudi H, Alabdulkareem H, Alrashidan A, Alzahrani M, Alrajhi A. Revitalizing oncology medications access in Saudi Arabia: Current challenges and recommendations by the Saudi Oncology Pharmacy Assembly. J Oncol Pharm Pract 2025; 31:245-250. [PMID: 38377985 PMCID: PMC11898367 DOI: 10.1177/10781552241232697] [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: 10/25/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
BackgroundCancer care is posing immense challenges to healthcare systems globally. Advances in screening, monitoring, and treating cancer improved patient outcomes and survival rates yet amplified the disease burden. Multiple barriers might impede early access to innovative therapies. We thoroughly examined the current challenges in oncology medication access in Saudi Arabia and provided consensus recommendations to revitalize the process.MethodsA focus group discussion was conducted. Expert healthcare providers (pharmacists and physicians) were invited to participate based on prespecified criteria. The research team conducted a qualitative analysis of the discussion to identify themes and formulate recommendations.ResultsFourteen experts were equally distributed into two groups, limiting the number in each group to 7. Pharmacists were 12 (∼86%), and physicians were 2 (∼14%). Ten were practicing in governmental hospitals, four representing different sectors; regulatory bodies, including Ministry of Health, National Unified Procurement Company, and Saudi Food and Drug Authority. Five themes were identified: national cancer burden, local data availability, pharmacoeconomic evaluation, patients reported outcomes, administration, and procurement. Consensus recommendations were formulated to optimize the formulary management process, enabling informed decision-making and facilitating early medication access for cancer patients.ConclusionsThe formulary management process can be enhanced by addressing the national cancer burden, promoting local data availability, conducting pharmacoeconomic evaluations, focusing on patient outcomes, and improving administration and procurement procedures. Implementing these recommendations can improve access to oncology medications and improve patient care outcomes in Saudi Arabia.
Collapse
Affiliation(s)
- Nora Alkhudair
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Jude Howaidi
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Mohammed Alnuhait
- Department of Clinical Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Majed Alshamrani
- Pharmaceutical Care Services, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Mansour Khan
- Pharmaceutical Care Services, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Atika Alharbi
- Pharmaceutical Care Services, Ministry of National Guard Health Affairs, Jeddah, Saudi Arabia
| | - Fouad Alnajjar
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Eshtyag Bajnaid
- Pharmaceutical Care Services Administration, King Abdullah Medical City, Makkah, Saudi Arabia
| | - Hajer Almodaheem
- Deputyship of Therapeutic Affairs, Ministry of Health, Riyadh, Saudi Arabia
| | - Mansour Alhowimel
- Unified Procurement, National Unified Procurement Company, Riyadh, Saudi Arabia
| | - Ali Alzahrani
- Medical Oncology Department, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Amr Khardaly
- Deputyship of Therapeutic Affairs, Ministry of Health, Riyadh, Saudi Arabia
| | - Mohammed Alnahedh
- Pharmaceutical Care Division, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Hamdi Elsoudi
- Clinical Pharmacy Department, Pharmaceutical Care Services Administration, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Hana Alabdulkareem
- Drug Policy and Economic Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Doctoral School of Applied Informatics and Applied Mathematics, Óbuda University, Budapest, Hungary
| | - Ahmed Alrashidan
- Corporate pharmaceutical planning, logistic & contracts management, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Musa Alzahrani
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah Alrajhi
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh, Saudi Arabia
- Department of Pharmacy Practice, College of Pharmacy, AlFaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Hopkins L, Clemons M, Bemister K, Booth C, Kadar S, Karanicolas P, Mulligan J, Savard MF, Tannock I, Tone A, MacKay H. Putting Patients First: Pragmatic Trials in Gynecologic Oncology. Curr Oncol 2025; 32:139. [PMID: 40136343 PMCID: PMC11941110 DOI: 10.3390/curroncol32030139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
In November 2024, the Society of Gynecologic Oncology of Canada hosted a 2-day, interdisciplinary Pragmatic Clinical Trials (PCTs) Workshop with the goal of launching an initiative to develop and promote PCTs within the Canadian gynecologic oncology research environment. The programme brought together multiple stakeholders, including patients with ovarian cancer, patient advocates, experts in PCTs, gynecologic oncologists, medical oncologists and clinical fellows. Foundational elements of pragmatism were emphasized in the context of the primary goal of PCTs, showing the real-world effectiveness of interventions in broad patient groups. Examples of how PCT outcomes can inform and influence clinical decision making and health policy were presented in the context of those outcomes that matter most to patients with cancer. The patients and patient advocates had the essential role of helping clinical investigators co-design PCT protocols to answer common, important, and practical questions that focus on outcomes that matter to patients. These endpoints included overall survival, quality of life and promotion of informed patient decision making. Tangible workshop outcomes included the development of several new proposals for PCTs inspirited and directed by the patient voice. Further educational initiatives to engage clinical gynecologic oncology investigators at all stages in their career are being planned.
Collapse
Affiliation(s)
- Laura Hopkins
- Department of Oncology, University of Saskatchewan, Saskatoon, SK S7V 4H4, Canada
| | - Mark Clemons
- Department of Oncology, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (M.C.); (M.-F.S.)
| | - Karen Bemister
- Patient Partners in Research, Ovarian Cancer Canada, Toronto, ON M2P 2A9, Canada; (K.B.)
| | - Chris Booth
- Department of Oncology, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Shannon Kadar
- Patient Partners in Research, Ovarian Cancer Canada, Toronto, ON M2P 2A9, Canada; (K.B.)
| | - Paul Karanicolas
- Department of Surgery, University of Toronto, Toronto, ON M5S 3H2, Canada;
| | - Julie Mulligan
- Patient Partners in Research, Ovarian Cancer Canada, Toronto, ON M2P 2A9, Canada; (K.B.)
| | - Marie-France Savard
- Department of Oncology, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (M.C.); (M.-F.S.)
| | - Ian Tannock
- Division of Medical Oncology, University of Toronto, Toronto, ON M5S 3H2, Canada;
| | - Alicia Tone
- Ovarian Cancer Canada, Toronto, ON M2P 2A9, Canada;
| | - Helen MacKay
- Division of Medical Oncology and Hematology, University of Toronto, Toronto, ON M4N 3M5, Canada;
| |
Collapse
|
9
|
Saccenti L, Varble N, Borde T, Mikhail AS, Kassin M, Levy E, Xu S, Hazen LA, Ukeh I, Vasco C, Duffy AG, Xie C, Monge C, Mabry D, Greten TF, Wood BJ. Quantifying morphologic variations as an alternate to standard response criteria for unresectable primary liver tumors after checkpoint inhibition therapy. LA RADIOLOGIA MEDICA 2025; 130:226-234. [PMID: 39656418 PMCID: PMC11870906 DOI: 10.1007/s11547-024-01937-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE The aim of this study was to assess the feasibility of quantifying morphologic changes in tumors during immunotherapy, as a reflection of response or survival. METHODS AND MATERIALS A retrospective single-center analysis was performed in patients with unresectable liver cancer previously enrolled in clinical trials combining immunotherapy (tremelimumab ± durvalumab) and locoregional treatment (either ablation or transarterial chemoembolization). Conventional response (RECIST 1.1) was assessed at 6-month follow-up. For morphologic assessment, the largest target lesion was manually segmented on axial slices in two dimensions using contrast-enhanced CT. Solidity and circularity of tumors were calculated at baseline, 3-month follow-up, and at 6-months follow-up. Survival analysis was performed. RESULTS From the 68 patients enrolled in clinical trials, 28 did not have target lesions separate from lesions treated by locoregional therapies, and 3 had no follow-up imaging. Thirty-seven patients (9 with biliary cancer and 28 with hepatocellular carcinoma) were included. Shape features and shape variation were not correlated with RECIST 1.1 status at 6-month follow-up. However, patients with low solidity tumors at 6-month follow-up showed poorer prognosis compared with patients with high solidity tumors at 6-month follow-up (p = 0.01). Solidity variation analysis confirmed that a decrease of tumor solidity at 6-month follow-up was associated with poorer prognosis (p = 0.01). No association was found between shape features at baseline or shape features at 3-month follow-up with overall survival. CONCLUSION Evolution and variation of tumor morphology during treatment may reflect or correlate with outcomes and contribute toward adapted response criteria.
Collapse
Affiliation(s)
- Laetitia Saccenti
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA.
- Henri Mondor's Institute of Biomedical Research - Inserm, U955 Team No. 18, Créteil, France.
| | - Nicole Varble
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
- Philips Research of North America, Cambridge, MA, USA
| | - Tabea Borde
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Andrew S Mikhail
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Michael Kassin
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Elliot Levy
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Sheng Xu
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Lindsey A Hazen
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Ifechi Ukeh
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Cyndi Vasco
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Austin G Duffy
- Gastrointestinal Malignancies Section, Thoracic and GI Malignancies Branch, Center for Cancer, Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Changqing Xie
- Gastrointestinal Malignancies Section, Thoracic and GI Malignancies Branch, Center for Cancer, Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Cecilia Monge
- Gastrointestinal Malignancies Section, Thoracic and GI Malignancies Branch, Center for Cancer, Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Donna Mabry
- Gastrointestinal Malignancies Section, Thoracic and GI Malignancies Branch, Center for Cancer, Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tim F Greten
- Gastrointestinal Malignancies Section, Thoracic and GI Malignancies Branch, Center for Cancer, Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
10
|
Gyawali B, Eisenhauer EA, van der Graaf W, Booth CM, Cherny NI, Goodman AM, Koven R, Pe ML, Marini BL, Mohyuddin GR, Pond GR, Sengar M, Soto-Perez-de-Celis E, Trapani D, Tregear M, Wilson BE, Tannock IF. Common Sense Oncology principles for the design, analysis, and reporting of phase 3 randomised clinical trials. Lancet Oncol 2025; 26:e80-e89. [PMID: 39914429 DOI: 10.1016/s1470-2045(24)00451-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 05/07/2025]
Abstract
Common Sense Oncology (CSO) prioritises treatments providing meaningful benefits for people with cancer. Here, we describe CSO principles aimed at improving the design, analysis, and reporting of randomised, controlled, phase 3 clinical trials evaluating cancer treatments. These principles include: (1) control treatment should be the best current standard of care; (2) the preferred primary endpoint is overall survival or a validated surrogate; (3) an absolute measure of benefit should be provided, such as the difference between groups in median overall survival times or the proportion of surviving patients at a prespecified time; (4) health-related quality of life should be at least a secondary endpoint; (5) toxicity should be described objectively without subjective language diminishing its importance; (6) trials should be designed to show or rule out clinically meaningful differences in outcomes, rather than a statistically significant difference alone; (7) censoring should be detailed, and sensitivity analyses done to determine its possible effects; (8) experimental treatments known to improve overall survival at later disease stages should be offered and funded for patients progressing in the control group; and (9) reports of trials should include a lay-language summary. We include checklists to guide compliance with these principles. By encouraging adherence, CSO aims to ensure that clinical trials yield results that are scientifically robust and meaningful to patients.
Collapse
Affiliation(s)
- Bishal Gyawali
- Division of Cancer Care and Epidemiology, Cancer Research Institute, Queen's University, Kingston, ON, Canada; Department of Oncology, Queen's University, Kingston, ON, Canada; Department of Public Health Sciences, Queen's University, Kingston, ON, Canada
| | | | - Winette van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Medical Oncology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Christopher M Booth
- Division of Cancer Care and Epidemiology, Cancer Research Institute, Queen's University, Kingston, ON, Canada; Department of Oncology, Queen's University, Kingston, ON, Canada; Department of Public Health Sciences, Queen's University, Kingston, ON, Canada
| | - Nathan I Cherny
- Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Aaron M Goodman
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, San Diego, CA, USA
| | - Rachel Koven
- Patient & Family Advisory Council, Southeast Regional Cancer Program, Kingston Health Sciences Centre, Kingston, ON, Canada; Division of Cancer Care and Epidemiology, Queen's University, Kingston, ON, Canada
| | - Madeline L Pe
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - Bernard L Marini
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Ghulam Rehman Mohyuddin
- Division of Hematology and Hematologic Malignancies, Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Gregory R Pond
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Manju Sengar
- Tata Memorial Centre, Affiliated to Homi Bhabha National Institute, Mumbai, India
| | - Enrique Soto-Perez-de-Celis
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Geriatrics, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Dario Trapani
- European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Brooke E Wilson
- Division of Cancer Care and Epidemiology, Cancer Research Institute, Queen's University, Kingston, ON, Canada; Department of Oncology, Queen's University, Kingston, ON, Canada; Department of Public Health Sciences, Queen's University, Kingston, ON, Canada
| | - Ian F Tannock
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Singh J, Anwer S, Palmer S, Saramago P, Thomas A, Dias S, Soares MO, Bujkiewicz S. Multi-indication Evidence Synthesis in Oncology Health Technology Assessment: Meta-analysis Methods and Their Application to a Case Study of Bevacizumab. Med Decis Making 2025; 45:17-33. [PMID: 39555661 PMCID: PMC11645851 DOI: 10.1177/0272989x241295665] [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: 11/07/2023] [Accepted: 08/15/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Multi-indication cancer drugs receive licensing extensions to include additional indications, as trial evidence on treatment effectiveness accumulates. We investigate how sharing information across indications can strengthen the inferences supporting health technology assessment (HTA). METHODS We applied meta-analytic methods to randomized trial data on bevacizumab, to share information across oncology indications on the treatment effect on overall survival (OS) or progression-free survival (PFS) and on the surrogate relationship between effects on PFS and OS. Common or random indication-level parameters were used to facilitate information sharing, and the further flexibility of mixture models was also explored. RESULTS Treatment effects on OS lacked precision when pooling data available at present day within each indication separately, particularly for indications with few trials. There was no suggestion of heterogeneity across indications. Sharing information across indications provided more precise estimates of treatment effects and surrogacy parameters, with the strength of sharing depending on the model. When a surrogate relationship was used to predict treatment effects on OS, uncertainty was reduced only when sharing effects on PFS in addition to surrogacy parameters. Corresponding analyses using the earlier, sparser (within and across indications) evidence available for particular HTAs showed that sharing on both surrogacy and PFS effects did not notably reduce uncertainty in OS predictions. Little heterogeneity across indications meant limited added value of the mixture models. CONCLUSIONS Meta-analysis methods can be usefully applied to share information on treatment effectiveness across indications in an HTA context, to increase the precision of target indication estimates. Sharing on surrogate relationships requires caution, as meaningful precision gains in predictions will likely require a substantial evidence base and clear support for surrogacy from other indications. HIGHLIGHTS We investigated how sharing information across indications can strengthen inferences on the effectiveness of multi-indication treatments in the context of health technology assessment (HTA).Multi-indication meta-analysis methods can provide more precise estimates of an effect on a final outcome or of the parameters describing the relationship between effects on a surrogate endpoint and a final outcome.Precision of the predicted effect on the final outcome based on an effect on the surrogate endpoint will depend on the precision of the effect on the surrogate endpoint and the strength of evidence of a surrogate relationship across indications.Multi-indication meta-analysis methods can be usefully applied to predict an effect on the final outcome, particularly where there is limited evidence in the indication of interest.
Collapse
Affiliation(s)
- Janharpreet Singh
- Biostatistics Research Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Sumayya Anwer
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Stephen Palmer
- Centre for Health Economics, University of York, York, UK
| | - Pedro Saramago
- Centre for Health Economics, University of York, York, UK
| | - Anne Thomas
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Sofia Dias
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Marta O Soares
- Centre for Health Economics, University of York, York, UK
| | - Sylwia Bujkiewicz
- Biostatistics Research Group, Department of Population Health Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
12
|
Wu J, Ge Y, Zhu G, Gao R, Zhu X, Zhang Y, Li J. Combination of Compound Kushen injection with first-line treatment versus first-line treatment alone for advanced colorectal cancer: a study protocol for a multicenter, openlabel, randomized controlled trial. BMC Complement Med Ther 2024; 24:429. [PMID: 39741233 DOI: 10.1186/s12906-024-04725-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND The treatment of advanced colorectal cancer (CRC) has progressed slowly, with chemotherapy combined with targeted therapy being the first-line treatment for the disease, but the improvement in efficacy is not satisfactory. Compound Kushen injection (CKI) is one of the representative drugs of anti-cancer Chinese herbal injection drugs, which has been widely used in the adjunct treatment of cancer in China. The aim of this trial is to evaluate the efficacy and safety of CKI combined with first-line treatment of advanced CRC. METHODS This is a multicenter, randomized, open-label controlled clinical trial in which 320 patients with advanced CRC will be randomly assigned to the treatment group or the control group in a 1:1 ratio. Both groups will receive at least 4 cycles of first-line therapy (FOLFOX/FOLFIRI/CAPEOX ± cetuximab/bevacizumab) in 14-21 day cycles, and the experimental group will receive additional CKI with a cumulative dose of 200 ml per cycle. Patients who achieve a complete response, partial response, or stable disease after 4-6 months will receive maintenance therapy until disease progression or another endpoint event, such as toxicity or death, occurs.. Follow-up will occur every 3 months until death or loss to follow-up. The primary outcome of this study will be progression-free survival (PFS). Secondary outcomes will be overall survival (OS), 1-year OS rate, 1-year PFS rate, objective response rate,disease control rate, symptoms and quality of life evaluation. Safety outcomes will be incidence of adverse events. DISCUSSION This study will be the first randomized controlled trial to investigate the efficacy and safety of CKI when combined with first-line treatment in the treatment of advanced CRC, with PFS as the primary outcome. It aims to clarify the clinical advantages and therapeutic effect of CKI in the treatment of advanced CRC. To identify the benefit population of CKI in the treatment of patients with advanced CRC, an enrichment design based on biomarkers will be utilized. Metabolomics and gut microbiota analysis will be conducted on biological samples to explore the metabolic and gut microbiota differences associated with the efficacy of CKI, guiding further research into its mechanism of action. TRIAL REGISTRATION ClinicalTrials.govNCT05894694. Registered on 4 August 2023.
Collapse
Affiliation(s)
- Jingyuan Wu
- Oncology Department, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beixian Pavilion, No.5, Xicheng District, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yuansha Ge
- Oncology Department, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beixian Pavilion, No.5, Xicheng District, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Guanghui Zhu
- Oncology Department, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beixian Pavilion, No.5, Xicheng District, Beijing, China
| | - Ruike Gao
- Oncology Department, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beixian Pavilion, No.5, Xicheng District, Beijing, China
| | - Xiaoyu Zhu
- Oncology Department, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beixian Pavilion, No.5, Xicheng District, Beijing, China
| | - Ying Zhang
- Oncology Department, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beixian Pavilion, No.5, Xicheng District, Beijing, China.
| | - Jie Li
- Oncology Department, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beixian Pavilion, No.5, Xicheng District, Beijing, China.
| |
Collapse
|
13
|
Ording AG, Skjøth F, Poulsen LØ, Szejniuk WM, Jakobsen E, Christensen TD, Noble S, Overvad TF. Time Toxicity of Systemic Anticancer Therapy for Metastatic Lung Cancer in Routine Clinical Practice: A Nationwide Cohort Study. JCO Oncol Pract 2024:OP2400526. [PMID: 39705615 DOI: 10.1200/op-24-00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/26/2024] [Accepted: 11/05/2024] [Indexed: 12/22/2024] Open
Abstract
PURPOSE The concept of time toxicity of cancer treatment, defined as proportion of days from physical contact with the health care system, has been suggested as simple, patient-centered measure useful for shared decision making, particularly in incurable cancer. We investigated the extent of health care contacts in clinical practice in Danish patients with stage IV lung cancer starting first-line systemic anticancer therapies. METHODS This is a nationwide cohort study of newly diagnosed patients with stage IV lung cancer in Denmark who initiated treatment during 2019-2021 and followed for up to 1 year. The time toxicity after treatment initiation was calculated as the proportion and mean cumulative number of days with physical health care system contacts recorded in Danish registries. The remaining days without any physical contact were defined as home days. One-year cumulative mortality was also assessed. RESULTS We included 4,384 patients with stage IV lung cancer. One year survival was 45% after treatment initiation. Of days alive, the mean cumulative number of days with physical health care contacts was 56 days within 1 year. The corresponding number of home days was 198. Overall, 22% of days alive involved physical contact with the health care system, broadly similar for patients with non-small cell lung cancer (22%) and small cell lung cancer (24%). For specific regimens, the corresponding proportions were chemotherapy (24%), immunotherapy (21%), immunochemotherapy (21%), and targeted therapy (16%). CONCLUSION More than 1 in 5 days after initiation of systemic treatment for metastatic lung cancer was spent in physical contact with the health care system. This information may aid shared decision making by informing about expected burdens in relation to cancer therapy.
Collapse
Affiliation(s)
- Anne Gulbech Ording
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Flemming Skjøth
- Department of Data, Innovation, and Research, Lillebælt Hospital, Vejle, Denmark
| | - Laurids Østergaard Poulsen
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Weronika Maria Szejniuk
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Erik Jakobsen
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, Odense, Denmark
| | - Thomas Decker Christensen
- Department of Cardiothoracic and Vascular Surgery and Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Noble
- Marie Curie Palliative Care Research Centre, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
14
|
Ul Haq MZ, Heredia C, Buadu A, Rizvi A, Workentin A, Persaud N. Changes in essential cancer medicines and association with cancer outcomes: an observational study of 158 countries. BMC Cancer 2024; 24:1526. [PMID: 39696026 DOI: 10.1186/s12885-024-13247-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Cancer is a major cause of mortality worldwide, and differences in cancer mortality rates between countries are, in part, due to differences in access to cancer care, including medicines. National essential medicines lists (NEMLs) play a role in prioritization of healthcare expenditure and access to medicines. We examined the association between amenable cancer mortality and listing medicines used in the management of eight cancers (non-melanoma skin, uterine, breast, Hodgkin lymphoma, colon, leukemia, cervical, and testicular) in national essential medicines lists of 158 countries and summarized changes to the inclusion of cancer treatments in NEMLs. METHODS We conducted a cross-sectional examination of NEMLs for 158 countries, which were obtained in May 2023. We identified medicines used to treat each of the eight cancers and determined the number of medicines listed by NEMLs for each cancer. We conducted multiple linear regressions to examine the association between the number of medicines listed on the NEMLs and cancer mortality. RESULTS We found associations between cancer medicine listing and outcomes for six of the eight examined cancers (non-melanoma skin cancer (p = 0.001), uterine cancer (p = 0.006), breast cancer (p = 0.001), Hodgkin lymphoma (p = 0.021), colon cancer (p = 0.006), and leukemia (p = 0.002)), when adjusting for healthcare expenditure and population size. CONCLUSION There was an association between listing cancer medicines on NEMLs and cancer mortality. Further research is required to explore how cancer mortality may be impacted by other cancer interventions, as well as policies to improve equitable access to cancer care.
Collapse
Affiliation(s)
- Moizza Zia Ul Haq
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, 209 Victoria St, Toronto, ON, M5B 1T8, Canada
| | - Camila Heredia
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, 209 Victoria St, Toronto, ON, M5B 1T8, Canada
| | - Adelaide Buadu
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, 209 Victoria St, Toronto, ON, M5B 1T8, Canada
| | - Amal Rizvi
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, 209 Victoria St, Toronto, ON, M5B 1T8, Canada
| | - Aine Workentin
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, 209 Victoria St, Toronto, ON, M5B 1T8, Canada
| | - Nav Persaud
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, 209 Victoria St, Toronto, ON, M5B 1T8, Canada.
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
15
|
Verduzco-Aguirre H, Wilson BE. Balancing clinical benefit and social value: challenges in HTA assessments. Lancet Oncol 2024; 25:1518-1519. [PMID: 39637884 DOI: 10.1016/s1470-2045(24)00557-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Haydee Verduzco-Aguirre
- Department of Oncology, Queen's University, Kingston, ON K7L 5P9, Canada; Division of Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada; Department of Hematology and Oncology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Brooke E Wilson
- Department of Oncology, Queen's University, Kingston, ON K7L 5P9, Canada; Division of Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
16
|
Locher L, Serra-Burriel M, Trapani D, Nussli E, Vokinger KN. Why effect sizes are systematically larger for progression-free survival than overall survival in cancer drug trials: Prognostic scores as a way forward. Eur J Cancer 2024; 213:115106. [PMID: 39550905 DOI: 10.1016/j.ejca.2024.115106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/19/2024]
Abstract
Cancer drugs have accumulated the most approvals over the past years. Overall survival (OS) is considered the gold standard for cancer trial outcomes. However, its use has declined over the past years, in favor of surrogate endpoints, such as progression-free survival (PFS). PFS allows to assess outcomes earlier and, thus, accelerates approval of cancer drugs. Previous studies have demonstrated a poor correlation between PFS and OS. Using simulation models, we examined why PFS usually overestimates survival benefit. We created a publicly accessible web application that allows users to run the simulations with different parameter settings. Based on the findings, we propose that assessment of preliminary evidence should be based on a combination of OS result and prognostic scores that reflect the health status of surviving patients.
Collapse
Affiliation(s)
- Luca Locher
- Academic Chair for Regulation in Law, Medicine and Technology, Faculty of Law, University of Zurich, Zurich, Switzerland; Academic Chair for Regulation in Law, Medicine and Technology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Miquel Serra-Burriel
- Academic Chair for Regulation in Law, Medicine and Technology, Faculty of Law, University of Zurich, Zurich, Switzerland; Academic Chair for Regulation in Law, Medicine and Technology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland; Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy; European Institute of Oncology, IRCCS, Milan, Italy; Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara 28100, Italy
| | - Emanuel Nussli
- Academic Chair for Regulation in Law, Medicine and Technology, Faculty of Law, University of Zurich, Zurich, Switzerland
| | - Kerstin N Vokinger
- Academic Chair for Regulation in Law, Medicine and Technology, Faculty of Law, University of Zurich, Zurich, Switzerland; Academic Chair for Regulation in Law, Medicine and Technology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
17
|
Gupta A, Brundage MD, Galica J, Karim S, Koven R, Ng TL, O'Donnell J, tenHove J, Robinson A, Booth CM. Patients' considerations of time toxicity when assessing cancer treatments with marginal benefit. Oncologist 2024; 29:978-985. [PMID: 39045654 PMCID: PMC11546709 DOI: 10.1093/oncolo/oyae187] [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: 04/13/2024] [Accepted: 06/27/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Effective techniques for eliciting patients' preferences regarding their own care, when treatment options offer marginal gains and different risks, is an important clinical need. We sought to evaluate the association between patients' considerations of the time burdens of care ("time toxicity") with decisions about hypothetical treatment options. METHODS We conducted a secondary analysis of a multicenter, mixed-methods study that evaluated patients' attitudes and preferences toward palliative-intent cancer treatments that delayed imaging progression-free survival (PFS) but did not improve overall survival (OS). We classified participants based on if they spontaneously volunteered one or more consideration of time burdens during qualitative interviews after treatment trade-off exercises. We compared the percentage of participants who opted for treatments with no PFS gain, some PFS gain, or who declined treatment regardless of PFS gain (in the absence of OS benefit). We conducted narrative analysis of themes related to time burdens. RESULTS The study cohort included 100 participants with advanced cancer (55% women, 63% age > 60 years, 38% with gastrointestinal cancer, and 80% currently receiving cancer-directed treatment. Forty-six percent (46/100) spontaneously described time burdens as a factor they considered in making treatment decisions. Participants who mentioned time (vs not) had higher thresholds for PFS gains required for choosing additional treatments (P value .004). Participants who mentioned time were more likely to decline treatments with no OS benefit irrespective of the magnitude of PFS benefit (65%, vs 31%). On qualitative analysis, we found that time burdens are influenced by several treatment-related factors and have broad-ranging impact, and illustrate how patients' experiences with time burdens and their preferences regarding time influence their decisions. CONCLUSIONS Almost half of participating patients spontaneously raised the issue of time burdens of cancer care when making hypothetical treatment decisions. These patients had notable differences in treatment preferences compared to those who did not mention considerations of time. Decision science researchers and clinicians should consider time burdens as an important attribute in research and in clinic.
Collapse
Affiliation(s)
- Arjun Gupta
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Michael D Brundage
- Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON K7L3N6, Canada
| | - Jacqueline Galica
- Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON K7L3N6, Canada
| | - Safiya Karim
- Tom Baker Cancer Centre, Calgary, AB T2N4N2, Canada
| | - Rachel Koven
- Patient Advocate on behalf of Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON K7L3N6, Canada
| | - Terry L Ng
- Division of Medical Oncology, University of Ottawa, Ottawa ON K1H8L6, Canada
| | - Jennifer O'Donnell
- Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON K7L3N6, Canada
| | - Julia tenHove
- Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON K7L3N6, Canada
| | - Andrew Robinson
- Cancer Centre of Southeastern Ontario, Kingston General Hospital, Kingston, ON K7L2V7, Canada
| | - Christopher M Booth
- Cancer Care and Epidemiology, Sinclair Cancer Research Institute, Queen's University, Kingston, ON K7L3N6, Canada
| |
Collapse
|
18
|
Lu D, Chen X, Mu Y, Kong L, Zhang L, Li J. Discontinuation and non-publication of randomized controlled trials on cervical cancer or precancer. Jpn J Clin Oncol 2024; 54:1141-1149. [PMID: 39041316 DOI: 10.1093/jjco/hyae096] [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: 05/08/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Research waste is a considerable problem in clinical trials, with nonpublication being a significant contributor. We aimed to determine the prevalence of discontinuation and nonpublication of randomized controlled trials (RCTs) on cervical cancer or precancer. METHODS We searched ClinicalTrials.gov for registered RCTs investigating cervical cancer or precancer that started between January 2000 and December 2020. The primary and secondary outcomes were trial nonpublication and premature discontinuation, respectively. Publication status was determined by systematic searches of peer-reviewed journals using the PubMed and Scopus databases. RESULTS A total of 113 RCTs met the inclusion criteria. Among the 85 trials completed before December 2020, 44 (51.8%) were prematurely discontinued and 40 (47.1%) were unpublished. A single-center design (61.4% vs. 34.1%, P = .012) and lack of external funding (59.1% vs. 36.6%, P = .038) were significantly associated with trial discontinuation. Large-scale (target sample size >400; 46.7% vs. 17.5%, P = .004) and externally funded trials (66.7% vs. 35.0%, P = .004) were more likely to be published. Multivariate logistic analysis revealed that a large sample size [odd ratio (OR): 4.125, 95% confidence interval (CI): 1.511-11.259, P = .006] and presence of external funding (OR: 3.714, 95% CI: 1.513-9.117, P = .004) were independent positive factors for trial publication. CONCLUSION A significant proportion of RCTs related to cervical cancer or precancer were discontinued early or remain unpublished, resulting in a waste of research resources.
Collapse
Affiliation(s)
- Dongfang Lu
- Department of Gynecology, Xingtai Central Hospital, Xingtai 054000, China
| | - Xiaolin Chen
- Department of Gynecology, Xingtai Central Hospital, Xingtai 054000, China
| | - Yanmin Mu
- Department of Gynecology, Xingtai Central Hospital, Xingtai 054000, China
| | - Lingxiao Kong
- Department of Gynecology, Xingtai Central Hospital, Xingtai 054000, China
| | - Ling Zhang
- Department of Gynecology, Xingtai Central Hospital, Xingtai 054000, China
| | - Juan Li
- Department of Gynecology, Xingtai Central Hospital, Xingtai 054000, China
| |
Collapse
|
19
|
Gale RP, Barosi G. The Gordian knot: ruxolitinib or transplants for high-risk myelofibrosis. Haematologica 2024; 109:3469-3470. [PMID: 38988273 PMCID: PMC11532677 DOI: 10.3324/haematol.2024.285972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial College London, London.
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis. IRCCS Policlinico S. Matteo Foundation, Pavia
| |
Collapse
|
20
|
Martínez-Barros H, Pousada-Fonseca Á, Pedreira-Bouzas J, Clopés-Estela A. [Translated article] Characteristics, clinical benefit, and reimbursement of new authorisations for oncohaematology drugs in Spain between 2017 and 2020. FARMACIA HOSPITALARIA 2024; 48:T272-T277. [PMID: 39069449 DOI: 10.1016/j.farma.2024.07.006] [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: 12/13/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE To describe the authorisations and funding resolutions for new onco-haematological drugs in Spain between 2017 and 2020, as well as the results of their main trials. METHODS Observational, cross-sectional, descriptive study conducted between October and December 2022. Onco-haematology drugs approved by the European Medicines Agency between 2017 and 2020 were included, according to EFPIA patients W.A.I.T Indicator 2021 Survey. Authorisation information was obtained from the main study of the European Public Assessment Report. Data were collected on medicines, their authorisation and main study, benefit shown, cost, and status and time to reimbursement. RESULTS Forty-one new drugs authorised for 49 indications were identified. More than half (58.5%) were targeted therapies, and 61.2% were for the treatment of solid tumours (61.2%). Most had palliative intent (71.4%) and were indicated in relapsed or refractory disease (55.1%). Of the clinical trials, 57.1% were phase III and 63.3% were randomised. The primary endpoint was overall survival in 16.3%, increasing to 25.8% among randomised clinical trials. Regarding licensed drugs based on response rate, the median response rate was 56.4% [IQI 40-66.3]. In those authorised on the basis of surrogate time-to-event endpoints, the median hazard ratio was 0.54 [IQI 0.38-0.57], and among those using overall survival was 0.71 [IQI 0.59-0.77]. Globally, 22.4% had shown benefit in overall survival, with a median gain of 4 months [IQI 3.6-16.7]. One-third (33.3%) of the indications evaluable according to the European Society for Medical Oncology Magnitude of Clinical Benefit Scale showed substantial clinical benefit. Of the indications, 75.5% were funded, half (48.6%; 36.7% of the total) with restrictions. The median time to funding was 19.5 months [IQI 11.4-29.3]. CONCLUSIONS Most main clinical trials of new onco-haematology drugs approved in Spain used surrogate primary endpoint and, at the time of authorisation, few had shown to prolong overall survival. More than a third were uncontrolled clinical trials.
Collapse
Affiliation(s)
| | | | | | - Ana Clopés-Estela
- Servicio de Farmacia, Instituto Catalán de Oncología (ICO) t, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Universidad Ramon Llull, Barcelona, Spain
| |
Collapse
|
21
|
Sanford NN, Hall WA. Accrual to Radiotherapy Trials in the US-Pitfalls and Potential Solutions. JAMA Oncol 2024; 10:1493-1494. [PMID: 39264636 DOI: 10.1001/jamaoncol.2024.3663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
This Viewpoint discusses accrual difficulties of radiotherapy cooperative group trials and proposes solutions.
Collapse
Affiliation(s)
- Nina N Sanford
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas
| | - William A Hall
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
22
|
Martínez-Barros H, Pousada-Fonseca Á, Pedreira-Bouzas J, Clopés-Estela A. Characteristics, clinical benefit and reimbursement of new authorisations for oncohaematology drugs in Spain between 2017 and 2020. FARMACIA HOSPITALARIA 2024; 48:272-277. [PMID: 38797624 DOI: 10.1016/j.farma.2024.04.022] [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: 12/13/2023] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVE To describe the authorisations and funding resolutions for new onco-hematological drugs in Spain between 2017 and 2020, as well as the results of their main trials. METHODS Observational, cross-sectional, descriptive study conducted between October and December 2022. Onco-hematology drugs approved by the European Medicines Agency between 2017 and 2020 were included, according to EFPIA patients W.A.I.T Indicator 2021 Survey. Authorisation information was obtained from the main study of the European Public Assessment Report (EPAR). Data were collected on medicines, their authorisation and main study, benefit shown, cost, and status and time to reimbursement. RESULTS Forty-one new drugs authorised for 49 indications were identified. More than half (58.5%) were targeted therapies, and 61.2% were for the treatment of solid tumors (61.2%). Most had palliative intent (71.4%) and were indicated in relapsed or refractory disease (55.1%). Of the clinical trials, 57.1% were phase III and 63.3% were randomised. The primary endpoint was overall survival in 16.3%, increasing to 25.8% among randomised clinical trials. Regarding licensed drugs based on response rate, the median response rate was 56.4% (IQI 40.0-66.3). In those authorised on the basis of surrogate time-to-event endpoints, the median Hazard Ratio was 0.54 (IQI 0.38-0.57), and among those using overall survival was 0.71 (IQI 0.59-0.77). Globally, 22.4% had shown benefit in overall survival, with a median gain of 4 months (IQI 3.6-16.7). One third (33.3%) of the indications evaluable according to the European Society for Medical Oncology Magnitude of Clinical Benefit Scale showed substantial clinical benefit. Of the indications, 75.5% were funded, half (48.6%; 36.7% of the total) with restrictions. The median time to funding was 19.5 months (IQI 11.4-29.3). CONCLUSIONS Most main clinical trials of new onco-haematology drugs approved in Spain used surrogate primary endpoint and, at the time of authorisation, few had shown to prolong overall survival. More than a third were uncontrolled clinical trials.
Collapse
Affiliation(s)
| | | | | | - Ana Clopés-Estela
- Servicio de Farmacia, Instituto Catalán de Oncología (ICO), Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Universidad Ramon Llull, Barcelona, España
| |
Collapse
|
23
|
Fakih M, Prager GW, Tabernero J, Amellal N, Calleja E, Taieb J. Clinically meaningful outcomes in refractory metastatic colorectal cancer: a decade of defining and raising the bar. ESMO Open 2024; 9:103931. [PMID: 39395264 PMCID: PMC11693422 DOI: 10.1016/j.esmoop.2024.103931] [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: 05/23/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 10/14/2024] Open
Abstract
Currently, there is no consensus definition for clinically meaningful outcomes in randomized clinical trials (RCTs) designed to evaluate new treatments for patients with refractory metastatic colorectal cancer (mCRC). Since 2014, recommended targets for improvements in overall survival and progression-free survival have been published by several societies, including those from the American Society of Clinical Oncology (ASCO) Clinically Meaningful Outcomes Working Group in 2014, the European Society for Medical Oncology-Magnitude of Clinical Benefit Scale (ESMO-MCBS) in 2015, and Colorectal Cancer Canada (CCC) consensus statements in 2019. However, evidence from several systematic reviews suggests that in a substantial proportion of RCTs that led to oncology drug approvals, the recommended thresholds of ASCO and ESMO-MCBS were not met. In addition to efficacy and safety, quality of life (QoL) is important to patients with mCRC, especially for those who are receiving later-line therapy or end-of-life care. As such, both ESMO-MCBS and CCC recommend the inclusion of QoL assessments in the design of mCRC clinical trials. Since the publication of the ASCO recommendations in 2014, there has been significant progress in the development of treatment options for patients with refractory mCRC; these include the approvals of trifluridine/tipiracil (FTD/TPI) as a single agent and in combination with bevacizumab, and the approval of fruquintinib. Among the phase III RCTs in third-line mCRC, only the SUNLIGHT trial of FTD/TPI plus bevacizumab met all recommended thresholds for clinically meaningful improvements, while also demonstrating a manageable safety profile and slower deterioration in multiple measures of QoL compared with FTD/TPI alone. The results from the SUNLIGHT study show that incremental gains in several clinically meaningful endpoints are achievable, thus raising the bar in defining clinically meaningful outcomes for emerging therapies in refractory mCRC.
Collapse
Affiliation(s)
- M Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, USA.
| | - G W Prager
- Department of Medicine I, Medical University Vienna, Vienna, Austria
| | - J Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, Barcelona, Spain
| | - N Amellal
- Servier International Research Institute, Suresnes, France
| | - E Calleja
- Taiho Oncology, Inc., Princeton, USA
| | - J Taieb
- Gastroenterology and Gastrointestinal Oncology Department, Hôpital Européen Georges-Pompidou, University Paris-Cité (Paris Descartes), SIRC CARPEM, Paris, France
| |
Collapse
|
24
|
Banerjee S, Booth CM, Bruera E, Büchler MW, Drilon A, Fry TJ, Ghobrial IM, Gianni L, Jain RK, Kroemer G, Llovet JM, Long GV, Pantel K, Pritchard-Jones K, Scher HI, Tabernero J, Weichselbaum RR, Weller M, Wu YL. Two decades of advances in clinical oncology - lessons learned and future directions. Nat Rev Clin Oncol 2024; 21:771-780. [PMID: 39354161 DOI: 10.1038/s41571-024-00945-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/03/2024]
Affiliation(s)
- Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK.
- The Institute of Cancer Research, London, UK.
| | | | - Eduardo Bruera
- Department of Palliative, Rehabilitation, and Integrative Medicine, The University of Texas MD Anderson Cancer, Unit 1414, Houston, TX, USA.
| | - Markus W Büchler
- Botton-Champalimaud Pancreatic Cancer, Champalimaud Foundation, Lisbon, Portugal.
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA.
| | - Terry J Fry
- Department of Paediatrics and Immunology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Aurora, CO, USA.
| | - Irene M Ghobrial
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medical Oncology, Harvard Medical School, Boston, MA, USA.
| | | | - Rakesh K Jain
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Josep M Llovet
- Mount Sinai Liver Cancer Program, Divisions of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia.
| | - Klaus Pantel
- Institute of Tumour Biology, University Cancer Center Hamburg, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital (HUVH), Barcelona, Spain.
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA.
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
25
|
Olivier T, Haslam A, Ochoa D, Fernandez E, Prasad V. Bedside implications of the use of surrogate endpoints in solid and haematological cancers: implications for our reliance on PFS, DFS, ORR, MRD and more. BMJ ONCOLOGY 2024; 3:e000364. [PMID: 39886154 PMCID: PMC11557723 DOI: 10.1136/bmjonc-2024-000364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 09/12/2024] [Indexed: 02/01/2025]
Abstract
Clinical endpoints, such as overall survival, directly measure relevant outcomes. Surrogate endpoints, in contrast, are intermediate, stand-in measures of various tumour-related metrics and include tumour growth, tumour shrinkage, blood results, etc. Surrogates may be a time point measurement, that is, tumour shrinkage at some point (eg, response rate) or biomarker-assessed disease status, measured at given time points (eg, circulating tumour DNA, ctDNA). They can also be measured over time, as with progression-free survival, which is the time until a patient presents with either disease progression or death. Surrogates are increasingly used in trials supporting the marketing authorisation of novel oncology drugs. Yet, the trial-level correlation between surrogates and clinical endpoints-meaning to which extent an improvement in the surrogate predicts an improvement in the direct endpoint-is often moderate to low. Here, we provide a comprehensive classification of surrogate endpoints: time point measurements and time-to-event endpoints in solid and haematological malignancies. Also, we discuss an overlooked aspect of the use of surrogates: the limitations of surrogates outside trial settings, at the bedside. Surrogates can result in the inappropriate stopping or switching of therapy. Surrogates can be used to usher in new strategies (eg, ctDNA in adjuvant treatment of colon cancer), which may erode patient outcomes. In liquid malignancies, surrogates can mislead us to use novel drugs and replace proven standards of care with costly medications. Surrogates can lead one to intensify treatment without clear improvement and possibly worsening quality of life. Clinicians should be aware of the role of surrogates in the development and regulation of drugs and how their use can carry real-world, bedside implications.
Collapse
Affiliation(s)
- Timothée Olivier
- Department of Oncology, Geneva University Hospitals, Geneve, Switzerland
| | - Alyson Haslam
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Dagney Ochoa
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Eduardo Fernandez
- Jane Anne Nohl Division of Hematology and Center for the Study of Blood Diseases, USC Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
26
|
Tregear M, Visco F. Outcomes that matter to patients with cancer: living longer and living better. EClinicalMedicine 2024; 76:102833. [PMID: 39309725 PMCID: PMC11415949 DOI: 10.1016/j.eclinm.2024.102833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Oncologists and cancer patients generally agree that the primary goals of advanced cancer treatment are to lengthen and/or improve patient survival. Yet over the last two decades, clinical trials of new cancer treatments have moved away from measuring outcomes that matter to patients. Increasingly, new drugs for advanced cancer treatment reach the market by demonstrating improvements in surrogate endpoints such as progression-free survival (PFS), which is not a measure of how a patient feels, functions, or survives. Research has shown that when patients are fully informed about the meaning of PFS, about half would not choose additional treatment for any magnitude of gain in PFS in the absence of an overall survival improvement. It's time to get back to designing trials that answer clinically meaningful questions and measure the outcomes that truly matter to patients. Engaging educated patient advocates in meaningful ways in clinical trial design and reporting would be a step in this direction.
Collapse
Affiliation(s)
| | - Fran Visco
- National Breast Cancer Coalition, Washington, DC, USA
| |
Collapse
|
27
|
Pokorny AMJ. (Un)common sense in oncology. Drug Ther Bull 2024; 62:146. [PMID: 39242152 DOI: 10.1136/dtb.2024.000043] [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: 09/09/2024]
Affiliation(s)
- Adrian M J Pokorny
- Alice Springs Hospital, Alice Springs, Northern Territory, Australia
- Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| |
Collapse
|
28
|
Somers AMJ, Duits AJ, Samson MJ, Schnog JJB. Pharmaceutical company funding of cancer patient advocacy organizations in the Netherlands. J Cancer Policy 2024; 41:100493. [PMID: 38876202 DOI: 10.1016/j.jcpo.2024.100493] [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: 01/20/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Financial conflicts of interest (FCOI) of medical professionals and associated organizations with pharmaceutical companies (pharma) might contribute to the use of low value oncological treatments. Value criteria for oncological drug approvals in the Netherlands have recently become more stringent leading to objections by cancer patient advocacy organizations (cPAOs). Considering the importance of cPAOs input in cancer patient care we analyzed whether pharma funding of cPAOs occurs in the Netherlands. METHODS The cPAO websites and available annual reports were evaluated for disclosure of pharma funding for the years 2021 and 2022. Also, data from the Dutch Healthcare Transparency Registry (DHTR) were extracted. RESULTS Twenty-one of 34 (61.8 %) cPAOs received pharma funding (with 20 registered in the DHTR), and for 13 (29.4 %) cPAOs no reporting of pharma funding could be found. Three of the cPAOs disclosed pharma funding directly on their main website. Online educational material was available from 22 cPAOs on their websites with pharma funding disclosed on the educational material in 5. The total registered amount of pharmaceutical funding was €667,232.00 in 2021 and €536,098.00 in 2022. The median (and interquartile ranges) DHTR registered amount of support per cPAO that received funding in the studied period was €23,799.50 (14,823.75-84,663.30). The most common funding category as defined in the DHTR was project sponsorship. CONCLUSIONS Financial support by the pharmaceutical industry is common for Dutch cPAOs. Given the importance of cPAOs and their objective input in the societal debate on the availability of cancer drugs, the potential influence of pharma sponsoring should be critically evaluated.
Collapse
Affiliation(s)
- Anne M J Somers
- Department of Hematology-Medical Oncology, Curaçao Medical Center, J.H.J. Hamelbergweg, Willemstad, Curaçao
| | - Ashley J Duits
- Curaçao Biomedical & Health Research Institute, Pater Eeuwensweg 36, Willemstad, Curaçao; Department of Medical Education, Curaçao Medical Center, J.H.J. Hamelbergweg, Willemstad, Curaçao; Institute for Medical Education, University Medical Center Groningen, Hanzeplein 1, the Netherlands; Red Cross Blood Bank Foundation, Pater Eeuwensweg 36, Willemstad, Curaçao
| | - Michael J Samson
- Department of Radiation Oncology, Curaçao Medical Center, J.H.J. Hamelbergweg, Willemstad, Curaçao.
| | - John-John B Schnog
- Department of Hematology-Medical Oncology, Curaçao Medical Center, J.H.J. Hamelbergweg, Willemstad, Curaçao; Curaçao Biomedical & Health Research Institute, Pater Eeuwensweg 36, Willemstad, Curaçao.
| |
Collapse
|
29
|
Tibau A, Hwang TJ, Avorn J, Kesselheim AS. Clinical value of guideline recommended molecular targets and genome targeted cancer therapies: cross sectional study. BMJ 2024; 386:e079126. [PMID: 39164034 PMCID: PMC11333991 DOI: 10.1136/bmj-2023-079126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/22/2024]
Abstract
OBJECTIVE To assess the clinical benefit and actionability of molecular targets for genome targeted cancer drugs recommended for clinical practice by the National Comprehensive Cancer Network (NCCN). DESIGN Cross sectional study. PARTICIPANTS/SETTING Genome targeted cancer drugs recommended by NCCN guidelines in the advanced setting. MAIN OUTCOME MEASURES Molecular target actionability was assessed using the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT). Clinical benefit of genome targeted oncology therapies was evaluated using the ESMO-Magnitude of Clinical Benefit Scale (ESMO-MCBS). Molecular targets at ESCAT category level I associated with studies showing substantial clinical benefit by ESMO-MCBS (grades 4-5) were designated as high benefit, and those linked to studies achieving an ESMO-MCBS grade of 3 were categorized as being of promising but unproven benefit. RESULTS 411 recommendations related to 74 genome targeted drugs targeting 50 driver alterations were examined. Most recommendations (346/411; 84%) were associated with clinical trials of various phases, but 16% (65/411) relied on only case reports or pre-clinical studies. However, clinical trials mostly comprised phase I or phase II (271/346; 78%), single arm (262/346; 76%) studies. The primary endpoint assessed in most trials was overall response rate (271/346; 78%) rather than survival. ESCAT tier I targetability encompassed 60% (246/411) of target recommendations, 35% (142/411) were classified as tier II or III, and 6% (23/411) had their relevance yet to be determined (tiers IV to X). When ESMO-MCBS was applied to 267 scorable trials, only 12% (32/267) showed substantial clinical benefit (grades 4-5) and 45% (121/267) were grade 3. When both frameworks were combined, 12% (32/267) of trials supported a determination of high benefit and 33% (88/267) indicated promising but unproven benefit. Of the 118 interventions endorsed by NCCN authors as preferred, 62 (53%) applied to treatments with high or promising but unproven benefit. CONCLUSION According to the ESCAT and ESMO-MCBS frameworks, about one eighth of genome based treatments for solid cancer were rated as likely to offer a high benefit to patients, whereas around a third were identified as offering a promising but unproven substantial benefit. Ensuring that NCCN recommendations are aligned with expected clinical benefits is crucial for promoting informed, evidence based, genomic guided treatment decisions.
Collapse
Affiliation(s)
- Ariadna Tibau
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Oncology Department, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau, and Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Thomas J Hwang
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Cancer Innovation and Regulation Initiative, Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Urological Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA USA
| | - Jerry Avorn
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Aaron S Kesselheim
- Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Aggarwal A, Simcock R, Price P, Rachet B, Lyratzopoulos G, Walker K, Spencer K, Roques T, Sullivan R. NHS cancer services and systems-ten pressure points a UK cancer control plan needs to address. Lancet Oncol 2024; 25:e363-e373. [PMID: 38991599 DOI: 10.1016/s1470-2045(24)00345-0] [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: 05/30/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024]
Abstract
In this Policy Review we discuss ten key pressure points in the NHS in the delivery of cancer care services that need to be urgently addressed by a comprehensive national cancer control plan. These pressure points cover areas such as increasing workforce capacity and its productivity, delivering effective cancer survivorship services, addressing variation in quality, fixing the reimbursement system for cancer care, and balancing of the cancer research agenda. These areas have been selected based on their relative importance to ensuring sustainable cancer services, persistence as key issues in the NHS, and their impact on delivering better and more equitable and affordable patient outcomes. Many of these pressure points are not acknowledged explicitly in any current discourse. The evidence we provide points to their impact on the ability to deliver world class cancer care, but also to their amenability to affordable solutions if given the relevant prioritisation and investment. The current narrative needs to move away from a technocentric approach to improving care, to one focused on understanding the complexity of cancer services and the wider health system to drive improvements in survival, quality of life, and experience for patients.
Collapse
Affiliation(s)
- Ajay Aggarwal
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK; Department of Oncology, Guy's & St Thomas' NHS Trust, London, UK.
| | - Richard Simcock
- Department of Oncology, University Hospitals Sussex NHS Trust, Brighton, UK
| | - Pat Price
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Bernard Rachet
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Kate Walker
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Katie Spencer
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK; Department of Oncology, Leeds Teaching Hospitals NHS Trust, Leeds
| | - Tom Roques
- Department of Oncology, Norfolk and Norwich NHS Foundation Trust, Norwich, UK
| | | |
Collapse
|
31
|
Baldwin D, Carmichael J, Cook G, Navani N, Peach J, Slater R, Wheatstone P, Wilkins J, Allen-Delingpole N, Kerr CEP, Siddiqui K. UK Stakeholder Perspectives on Surrogate Endpoints in Cancer, and the Potential for UK Real-World Datasets to Validate Their Use in Decision-Making. Cancer Manag Res 2024; 16:791-810. [PMID: 39044745 PMCID: PMC11264281 DOI: 10.2147/cmar.s441359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Duration of overall survival in patients with cancer has lengthened due to earlier detection and improved treatments. However, these improvements have created challenges in assessing the impact of newer treatments, particularly those used early in the treatment pathway. As overall survival remains most decision-makers' preferred primary endpoint, therapeutic innovations may take a long time to be introduced into clinical practice. Moreover, it is difficult to extrapolate findings to heterogeneous populations and address the concerns of patients wishing to evaluate everyday quality and extension of life. There is growing interest in the use of surrogate or interim endpoints to demonstrate robust treatment effects sooner than is possible with measurement of overall survival. It is hoped that they could speed up patients' access to new drugs, combinations, and sequences, and inform treatment decision-making. However, while surrogate endpoints have been used by regulators for drug approvals, this has occurred on a case-by-case basis. Evidence standards are yet to be clearly defined for acceptability in health technology appraisals or to shape clinical practice. This article considers the relevance of the use of surrogate endpoints in cancer in the UK context, and explores whether collection and analysis of real-world UK data and evidence might contribute to validation.
Collapse
Affiliation(s)
- David Baldwin
- Department of Respiratory Medicine, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Jonathan Carmichael
- Department of Oncology, The National Institute for Health Research Leeds In Vitro Diagnostics Co-Operative (NIHR Leeds MIC), Leeds, UK
| | - Gordon Cook
- Cancer Research UK Trials Unit, LICTR, University of Leeds & NIHR (Leeds) IVD MIC, Leeds, UK
| | - Neal Navani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
- Department of Thoracic Medicine, University College London Hospital, London, UK
| | - James Peach
- Human Centric Drug Discovery, Wood Centre for Innovation, Oxford, UK
| | | | - Pete Wheatstone
- Patient and Public Involvement and Engagement Group, DATA-CAN, London, UK
| | | | | | | | | |
Collapse
|
32
|
Bommier C, Maurer MJ, Lambert J. What clinicians should know about surrogate end points in hematologic malignancies. Blood 2024; 144:11-20. [PMID: 38603637 DOI: 10.1182/blood.2023022269] [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: 10/10/2023] [Revised: 03/14/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
ABSTRACT Use of surrogates as primary end points is commonplace in hematology/oncology clinical trials. As opposed to prognostic markers, surrogates are end points that can be measured early and yet can still capture the full effect of treatment, because it would be captured by the true outcome (eg, overall survival). We discuss the level of evidence of the most commonly used end points in hematology and share recommendations on how to apply and evaluate surrogate end points in research and clinical practice. Based on the statistical literature, this clinician-friendly review intends to build a bridge between clinicians and surrogacy specialists.
Collapse
Affiliation(s)
- Côme Bommier
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Epidemiology and Clinical Statistics for Tumor, Respiratory, and Resuscitation Assessments Team, INSERM, U1153, Assistance Publique-Hôpitaux de Paris Hôpital St Louis, Université Paris Cité, Paris, France
| | - Matthew John Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Jerome Lambert
- Epidemiology and Clinical Statistics for Tumor, Respiratory, and Resuscitation Assessments Team, INSERM, U1153, Assistance Publique-Hôpitaux de Paris Hôpital St Louis, Université Paris Cité, Paris, France
| |
Collapse
|
33
|
Wilson BE, Eisenhauer EA, Booth CM. Study Participants, Future Patients, and Outcomes That Matter in Cancer Clinical Trials. JAMA 2024; 331:2081-2083. [PMID: 38767591 DOI: 10.1001/jama.2024.1281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Affiliation(s)
- Brooke E Wilson
- Department of Oncology, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
- School of Population Health, Faculty of Medicine and Health, Sydney, Australia
| | | | - Christopher M Booth
- Department of Oncology, Queen's University, Kingston, Ontario, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
| |
Collapse
|
34
|
Schnog JB, Duits AJ, Samson MJ. Design issues with lutetium-177 PSMA-617 registration studies that bias the outcome of the experimental arm reflect an increasing misalignment of contemporary oncology trials with true patient benefit. BJC REPORTS 2024; 2:45. [PMID: 39516569 PMCID: PMC11523959 DOI: 10.1038/s44276-024-00065-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 11/16/2024]
Abstract
In the PSMAfore randomized controlled trial patients with chemotherapy naïve castrate resistant metastasized prostate cancer (CRPC) progressing after one line of a second-generation androgen receptor signaling inhibitor (ARSI) were randomized to the experimental arm of lutetium-177 PSMA-617 or the control arm of another ARSI. The trial showed an increase in the primary endpoint radiographic progression free survival in the experimental arm. Previously, the VISION trial led to the approval of lutetium-177 PSMA-617 in patients with CRPC progressing after at least 1 second generation ARSI and at least 1 line of chemotherapy with a taxane. We highlight several shortcomings in both trials concerning use of putative surrogate endpoints, control arm treatments not reflective of contemporary standards of care, informative censoring and inappropriate cross-over, that all bias results in favor of the experimental arms. Additional regulatory approval of lutetium-177 PSMA-617 for patients prior to receiving chemotherapy would not only lead to further exposure of patients to a treatment without proper proof of benefit but to unsubstantiated health care spending as well.
Collapse
Affiliation(s)
- J B Schnog
- Department of Hematology-Medical Oncology, Curaçao Medical Center, Willemstad, Curaçao.
- Curaçao Biomedical & Health Research Institute, Willemstad, Curaçao.
| | - A J Duits
- Curaçao Biomedical & Health Research Institute, Willemstad, Curaçao
- Department of Medical Education, Curaçao Medical Center, Willemstad, Curaçao
- Institute for Medical Education, University Medical Center Groningen, Groningen, the Netherlands
- Red Cross Blood Bank Foundation, Willemstad, Curaçao
| | - M J Samson
- Curaçao Biomedical & Health Research Institute, Willemstad, Curaçao
- Department of Radiation Oncology, Curaçao Medical Center, Willemstad, Curaçao
| |
Collapse
|
35
|
Stockton SS, Ayers GD, Lee C, Laferriere H, Das S, Berlin J. Evolving or immutable - phase I solid tumor trials in the era of precision oncology. Invest New Drugs 2024; 42:326-334. [PMID: 38775890 PMCID: PMC11164775 DOI: 10.1007/s10637-024-01445-z] [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: 04/01/2024] [Accepted: 05/07/2024] [Indexed: 06/04/2024]
Abstract
In the era of precision oncology (PO), systemic therapies for patients (pts) with solid tumors have shifted from chemotherapy (CT) to targeted therapy (TT) and immunotherapy (IO). This systematic survey describes features of trials enrolling between 2010 and 2020, focusing on inclusion criteria, type of dose escalation scheme (DES) utilized, and use of expansion cohorts (ECs). A literature search identified phase I studies in adults with solid tumors published January 1, 2000- December 31, 2020 from 12 journals. We included only studies enrolling between 2010 and 2020 to better capture the PO era. Two reviewers abstracted data; a third established concordance. Of 10,744 studies, 10,195 were non-topical or enrolled prior to 2010; 437 studies were included. The most common drug classes were TT (47.6%), IO (22%), and CT (6.9%). In studies which reported race, patients were predominantly white (61.7%) or Asian (25.7%), followed by black (6.5%) or other (6.1%). Heterogeneity was observed in the reporting and specification of study inclusion criteria. Only 40.1% of studies utilized ECs, and among the studies which used ECS, 46.6% were defined by genomic selection. Rule-based DES were used in 89% of trials; a 3+3 design was used in 80.5%. Of all drugs tested, 37.5% advanced to phase II, while 10.3% garnered regulatory licensure (for an indication tested in phase I). In the era of PO, TT and IO have emerged as the most studied agents in phase I trials. Rule-based DES, which are more relevant for escalating CT, are still chiefly utilized.
Collapse
Affiliation(s)
- Shannon S Stockton
- Vanderbilt University Medical Center, 1211 Medical Center Drive, 37232, Nashville, TN, USA.
| | - G Dan Ayers
- Vanderbilt University Medical Center, 1211 Medical Center Drive, 37232, Nashville, TN, USA
| | - Cody Lee
- Vanderbilt University Medical Center, 1211 Medical Center Drive, 37232, Nashville, TN, USA
| | | | | | - Jordan Berlin
- Vanderbilt University Medical Center, 1211 Medical Center Drive, 37232, Nashville, TN, USA
| |
Collapse
|
36
|
Naci H, Zhang Y, Woloshin S, Guan X, Xu Z, Wagner AK. Overall survival benefits of cancer drugs initially approved by the US Food and Drug Administration on the basis of immature survival data: a retrospective analysis. Lancet Oncol 2024; 25:760-769. [PMID: 38754451 DOI: 10.1016/s1470-2045(24)00152-9] [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: 02/24/2024] [Accepted: 03/14/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND New cancer drugs can be approved by the US Food and Drug Administration (FDA) on the basis of surrogate endpoints while data on overall survival are still incomplete or immature, with too few deaths for meaningful analysis. We aimed to evaluate whether clinical trials with immature survival data generated evidence of overall survival benefit during the period after marketing authorisation, and where that evidence was reported. METHODS In this retrospective analysis, we searched Drugs@FDA to identify cancer drug indications approved between Jan 1, 2001, and Dec 31, 2018, on the basis of immature survival data. We systematically collected publicly available data on postapproval overall survival results in labelling (Drugs@FDA), journal publications (MEDLINE via PubMed), and clinical trial registries (ClinicalTrials.gov). The primary outcome was availability of statistically significant overall survival benefits during the period after marketing authorisation (until March 31, 2023). Additionally, we evaluated the availability and timing of overall survival findings in labelling, journal publications, and ClinicalTrials.gov records. FINDINGS During the study period, the FDA granted marketing authorisation to 223 cancer drug indications, 95 of which had overall survival as an endpoint. 39 (41%) of these 95 indications had immature survival data. After a minimum of 4·3 years of follow-up during the period after marketing authorisation (and median 8·2 years [IQR 5·3-12·0] since FDA approval), additional survival data from the pivotal trials became available in either revised labelling or publications, or both, for 38 (97%) of 39 indications. Additional data on overall survival showed a statistically significant benefit in 12 (32%) of 38 indications, whereas mature data yielded statistically non-significant overall survival findings for 24 (63%) indications. Statistically significant evidence of overall survival benefit was reported in either labelling or publications a median of 1·5 years (IQR 0·8-2·3) after initial approval. The median time to availability of statistically non-significant overall survival results was 3·3 years (2·2-4·5). The availability of overall survival results on ClinicalTrials.gov varied considerably. INTERPRETATION Fewer than a third of indications approved with immature survival data showed a statistically significant overall survival benefit after approval. Notable inconsistencies in timing and availability of information after approval across different sources emphasise the need for better reporting standards. FUNDING None.
Collapse
Affiliation(s)
- Huseyin Naci
- Department of Health Policy, London School of Economics and Political Science, London, UK; The Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA.
| | - Yichen Zhang
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Steven Woloshin
- The Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA; The Center for Medicine in the Media, Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Xiaodong Guan
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ziyue Xu
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Anita K Wagner
- The Lisa Schwartz Foundation for Truth in Medicine, Norwich, VT, USA; Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| |
Collapse
|
37
|
Leng JX, Carpenter DJ, Huang C, Qazi J, Arshad M, Mullikin TC, Reitman ZJ, Kirkpatrick JP, Floyd SR, Fecci PE, Chmura SJ, Hong JC, Salama JK. Determinants of Symptomatic Intracranial Progression After an Initial Stereotactic Radiosurgery Course. Adv Radiat Oncol 2024; 9:101475. [PMID: 38690297 PMCID: PMC11059392 DOI: 10.1016/j.adro.2024.101475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/04/2024] [Indexed: 05/02/2024] Open
Abstract
Purpose Clinical and imaging surveillance of patients with brain metastases is important after stereotactic radiosurgery (SRS) because many will experience intracranial progression (ITCP) requiring multidisciplinary management. The prognostic significance of neurologic symptoms at the time of ITCP is poorly understood. Methods and Materials This was a multi-institutional, retrospective cohort study from 2015 to 2020, including all patients with brain metastases completing an initial course of SRS. The primary outcome was overall survival (OS) by presence of neurologic symptoms at ITCP. OS, freedom from ITCP (FF-ITCP), and freedom from symptomatic ITCP (FF-SITCP) were assessed via Kaplan-Meier method. Cox proportional hazard models tested parameters impacting FF-ITCP and FF-SITCP. Results Among 1383 patients, median age was 63.4 years, 55% were female, and common primaries were non-small cell lung (49%), breast (15%), and melanoma (9%). At a median follow-up of 8.72 months, asymptomatic and symptomatic ITCP were observed in 504 (36%) and 194 (14%) patients, respectively. The majority of ITCP were distant ITCP (79.5%). OS was worse with SITCP (median, 10.2 vs 17.9 months, P < .001). SITCP was associated with clinical factors including total treatment volume (P = .012), melanoma histology (P = .001), prior whole brain radiation therapy (P = .003), number of brain metastases (P < .001), interval of 1 to 2 years from primary and brain metastasis diagnosis (P = .012), controlled extracranial disease (P = .042), and receipt of pre-SRS chemotherapy (P = .015). Patients who were younger and received post-SRS chemotherapy (P = .001), immunotherapy (P < .001), and targeted or small-molecule inhibitor therapy (P < .026) had better FF-SITCP. Conclusions In this cohort study of patients with brain metastases completing SRS, neurologic symptoms at ITCP is prognostic for OS. This data informs post-SRS surveillance in clinical practice as well as future prospective studies needed in the modern management of brain metastases.
Collapse
Affiliation(s)
- Jim X. Leng
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - David J. Carpenter
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
- Department of Radiation Oncology, Wellstar Paulding Hospital, Hiram, Georgia
| | - Christina Huang
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Jamiluddin Qazi
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Muzamil Arshad
- Department of Radiation and Cellular Oncology, University of Chicago Medical Center, Chicago, Illinois
| | - Trey C. Mullikin
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Zachary J. Reitman
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - John P. Kirkpatrick
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Scott R. Floyd
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Peter E. Fecci
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Steven J. Chmura
- Department of Radiation and Cellular Oncology, University of Chicago Medical Center, Chicago, Illinois
| | - Julian C. Hong
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California
- Joint Program in Computational Precision Health, University of California, San Francisco, California and University of California, Berkeley, California
| | - Joseph K. Salama
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
- Radiation Oncology Clinical Service, Durham VA Health Care System, Durham, North Carolina
| |
Collapse
|
38
|
Wilson BE, Sengar M, Tregear M, van der Graaf WTA, Luca Battisti NM, Csaba DL, Soto-Perez-de-Celis E, Gyawali B, Booth CM. Common Sense Oncology: Equity, Value, and Outcomes That Matter. Am Soc Clin Oncol Educ Book 2024; 44:e100039. [PMID: 38788178 DOI: 10.1200/edbk_100039] [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: 05/26/2024]
Abstract
While some recent drug treatments have been transformative for patients with cancer, many treatments offer small benefits despite high clinical toxicity, time toxicity and financial toxicity. Moreover, treatments that do provide substantial clinical benefits are not available to many patients globally due to issues with availability and affordability. The Common Sense Oncology's vision is that patients will have access to treatments that provide meaningful improvements in outcomes that matter, regardless of where they live. In recognition of the growing challenges in the field of oncology, Common Sense Oncology seeks to achieve this vision by improving evidence generation, evidence interpretation and evidence communication.
Collapse
Affiliation(s)
- Brooke E Wilson
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
- School of Population Health, Faculty of Medicine, University of New South Wales, Randwick, Australia
| | - Manju Sengar
- Tata Memorial Hospital, Affiliated to Homi Bhabha National Institute, Mumbai, India
| | | | - Winette T A van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nicolò Matteo Luca Battisti
- Department of Medicine, Breast Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Inequalities Focused Topic Network, European Cancer Organisation, Brussels, Belgium
- International Society of Geriatric Oncology, Geneva, Switzerland
| | - Degi Laszlo Csaba
- Faculty of Sociology and Social Work, Babeş-Bolyai University, Cluj Napoca, Romania
| | - Enrique Soto-Perez-de-Celis
- Department of Geriatrics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Bishal Gyawali
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
| | - Christopher M Booth
- Department of Oncology, Queen's University, Kingston, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, Canada
| |
Collapse
|
39
|
Schnog JB, Samson MJ, Gersenbluth I, Duits AJ. Pharmaceutical Industry Payments to Medical Oncologists in the Netherlands: Trends and Patterns Provided by an Open-Access Transparency Data Set. JCO Oncol Pract 2024; 20:843-851. [PMID: 38354335 DOI: 10.1200/op.23.00533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/24/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
PURPOSE Health care expenditure related to oncologic treatments is skyrocketing although many treatments offer marginal, if any, clinical benefit. Financial conflicts of interest (fCOI) resulting from pharmaceutical industry (pharma) payments to physicians is increasingly recognized as a predictive factor for regulatory board approval and guideline incorporation of low-value treatments. We sought to study the extent to which pharma payments to medical oncologists occur in the Netherlands, the amount of money involved, and whether these occur more frequently and are higher for key opinion leaders (KOLs). METHODS In our cross-sectional retrospective database study, we used several Dutch open-access databases and extracted data registered between 2019 and 2021. RESULTS A cumulative amount of €899,863 was paid to 48.8% of the 408 registered medical oncologists. Over time, there was a marked decline in both the proportion of medical oncologists receiving payments (from 40.4% in 2019 to 19.1% in 2021) and the mean annual value of payments (from €2,962 in 2019 to €2,188 in 2021) with the latter mainly resulting from a decline in hospitality-related transactions. KOLs were more likely to receive industry payments and received a higher median payment value. DISCUSSION Our findings should contribute to the increasing awareness in the Netherlands of the potential effects of fCOI.
Collapse
Affiliation(s)
- J B Schnog
- Department of Hematology-Medical Oncology, Curaçao Medical Center, Willemstad, Curaçao
- Curaçao Biomedical & Health Research Institute, Willemstad, Curaçao
| | - M J Samson
- Department of Radiation Oncology, Curaçao Medical Center, Willemstad, Curaçao
| | - I Gersenbluth
- Curaçao Biomedical & Health Research Institute, Willemstad, Curaçao
| | - A J Duits
- Curaçao Biomedical & Health Research Institute, Willemstad, Curaçao
- Department of Medical Education, Curaçao Medical Center, Willemstad, Curaçao
- Institute for Medical Education, University Medical Center Groningen, Groningen, the Netherlands
- Red Cross Blood Bank Foundation, Willemstad, Curaçao
| |
Collapse
|
40
|
Tibau A, Hwang TJ, Molto C, Avorn J, Kesselheim AS. Clinical Value of Molecular Targets and FDA-Approved Genome-Targeted Cancer Therapies. JAMA Oncol 2024; 10:634-641. [PMID: 38573645 PMCID: PMC11099684 DOI: 10.1001/jamaoncol.2024.0194] [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: 06/15/2023] [Accepted: 10/03/2023] [Indexed: 04/05/2024]
Abstract
Importance The number of new genome-targeted cancer drugs has increased, offering the possibility of personalized therapy, often at a very high cost. Objective To assess the validity of molecular targets and therapeutic benefits of US Food and Drug Administration-approved genome-targeted cancer drugs based on the outcomes of their corresponding pivotal clinical trials. Design and Settings In this cohort study, all genome-targeted cancer drugs that were FDA-approved between January 1, 2015, and December 31, 2022, were analyzed. From FDA drug labels and trial reports, key characteristics of pivotal trials were extracted, including the outcomes assessed. Main Outcomes and Measures The strength of evidence supporting molecular targetability was assessed using the European Society for Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT). Clinical benefit for their approved indications was evaluated using the ESMO-Magnitude of Clinical Benefit Scale (ESMO-MCBS). Substantial clinical benefit was defined as a grade of A or B for curative intent and 4 or 5 for noncurative intent. Molecular targets qualifying for ESCAT category level I-A and I-B associated with substantial clinical benefit by ESMO-MCBS were rated as high-benefit genomic-based cancer treatments. Results A total of 50 molecular-targeted drugs covering 84 indications were analyzed. Forty-five indications (54%) were approved based on phase 1 or phase 2 pivotal trials, 45 (54%) were supported by single-arm pivotal trials, and 48 (57%) were approved on the basis of subgroup analyses. By each indication, 46 of 84 primary end points (55%) were overall response rate (median [IQR] overall response rate, 57% [40%-69%]; median [IQR] duration of response, 11.1 [9.2-19.8] months). Among the 84 pivotal trials supporting these 84 indications, 38 trials (45%) had I-A ESCAT targetability, and 32 (38%) had I-B targetability. Overall, 24 of 84 trials (29%) demonstrated substantial clinical benefit via ESMO-MCBS. Combining these ratings, 24 of 84 indications (29%) were associated with high-benefit genomic-based cancer treatments. Conclusions and Relevance The results of this cohort study demonstrate that among recently approved molecular-targeted cancer therapies, fewer than one-third demonstrated substantial patient benefits at approval. Benefit frameworks such as ESMO-MCBS and ESCAT can help physicians, patients, and payers identify therapies with the greatest clinical potential.
Collapse
Affiliation(s)
- Ariadna Tibau
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Oncology Department, Hospital de la Santa Creu i Sant Pau, Institut d’Investigació Biomèdica Sant Pau, and Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Thomas J. Hwang
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Cancer Innovation and Regulation Initiative, Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Urological Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Consolacion Molto
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Jerry Avorn
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Aaron S. Kesselheim
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
41
|
Briercheck E, Pyle D, Adams C, Atun R, Booth C, Dent J, Garcia-Gonzalez P, Ilbawi A, Jazieh AR, Kerr D, Knaul F, Kobayashi E, Lim C, Maza M, Milner D, Navarro MF, O'Brien M, Rodriguez-Galindo C, Sullivan R, Torode J, Vokes E, Gralow J. Unification of Efforts to Improve Global Access to Cancer Therapeutics: Report From the 2022/2023 Access to Essential Cancer Medicines Stakeholder Summit. JCO Glob Oncol 2024; 10:e2300256. [PMID: 38781548 DOI: 10.1200/go.23.00256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/06/2023] [Accepted: 03/24/2024] [Indexed: 05/25/2024] Open
Abstract
PURPOSE There is an urgent need to improve access to cancer therapy globally. Several independent initiatives have been undertaken to improve access to cancer medicines, and additional new initiatives are in development. Improved sharing of experiences and increased collaboration are needed to achieve substantial improvements in global access to essential oncology medicines. METHODS The inaugural Access to Essential Cancer Medicines Stakeholder Meeting was organized by ASCO and convened at the June 2022 ASCO Annual Meeting in Chicago, IL, with two subsequent meetings, Union for International Cancer Control World Cancer Congress held in Geneva, Switzerland, in October 2022 and at the ASCO Annual Meeting in June of 2023. Invited stakeholders included representatives from cancer institutes, physicians, researchers, professional societies, the pharmaceutical industry, patient advocacy organizations, funders, cancer organizations and foundations, policy makers, and regulatory bodies. The session was moderated by ASCO. Past efforts and current and upcoming initiatives were initially discussed (2022), updates on progress were provided (2023), and broad agreement on resulting action steps was achieved with participants. RESULTS Summit participants recognized that while much work was ongoing to enhance access to cancer therapeutics globally, communication and synergy across projects and organizations could be enhanced by providing a platform for collaboration and shared expertise. CONCLUSION The summit resulted in new cross-stakeholder insights and planned collaboration addressing barriers to accessing cancer medications. Specific actions and timelines for implementation and reporting were established.
Collapse
Affiliation(s)
- Edward Briercheck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Doug Pyle
- American Society of Clinical Oncology, Alexandria, VA
| | - Cary Adams
- Union for International Cancer Control, Geneva, Switzerland
| | - Rifat Atun
- Department of Global Health and Population, Harvard University, Boston, MA
| | - Christopher Booth
- Division of Cancer Care and Epidemiology, Queen's University, Kingston, Canada
| | | | | | - Andre Ilbawi
- Cancer Programme, World Health Organization, Geneva, Switzerland
| | | | - David Kerr
- Nuffield Department of Clinical Laboratory Sciences, Oxford University, Oxford, United Kingdom
| | - Felicia Knaul
- Department of Public Health Sciences, University of Miami, Miami, FL
| | | | | | | | - Danny Milner
- Union for International Cancer Control, Geneva, Switzerland
| | | | | | | | - Richard Sullivan
- Institute of Cancer Policy, King's College London, London, United Kingdom
| | - Julie Torode
- Institute of Cancer Policy, King's College London, London, United Kingdom
| | - Everett Vokes
- Department of Medicine, University of Chicago, Chicago, IL
| | - Julie Gralow
- American Society of Clinical Oncology, Alexandria, VA
| |
Collapse
|
42
|
Chakraborty N, Brown M, Persaud S, Gallagher G, Trivedi NU, Bach PB, Mitchell AP. Trends in financial payments from industry to US cancer centers, 2014-2021. JNCI Cancer Spectr 2024; 8:pkae015. [PMID: 38825338 PMCID: PMC11144522 DOI: 10.1093/jncics/pkae015] [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: 11/29/2023] [Revised: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Industry payments to US cancer centers are poorly understood. METHODS US National Cancer Institute (NCI)-designated comprehensive cancer centers were identified (n = 51). Industry payments to NCI-designated comprehensive cancer centers from 2014 to 2021 were obtained from Open Payments and National Institutes of Health (NIH) grant funding from NIH Research Portfolio Online Reporting Tools (RePORT). Given our focus on cancer centers, we measured the subset of industry payments related to cancer drugs specifically and the subset of NIH funding from the NCI. RESULTS Despite a pandemic-related decline in 2020-2021, cancer-related industry payments to NCI-designated comprehensive cancer centers increased from $482 million in 2014 to $972 million in 2021. Over the same period, NCI research grant funding increased from $2 481 million to $2 724 million. The large majority of nonresearch payments were royalties and licensing payments. CONCLUSION Industry payments to NCI-designated comprehensive cancer centers increased substantially more than NCI funding in recent years but were also more variable. These trends raise concerns regarding the influence and instability of industry payments.
Collapse
Affiliation(s)
- Nirjhar Chakraborty
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meredith Brown
- US Digital Corps, Office of Technology Transformation Services, US General Services Administration, Washington, DC, USA
| | - Sonia Persaud
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Grace Gallagher
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Niti U Trivedi
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Delfi Diagnostics, Baltimore, MD, USA
| | - Peter B Bach
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Delfi Diagnostics, Baltimore, MD, USA
| | - Aaron P Mitchell
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
43
|
Stockton SS, Ayers GD, Lee C, Laferriere H, Das S, Berlin J. Evolving or Immutable - Phase I Solid Tumor Trials in the Era of Precision Oncology. RESEARCH SQUARE 2024:rs.3.rs-4202155. [PMID: 38746351 PMCID: PMC11092862 DOI: 10.21203/rs.3.rs-4202155/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Purpose In the era of precision oncology (PO), systemic therapies for patients (pts) with solid tumors have shifted from chemotherapy (CT) to targeted therapy (TT) and immunotherapy (IO). This systematic survey describes features of trials enrolling between 2010-2020, focusing on inclusion criteria, type of dose escalation scheme (DES) utilized, and use of expansion cohorts (ECs). Methods A literature search identified phase I studies in adults with solid tumors published January 1, 2000 - December 31, 2020 from 12 journals. We included only studies enrolling between 2010-2020 to better capture the PO era. Two reviewers abstracted data; a third established concordance. Results Of 10,744 studies, 10,195 were non-topical or enrolled prior to 2010; 437 studies were included. The most common drug classes were TT (47.6%), IO (22%), and CT (6.9%). In studies which reported race, patients were predominantly white (61.7%) or Asian (25.7%), followed by black (6.5%) or other (6.1%). Heterogeneity was observed in the reporting and specification of study inclusion criteria. Only 40.1% of studies utilized ECs, and among the studies which used ECS, 46.6% were defined by genomic selection. Rule-based DES were used in 89% of trials; a 3+3 design was used in 80.5%. Of all drugs tested, 37.5% advanced to phase II, while 10.3% garnered regulatory licensure (for an indication tested in phase I). Conclusion In the era of PO, TT and IO have emerged as the most studied agents in phase I trials. Rule-based DES, which are more relevant for escalating CT, are still chiefly utilized.
Collapse
Affiliation(s)
| | | | - Cody Lee
- Vanderbilt University Medical Center
| | | | | | | |
Collapse
|
44
|
Stewart DJ, Bradford JP, Sehdev S, Ramsay T, Navani V, Rawson NSB, Jiang DM, Gotfrit J, Wheatley-Price P, Liu G, Kaplan A, Spadafora S, Goodman SG, Auer RAC, Batist G. New Anticancer Drugs: Reliably Assessing "Value" While Addressing High Prices. Curr Oncol 2024; 31:2453-2480. [PMID: 38785465 PMCID: PMC11119944 DOI: 10.3390/curroncol31050184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Countries face challenges in paying for new drugs. High prices are driven in part by exploding drug development costs, which, in turn, are driven by essential but excessive regulation. Burdensome regulation also delays drug development, and this can translate into thousands of life-years lost. We need system-wide reform that will enable less expensive, faster drug development. The speed with which COVID-19 vaccines and AIDS therapies were developed indicates this is possible if governments prioritize it. Countries also differ in how they value drugs, and generally, those willing to pay more have better, faster access. Canada is used as an example to illustrate how "incremental cost-effectiveness ratios" (ICERs) based on measures such as gains in "quality-adjusted life-years" (QALYs) may be used to determine a drug's value but are often problematic, imprecise assessments. Generally, ICER/QALY estimates inadequately consider the impact of patient crossover or long post-progression survival, therapy benefits in distinct subpopulations, positive impacts of the therapy on other healthcare or societal costs, how much governments willingly might pay for other things, etc. Furthermore, a QALY value should be higher for a lethal or uncommon disease than for a common, nonlethal disease. Compared to international comparators, Canada is particularly ineffective in initiating public funding for essential new medications. Addressing these disparities demands urgent reform.
Collapse
Affiliation(s)
- David J. Stewart
- Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada (J.G.); (P.W.-P.)
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.R.); (R.A.C.A.)
- Life Saving Therapies Network, Ottawa, ON K1H 5E6, Canada; (J.-P.B.); (G.B.)
| | - John-Peter Bradford
- Life Saving Therapies Network, Ottawa, ON K1H 5E6, Canada; (J.-P.B.); (G.B.)
| | - Sandeep Sehdev
- Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada (J.G.); (P.W.-P.)
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.R.); (R.A.C.A.)
- Life Saving Therapies Network, Ottawa, ON K1H 5E6, Canada; (J.-P.B.); (G.B.)
| | - Tim Ramsay
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.R.); (R.A.C.A.)
| | - Vishal Navani
- Division of Medical Oncology, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Nigel S. B. Rawson
- Canadian Health Policy Institute, Toronto, ON M5V 0A4, Canada;
- Macdonald-Laurier Institute, Ottawa, ON K1N 7Z2, Canada
| | - Di Maria Jiang
- University of Toronto, Toronto, ON M5S 3H2, Canada; (D.M.J.); (G.L.); (A.K.); (S.G.G.)
- Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada
| | - Joanna Gotfrit
- Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada (J.G.); (P.W.-P.)
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.R.); (R.A.C.A.)
| | - Paul Wheatley-Price
- Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada (J.G.); (P.W.-P.)
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.R.); (R.A.C.A.)
- Life Saving Therapies Network, Ottawa, ON K1H 5E6, Canada; (J.-P.B.); (G.B.)
| | - Geoffrey Liu
- University of Toronto, Toronto, ON M5S 3H2, Canada; (D.M.J.); (G.L.); (A.K.); (S.G.G.)
- Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada
| | - Alan Kaplan
- University of Toronto, Toronto, ON M5S 3H2, Canada; (D.M.J.); (G.L.); (A.K.); (S.G.G.)
- Family Physicians Airway Group of Canada, Markham, ON L3R 9X9, Canada
| | - Silvana Spadafora
- Algoma District Cancer Program, Sault Ste Marie, ON P6B 0A8, Canada;
| | - Shaun G. Goodman
- University of Toronto, Toronto, ON M5S 3H2, Canada; (D.M.J.); (G.L.); (A.K.); (S.G.G.)
- St. Michael’s Hospital, Unity Health Toronto, and Peter Munk Cardiac Centre, University Health Network, Toronto, ON M5B 1W8, Canada
| | - Rebecca A. C. Auer
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (T.R.); (R.A.C.A.)
- Department of Surgery, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Gerald Batist
- Life Saving Therapies Network, Ottawa, ON K1H 5E6, Canada; (J.-P.B.); (G.B.)
- Centre for Translational Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
45
|
Martinez P, Baghli I, Gourjon G, Seyfried TN. Mitochondrial-Stem Cell Connection: Providing Additional Explanations for Understanding Cancer. Metabolites 2024; 14:229. [PMID: 38668357 PMCID: PMC11051897 DOI: 10.3390/metabo14040229] [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: 03/04/2024] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The cancer paradigm is generally based on the somatic mutation model, asserting that cancer is a disease of genetic origin. The mitochondrial-stem cell connection (MSCC) proposes that tumorigenesis may result from an alteration of the mitochondria, specifically a chronic oxidative phosphorylation (OxPhos) insufficiency in stem cells, which forms cancer stem cells (CSCs) and leads to malignancy. Reviewed evidence suggests that the MSCC could provide a comprehensive understanding of all the different stages of cancer. The metabolism of cancer cells is altered (OxPhos insufficiency) and must be compensated by using the glycolysis and the glutaminolysis pathways, which are essential to their growth. The altered mitochondria regulate the tumor microenvironment, which is also necessary for cancer evolution. Therefore, the MSCC could help improve our understanding of tumorigenesis, metastases, the efficiency of standard treatments, and relapses.
Collapse
Affiliation(s)
- Pierrick Martinez
- Scientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon, 84140 Montfavet, France;
| | - Ilyes Baghli
- International Society for Orthomolecular Medicine, Toronto, ON M4B 3M9, Canada;
| | - Géraud Gourjon
- Scientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon, 84140 Montfavet, France;
| | | |
Collapse
|
46
|
Gannon F. Clinical trials and tribulations. EMBO Rep 2024; 25:1690-1691. [PMID: 38316901 PMCID: PMC11014898 DOI: 10.1038/s44319-024-00079-9] [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: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
We need better post-approval monitoring and reporting to assess the efficiency of new cancer therapies in the real world beyond clinical trials.
Collapse
Affiliation(s)
- Frank Gannon
- QIMR Berghofer Medical Research Institute in Brisbane, Brisbane, QLD, Australia.
| |
Collapse
|
47
|
van Nassau SCMW, Bol GM, van der Baan FH, Roodhart JML, Vink GR, Punt CJA, May AM, Koopman M, Derksen JWG. Harnessing the Potential of Real-World Evidence in the Treatment of Colorectal Cancer: Where Do We Stand? Curr Treat Options Oncol 2024; 25:405-426. [PMID: 38367182 PMCID: PMC10997699 DOI: 10.1007/s11864-024-01186-4] [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] [Accepted: 01/17/2024] [Indexed: 02/19/2024]
Abstract
OPINION STATEMENT Treatment guidelines for colorectal cancer (CRC) are primarily based on the results of randomized clinical trials (RCTs), the gold standard methodology to evaluate safety and efficacy of oncological treatments. However, generalizability of trial results is often limited due to stringent eligibility criteria, underrepresentation of specific populations, and more heterogeneity in clinical practice. This may result in an efficacy-effectiveness gap and uncertainty regarding meaningful benefit versus treatment harm. Meanwhile, conduct of traditional RCTs has become increasingly challenging due to identification of a growing number of (small) molecular subtypes. These challenges-combined with the digitalization of health records-have led to growing interest in use of real-world data (RWD) to complement evidence from RCTs. RWD is used to evaluate epidemiological trends, quality of care, treatment effectiveness, long-term (rare) safety, and quality of life (QoL) measures. In addition, RWD is increasingly considered in decision-making by clinicians, regulators, and payers. In this narrative review, we elaborate on these applications in CRC, and provide illustrative examples. As long as the quality of RWD is safeguarded, ongoing developments, such as common data models, federated learning, and predictive modelling, will further unfold its potential. First, whenever possible, we recommend conducting pragmatic trials, such as registry-based RCTs, to optimize generalizability and answer clinical questions that are not addressed in registrational trials. Second, we argue that marketing approval should be conditional for patients who would have been ineligible for the registrational trial, awaiting planned (non) randomized evaluation of outcomes in the real world. Third, high-quality effectiveness results should be incorporated in treatment guidelines to aid in patient counseling. We believe that a coordinated effort from all stakeholders is essential to improve the quality of RWD, create a learning healthcare system with optimal use of trials and real-world evidence (RWE), and ultimately ensure personalized care for every CRC patient.
Collapse
Affiliation(s)
- Sietske C M W van Nassau
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, PO Box 85500, Utrecht, 3584 CX, The Netherlands.
| | - Guus M Bol
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, PO Box 85500, Utrecht, 3584 CX, The Netherlands
| | - Frederieke H van der Baan
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, PO Box 85500, Utrecht, 3584 CX, The Netherlands
- Department of Epidemiology & Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeanine M L Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, PO Box 85500, Utrecht, 3584 CX, The Netherlands
| | - Geraldine R Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, PO Box 85500, Utrecht, 3584 CX, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Cornelis J A Punt
- Department of Epidemiology & Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anne M May
- Department of Epidemiology & Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, PO Box 85500, Utrecht, 3584 CX, The Netherlands
| | - Jeroen W G Derksen
- Department of Epidemiology & Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
48
|
Fountzilas E, Tsimberidou AM, Hiep Vo H, Kurzrock R. Tumor-agnostic baskets to N-of-1 platform trials and real-world data: Transforming precision oncology clinical trial design. Cancer Treat Rev 2024; 125:102703. [PMID: 38484408 DOI: 10.1016/j.ctrv.2024.102703] [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: 01/08/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
Choosing the right drug(s) for the right patient via advanced genomic sequencing and multi-omic interrogation is the sine qua non of precision cancer medicine. Traditional cancer clinical trial designs follow well-defined protocols to evaluate the efficacy of new therapies in patient groups, usually identified by their histology/tissue of origin of their malignancy. In contrast, precision medicine seeks to optimize benefit in individual patients, i.e., to define who benefits rather than determine whether the overall group benefits. Since cancer is a disease driven by molecular alterations, innovative trial designs, including biomarker-defined tumor-agnostic basket trials, are driving ground-breaking regulatory approvals and deployment of gene- and immune-targeted drugs. Molecular interrogation further reveals the disruptive reality that advanced cancers are extraordinarily complex and individually distinct. Therefore, optimized treatment often requires drug combinations and N-of-1 customization, addressed by a new generation of N-of-1 trials. Real-world data and structured master registry trials are also providing massive datasets that are further fueling a transformation in oncology. Finally, machine learning is facilitating rapid discovery, and it is plausible that high-throughput computing, in silico modeling, and 3-dimensional printing may be exploitable in the near future to discover and design customized drugs in real time.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, St Luke's Clinic, Thessaloniki, Greece; European University Cyprus, German Oncology Center, Nicosia, Cyprus
| | - Apostolia-Maria Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA.
| | - Henry Hiep Vo
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, France; Medical College of Wisconsin, USA
| |
Collapse
|
49
|
Del Paggio JC, Naipaul R, Gavura S, Mercer RE, Koven R, Gyawali B, Wilson BE, Booth CM. Cost and value of cancer medicines in a single-payer public health system in Ontario, Canada: a cross-sectional study. Lancet Oncol 2024; 25:431-438. [PMID: 38547890 DOI: 10.1016/s1470-2045(24)00072-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND The financial impact of cancer medicines on health systems is not well known. We describe temporal trends in expenditure on cancer medicines within the single-payer health system of Ontario, Canada, and the extent of clinical benefit these treatments offer. METHODS In this cross-sectional study, we identified cancer medicines and expenditures from formularies and costing databases (the New Drug Funding Program, Ontario Drug Benefit Program, and The High-Cost Therapy Funding Program) during 10 consecutive years (April 1, 2012, to March 31, 2022) in Ontario, Canada. For intravenous medicines, we applied the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS) to identify expenditures associated with substantial clinical benefit. We also identified treatments associated with improved overall survival or quality of life. FINDINGS 69 intravenous and 98 oral or injectable medicines were funded during 2012-22. Annual expenditure on cancer medicines increased by approximately 15% per year during 2012-22; the increase was more rapid in the most recent 4 years. Total expenditure on cancer medicines in the 2021-22 financial year was CA$1·7 billion. Immune checkpoint inhibitors were the single biggest expense by class ($284 million), representing 17% of the entire cancer medicine annual budget. Drugs with the highest individual costs were lenalidomide ($178 million) and pembrolizumab ($163 million), each accounting for around 10% of the entire budget. 29 (76%) of 38 indications eligible for ESMO-MCBS scoring met the threshold for substantial clinical benefit. Eight (21%) indications had no randomised trial evidence of improved overall survival, and only four (11%) were associated with improved QOL. $346 million (67% of the expenditure on intravenous cancer medicines) was spent on drugs that improved median overall survival by more than 6 months, $82 million (16%) was spent on medicines with overall survival gains of 3-6 months, and $32 million (6%) was spent on medicines with overall survival gains of less than 3 months. $53 million (10%) was spent on medicines with no established improvement in overall survival. INTERPRETATION Costs of cancer medicines to the Canadian health system are increasing rapidly. Most funded indications met thresholds for substantial clinical benefit and two-thirds of the expenditure were for medicines that improve survival by more than 6 months. Whether this cost trajectory can be maintained in a sustainable, equitable, high-quality health system is unclear. Efforts are needed to ensure the price of medicines with substantial benefit is affordable and funding of treatments with very modest benefit might need to be re-assessed, particularly when alternative supportive and palliative therapies are available. FUNDING None.
Collapse
Affiliation(s)
- Joseph C Del Paggio
- Department of Oncology, Thunder Bay Regional Health Sciences Centre and Northern Ontario School of Medicine University, Thunder Bay, ON, Canada
| | - Rohini Naipaul
- Ontario Health (Cancer Care Ontario), Provincial Drug Reimbursement Programs, Toronto, ON, Canada
| | - Scott Gavura
- Ontario Health (Cancer Care Ontario), Provincial Drug Reimbursement Programs, Toronto, ON, Canada
| | - Rebecca E Mercer
- Ontario Health (Cancer Care Ontario), Provincial Drug Reimbursement Programs, Toronto, ON, Canada; Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada
| | - Rachel Koven
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Bishal Gyawali
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON, Canada; Department of Oncology, Queen's University, Kingston, ON, Canada; Public Health Sciences, Queen's University, Kingston, ON, Canada
| | - Brooke E Wilson
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON, Canada; Department of Oncology, Queen's University, Kingston, ON, Canada
| | - Christopher M Booth
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON, Canada; Department of Oncology, Queen's University, Kingston, ON, Canada; Public Health Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
50
|
Stewart DJ, Cole K, Bosse D, Brule S, Fergusson D, Ramsay T. Population Survival Kinetics Derived from Clinical Trials of Potentially Curable Lung Cancers. Curr Oncol 2024; 31:1600-1617. [PMID: 38534955 PMCID: PMC10968953 DOI: 10.3390/curroncol31030122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 05/26/2024] Open
Abstract
Using digitized data from progression-free survival (PFS) and overall survival Kaplan-Meier curves, one can assess population survival kinetics through exponential decay nonlinear regression analyses. To demonstrate their utility, we analyzed PFS curves from published curative-intent trials of non-small cell lung cancer (NSCLC) adjuvant chemotherapy, adjuvant osimertinib in resected EGFR-mutant NSCLC (ADAURA trial), chemoradiotherapy for inoperable NSCLC, and limited small cell lung cancer (SCLC). These analyses permit assessment of log-linear curve shape and estimation of the proportion of patients cured, PFS half-lives for subpopulations destined to eventually relapse, and probability of eventual relapse in patients remaining progression-free at different time points. The proportion of patients potentially cured was 41% for adjuvant controls, 58% with adjuvant chemotherapy, 17% for ADAURA controls, not assessable with adjuvant osimertinib, 15% with chemoradiotherapy, and 12% for SCLC. Median PFS half-life for relapsing subpopulations was 11.9 months for adjuvant controls, 17.4 months with adjuvant chemotherapy, 24.4 months for ADAURA controls, not assessable with osimertinib, 9.3 months with chemoradiotherapy, and 10.7 months for SCLC. For those remaining relapse-free at 2 and 5 years, the cure probability was 74%/96% for adjuvant controls, 77%/93% with adjuvant chemotherapy, 51%/94% with chemoradiation, and 39%/87% with limited SCLC. Relatively easy population kinetic analyses add useful information.
Collapse
Affiliation(s)
- David J. Stewart
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Katherine Cole
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA
| | - Dominick Bosse
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Stephanie Brule
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Dean Fergusson
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| | - Tim Ramsay
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada; (K.C.); (S.B.); (D.F.)
| |
Collapse
|