1
|
Houda I, Dickhoff C, Uyl-de Groot CA, Reguart N, Provencio M, Levy A, Dziadziuszko R, Pompili C, Di Maio M, Thomas M, Brunelli A, Popat S, Senan S, Bahce I. New systemic treatment paradigms in resectable non-small cell lung cancer and variations in patient access across Europe. THE LANCET REGIONAL HEALTH. EUROPE 2024; 38:100840. [PMID: 38476748 PMCID: PMC10928304 DOI: 10.1016/j.lanepe.2024.100840] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 03/14/2024]
Abstract
The treatment landscape of resectable early-stage non-small cell lung cancer (NSCLC) is set to change significantly due to encouraging results from randomized trials evaluating neoadjuvant and adjuvant immunotherapy, as well as adjuvant targeted therapy. As of January 2024, marketing authorization has been granted for four new indications in Europe, and regulatory approvals for other study regimens are expected. Because cost-effectiveness and reimbursement criteria for novel treatments often differ between European countries, access to emerging developments may lead to inequalities due to variations in recommended and available lung cancer care throughout Europe. This Series paper (i) highlights the clinical studies reshaping the treatment landscape in resectable early-stage NSCLC, (ii) compares and contrasts approaches taken by the European Medicines Agency (EMA) for drug approval to that taken by the United States Food and Drug Administration (FDA), and (iii) evaluates the differences in access to emerging treatments from an availability perspective across European countries.
Collapse
Affiliation(s)
- Ilias Houda
- Department of Pulmonary Medicine, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Chris Dickhoff
- Department of Cardiothoracic Surgery, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Carin A. Uyl-de Groot
- Erasmus School of Health Policy & Management/Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - Noemi Reguart
- Department of Medical Oncology, Hospital Clínic de Barcelona, C. de Villarroel, 170, 08036, Barcelona, Spain
| | - Mariano Provencio
- Department of Medical Oncology, Hospital Universitario Puerta De Hierro, C. Joaquín Rodrigo, 1, 28222, Majadahonda, Madrid, Spain
| | - Antonin Levy
- Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Université Paris Saclay, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Faculty of Medicine, Medical University of Gdańsk, 80-210, Gdańsk, Poland
| | - Cecilia Pompili
- Department of Thoracic Surgery, University and Hospital Trust – Ospedale Borgo Trento, P.Le A. Stefani, 1, 37126, Verona, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Medical Oncology 1U, AOU Città della Salute e della Scienza di Torino, 10126, Turin, Italy
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research, Heidelberg, Germany
| | - Alessandro Brunelli
- Department of Thoracic Surgery, St. James's University Hospital, Beckett Street, LS9 7TF, Leeds, United Kingdom
| | - Sanjay Popat
- Lung Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Suresh Senan
- Department of Radiation Oncology, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Idris Bahce
- Department of Pulmonary Medicine, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| |
Collapse
|
2
|
More TH, Hiller K, Seifert M, Illig T, Schmidt R, Gronauer R, von Hahn T, Weilert H, Stang A. Metabolomics analysis reveals novel serum metabolite alterations in cancer cachexia. Front Oncol 2024; 14:1286896. [PMID: 38450189 PMCID: PMC10915872 DOI: 10.3389/fonc.2024.1286896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Background Cachexia is a body wasting syndrome that significantly affects well-being and prognosis of cancer patients, without effective treatment. Serum metabolites take part in pathophysiological processes of cancer cachexia, but apart from altered levels of select serum metabolites, little is known on the global changes of the overall serum metabolome, which represents a functional readout of the whole-body metabolic state. Here, we aimed to comprehensively characterize serum metabolite alterations and analyze associated pathways in cachectic cancer patients to gain new insights that could help instruct strategies for novel interventions of greater clinical benefit. Methods Serum was sampled from 120 metastatic cancer patients (stage UICC IV). Patients were grouped as cachectic or non-cachectic according to the criteria for cancer cachexia agreed upon international consensus (main criterium: weight loss adjusted to body mass index). Samples were pooled by cachexia phenotype and assayed using non-targeted gas chromatography-mass spectrometry (GC-MS). Normalized metabolite levels were compared using t-test (p < 0.05, adjusted for false discovery rate) and partial least squares discriminant analysis (PLS-DA). Machine-learning models were applied to identify metabolite signatures for separating cachexia states. Significant metabolites underwent MetaboAnalyst 5.0 pathway analysis. Results Comparative analyses included 78 cachectic and 42 non-cachectic patients. Cachectic patients exhibited 19 annotable, significantly elevated (including glucose and fructose) or decreased (mostly amino acids) metabolites associating with aminoacyl-tRNA, glutathione and amino acid metabolism pathways. PLS-DA showed distinct clusters (accuracy: 85.6%), and machine-learning models identified metabolic signatures for separating cachectic states (accuracy: 83.2%; area under ROC: 88.0%). We newly identified altered blood levels of erythronic acid and glucuronic acid in human cancer cachexia, potentially linked to pentose-phosphate and detoxification pathways. Conclusion We found both known and yet unknown serum metabolite and metabolic pathway alterations in cachectic cancer patients that collectively support a whole-body metabolic state with impaired detoxification capability, altered glucose and fructose metabolism, and substrate supply for increased and/or distinct metabolic needs of cachexia-associated tumors. These findings together imply vulnerabilities, dependencies and targets for novel interventions that have potential to make a significant impact on future research in an important field of cancer patient care.
Collapse
Affiliation(s)
- Tushar H. More
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Martin Seifert
- Asklepios Precision Medicine, Asklepios Hospitals GmbH & Co KgaA, Königstein (Taunus), Germany
- Connexome GmbH, Fischen, Germany
| | - Thomas Illig
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
- Hannover Unified Biobank (HUB), Hannover, Germany
| | - Rudi Schmidt
- Asklepios Precision Medicine, Asklepios Hospitals GmbH & Co KgaA, Königstein (Taunus), Germany
- Immunetrue, Cologne, Germany
| | - Raphael Gronauer
- Asklepios Precision Medicine, Asklepios Hospitals GmbH & Co KgaA, Königstein (Taunus), Germany
- Connexome GmbH, Fischen, Germany
| | - Thomas von Hahn
- Asklepios Hospital Barmbek, Department of Gastroenterology, Hepatology and Endoscopy, Hamburg, Germany
- Asklepios Tumorzentrum Hamburg, Hamburg, Germany
- Semmelweis University, Asklepios Campus Hamburg, Budapest, Hungary
| | - Hauke Weilert
- Asklepios Tumorzentrum Hamburg, Hamburg, Germany
- Semmelweis University, Asklepios Campus Hamburg, Budapest, Hungary
- Asklepios Hospital Barmbek, Department of Hematology, Oncology and Palliative Care Medicine, Hamburg, Germany
| | - Axel Stang
- Asklepios Tumorzentrum Hamburg, Hamburg, Germany
- Semmelweis University, Asklepios Campus Hamburg, Budapest, Hungary
- Asklepios Hospital Barmbek, Department of Hematology, Oncology and Palliative Care Medicine, Hamburg, Germany
| |
Collapse
|
3
|
Ortiz Nunez A, Gonzalez Portela J, Zozaya N, Fernández I. Narrative review of value frameworks in urothelial carcinoma and positioning of enfortumab vedotin. J Med Econ 2024; 27:1222-1231. [PMID: 39258976 DOI: 10.1080/13696998.2024.2403351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/12/2024]
Abstract
AIMS Evaluate existing oncology value frameworks in terms of their methodology, structure, characteristics, and functionality using the example of enfortumab vedotin, an approved therapy for urothelial carcinoma. METHODS A search of PubMed, grey literature, and official websites of relevant international organizations was performed from January 2022 to March 2023. RESULTS Six frameworks were identified and analyzed, including the American Society of Clinical Oncology's assessment framework, European Society for Medical Oncology's Magnitude of Clinical Benefit Scale, the National Comprehensive Cancer Network's Evidence Blocks, Memorial Sloan Kettering Cancer Center's DrugAbacus, Institute for Clinical and Economic Review's assessment framework, and the Drug Assessment Framework. Comparisons across frameworks were challenging, owing to differing approaches, objectives, perspectives, methodology, and criteria. Based on the results of the EV-301 study (NCT03474107), the European Society for Medical Oncology's Magnitude of Clinical Benefit Scale assigned a score of 4 out of 5 to enfortumab vedotin administered after chemotherapy and immunotherapy. The National Comprehensive Cancer Network's Evidence Blocks enabled assessment of enfortumab vedotin compared with other treatments for locally advanced or metastatic urothelial carcinoma, resulting in the positioning of enfortumab vedotin as a preferred regimen after chemotherapy and immunotherapy. CONCLUSIONS Application of value frameworks in oncology can contribute to informed value-based decision-making. However, comparisons across frameworks should be made with caution and limited to the same lines of treatment. Enfortumab vedotin may contribute to optimizing outcomes in patients previously treated with chemotherapy and immunotherapy for locally advanced or metastatic urothelial carcinoma.
Collapse
Affiliation(s)
| | | | - Néboa Zozaya
- Department of Health Affairs & Policy Research, Vivactis Weber, Madrid, Spain
| | - Irene Fernández
- Department of Health Affairs & Policy Research, Vivactis Weber, Madrid, Spain
| |
Collapse
|
4
|
Aryal S, Bachman SL, Lyden K, Clay I. Measuring What Is Meaningful in Cancer Cachexia Clinical Trials: A Path Forward With Digital Measures of Real-World Physical Behavior. JCO Clin Cancer Inform 2023; 7:e2300055. [PMID: 37851933 PMCID: PMC10642875 DOI: 10.1200/cci.23.00055] [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: 04/03/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 10/20/2023] Open
Abstract
PURPOSE The burden of cancer cachexia on patients' health-related quality of life, specifically their physical functioning, is well documented, but clinical trials thus far have failed to show meaningful improvement in physical functioning. The purpose of this review is to summarize existing methods of assessing physical function in cancer cachexia, outline a path forward for measuring what is meaningful to patients using digital measures derived from digital health technologies (DHTs), and discuss the current landscape of digital measures from the clinical and regulatory standpoint. DESIGN For this narrative review, peer-reviewed articles were searched on PubMed, clinical trials records were searched on clinicaltrials.gov, and records of digital measures submitted for regulatory qualification were searched on the US Food and Drug Administration's Drug Development Tool Qualification Program database. RESULTS There are gaps in assessing aspects of physical function that matter to patients. Existing assessment methods such as patient-reported outcomes and objective performance outcomes have limitations, including their episodic nature and burden to patients. DHTs such as wearable sensors can capture real-world physical behavior continuously, passively, and remotely, and may provide a more comprehensive picture of patients' everyday functioning. Recent regulatory submissions showcase potential clinical implementation of digital measures in various therapeutic areas. CONCLUSION Digital measures of real-world physical behavior present an opportunity to detect and demonstrate improvements in physical functioning in cancer cachexia, but evidence-based development is critical. For their use in clinical and regulatory decision making, studies demonstrating meaningfulness to patients as well as feasibility and validation are necessary.
Collapse
|
5
|
Liu X, Lu X, Zhou W, Hahne J, Khoshnood K, Shi X, Zhong Y, Wang X. Informed consent in cancer clinical drug trials in China: a narrative literature review of the past 20 years. Trials 2023; 24:445. [PMID: 37415240 DOI: 10.1186/s13063-023-07482-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Although the number of cancer clinical drug trials is increasing rapidly in China, issues concerning informed consent in this research context are understudied. By performing a narrative literature review, we aim to describe the current situation and identify the most salient challenges affecting informed consent in cancer clinical drug trials among adult patients in China since 2000. METHODS We searched Web of Science (WOS), PubMed, Scopus, EMBASE, the Cochrane Library databases, China National Knowledge Infrastructure (CNKI), China Biomedical Literature Database on Disc (CBMdisc), Chinese Scientific Journals Fulltext Database (CQVIP), and WANFANG Data to identify relevant publications since 2000. Data were extracted by three reviewers on six items pertaining to study type, theme, and challenges. RESULTS We identified 37 unique manuscripts, from which 19 full texts were obtained and six were included in the review. All six studies were published in Chinese journals, and the publication years of the majority (five out of six) of the studies were 2015 or later. The authors of the six studies were all from clinical departments or ethical review committees at five hospitals in China. All of the included publications were descriptive studies. Publications reported challenges related to the following aspects of informed consent: information disclosure, patient understanding, voluntariness, authorization, and procedural steps. CONCLUSION Based on our analysis of publications over the past two decades, there are currently frequent challenges related to various aspects of informed consent in cancer clinical drug trials in China. Furthermore, only a limited number of high-quality research studies on informed consent in cancer clinical drug trials in China are available to date. Efforts toward improvement of informed consent practice, in the form of guidelines or further regulations in China, should draw on both experience from other countries and high-quality local evidence.
Collapse
Affiliation(s)
- Xing Liu
- Medical Ethics Committee, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | - Xiaoran Lu
- School of Humanities, Central South University, Changsha, 410075, Hunan, People's Republic of China
| | - Wei Zhou
- School of Public Administration, Hunan University, Changsha, 410023, Hunan, People's Republic of China
| | - Jessica Hahne
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Kaveh Khoshnood
- Yale School of Public Health, Yale University, 60 College Street, New Haven, CT, 06520, USA
| | - Xiaoting Shi
- Department of Environmental Health Sciences, Yale School of Public Health, 60 College St, New Haven, CT, 06520, USA
| | - Yuqiong Zhong
- School of Humanities, Central South University, Changsha, 410075, Hunan, People's Republic of China
| | - Xiaomin Wang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
6
|
Blakely CM, Weder W, Bubendorf L, He J, Majem M, Shyr Y, Chaft JE. Primary endpoints to assess the efficacy of novel therapeutic approaches in epidermal growth factor receptor-mutated, surgically resectable non-small cell lung cancer: A review. Lung Cancer 2023; 177:59-72. [PMID: 36736076 DOI: 10.1016/j.lungcan.2023.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
While the discovery of oncogenic driver mutations has personalized the metastatic non-small cell lung cancer (NSCLC) treatment landscape with effective targeted therapies, implementation of new treatments in resectable NSCLC has been limited due to the long follow-up needed for overall survival (OS). Until recently, treatment for patients with early-stage resectable NSCLC has been limited to perioperative chemotherapy, which provides modest benefits. However, the regulatory acceptance of two surrogate endpoints for OS has allowed recent approval of both adjuvant osimertinib and atezolizumab, providing patients with new treatment options to improve outcomes. In phase 3 oncology trials, OS has historically been viewed as the gold-standard efficacy measure, but disease-free survival and event-free survival (EFS) are now validated surrogate endpoints for OS in clinical trials and should be considered when mature OS data is unavailable. Another potential surrogate endpoint in the adjuvant NSCLC setting is circulating tumor DNA (ctDNA)-based minimal residual disease (MRD), although prospective validation is needed. For neoadjuvant targeted therapies, EFS, major pathologic response and ctDNA-based MRD are potential surrogate endpoints. To fully translate the success of the personalized treatment advances in the metastatic setting to earlier-stage disease, prospective validation studies of these potential surrogate endpoints that can accelerate the evaluation of drug efficacy are needed. A collaborative effort is also needed from all clinical and regulatory parties to collate surrogate endpoint data for large-scale validation. In this review we discuss the trends in surrogate endpoints used in oncology trials, with a focus on considerations for selecting appropriate primary endpoints in early-stage resectable EGFR-mutant NSCLC, an area of unmet need for novel treatment options.
Collapse
Affiliation(s)
- Collin M Blakely
- Department of Medicine and Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Walter Weder
- Department of Thoracic Surgery, University of Zurich (director Emeritus), Thoraxchirurgie, Klinik Bethanien, Zürich, Switzerland
| | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Jianxing He
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jamie E Chaft
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY 10021, USA.
| |
Collapse
|
7
|
Ludwig WD. Beschleunigte Zulassungsverfahren: Status quo, Probleme und Reformbedarf. INFO HÄMATOLOGIE + ONKOLOGIE 2022. [PMCID: PMC9470231 DOI: 10.1007/s15004-022-9164-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
8
|
Hwang TJ, Kesselheim AS, Tibau A, Lee CC, Vokinger KN. Clinical Benefit and Expedited Approval of Cancer Drugs in the United States, European Union, Switzerland, Japan, Canada, and Australia. JCO Oncol Pract 2022; 18:e1522-e1532. [PMID: 35731996 PMCID: PMC9509186 DOI: 10.1200/op.21.00909] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE: Regulatory agencies have sought to speed up the review of new cancer medicines and reduce delays in approval between countries. We examined trends in regulatory review times and association with clinical benefit for new cancer medicines in six jurisdictions: United States (Food and Drug Administration [FDA]), European Union (European Medicines Agency [EMA]), Switzerland (Swissmedic), Japan (Pharmaceuticals and Medical Devices Agency [PMDA]), Canada (Health Canada), and Australia (Therapeutic Goods Administration). METHODS: We studied all new cancer drugs approved in the six aforementioned jurisdictions from 2007 to 2020. We extracted all applicable expedited programs, total regulatory review times, and, for drugs first approved by the FDA, times to subsequent regulatory approval. Clinical benefit was assessed using the European Society for Medical Oncology-Magnitude of Clinical Benefit Scale value framework and ASCO-Cancer Research Committee's targets. Nonparametric Kruskal-Wallis test was used to compare total review times for high versus low clinical benefit drugs. RESULTS: One hundred and twenty eight drugs received initial approval in at least one of the six included jurisdictions. Most drugs approved by the FDA (91%) and Health Canada (59%) qualified for at least one expedited program within those jurisdictions, compared with 46% of EMA approvals and 18% of PMDA approvals. The FDA was the first regulator to approve 102 (80%) drugs. Delays in submission accounted for a median of 20.2% (EMA) to 83.8% (PMDA) of the time to subsequent approval. There was no association between high clinical benefit and shorter total review times. CONCLUSION: Most new cancer therapies were approved first by the FDA, and delays in submission of regulatory applications accounted for substantial delays in approving cancer drugs in other countries. Regulators should prioritize faster review for drugs with high clinical benefit.
Collapse
Affiliation(s)
- Thomas J. Hwang
- Cancer Innovation and Regulation Initiative, Lank Center for Genitourinary Cancer, Dana-Farber Cancer Institute and Division of Urological Surgery, Brigham and Women's Hospital, Boston, MA
- 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
| | - 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
| | - Ariadna Tibau
- Department of Oncology, Hospital de la Santa Creu i Sant Pau, Institut d’Investigació Biomèdica Sant Pau and Universitat Autònoma de Barcelona, Barcelona, Spain
| | - ChangWon C. Lee
- 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
| | - Kerstin N. Vokinger
- 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
- Institute of Law, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
|
10
|
The Importance of Disease-Free Survival as a Clinical Trial Endpoint: A Qualitative Study Among Canadian Survivors of Lung Cancer. PATIENT-PATIENT CENTERED OUTCOMES RESEARCH 2021; 15:307-316. [PMID: 34643935 DOI: 10.1007/s40271-021-00552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND In lung cancer trials, overall survival is a well-validated and widely used endpoint; yet, in the context of adjuvant or curative intent treatments, disease-free survival (DFS) may be a better indicator of transformative patient outcomes. Although use of DFS is growing, patient perceptions of its relevance have not been established. OBJECTIVE We aimed to understand the importance of DFS as a trial endpoint, from the perspective of survivors of lung cancer. METHODS Web-based qualitative interviews were conducted with Canadian survivors of stage Ib-IIIa lung cancer. Participants described their experiences of cancer diagnosis and treatment, including their treatment goals and priorities. Participants then provided their perspectives on DFS and overall survival, and how well each aligned with their treatment priorities. Thematic analysis was used to explore patterns in responses. RESULTS Among the 18 participants (mean age, 64 years), 83% were female, most (89%) had received surgery, and 56% received chemotherapy. Most participants viewed DFS as an intrinsically meaningful treatment outcome, for reasons such as alignment with treatment goals, and the perception that DFS would help maintain a high quality of life. One individual was interested in DFS only as a potential surrogate for overall survival. Participants desired access to new treatments that improve DFS and emphasized this within the context of promoting patient agency in treatment decision making. CONCLUSIONS These findings suggest DFS is a meaningful endpoint from the perspective of survivors of lung cancer; and may help inform decisions regarding regulatory approval and reimbursement of new treatments based on DFS data.
Collapse
|
11
|
Tivey A, Huddar P, Shotton R, Cheese I, Daniels S, Lorigan P, J Lee R. Patient engagement in melanoma research: from bench to bedside. Future Oncol 2021; 17:3705-3716. [PMID: 34213356 DOI: 10.2217/fon-2020-1165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Advances in research have transformed the management of melanoma in the past decade. In parallel, patient advocacy has gained traction, and funders are increasingly prioritizing patient and public involvement. Here we discuss the ways in which patients and the public can be engaged in different stages of the research process, from developing, prioritizing and refining the research question to preclinical studies and clinical trials, then finally to ongoing research in the clinic. We discuss the challenges and opportunities that exist at each stage in order to ensure that a representative population of patients and the public contribute to melanoma research both now and in the future.
Collapse
Affiliation(s)
- Ann Tivey
- The Christie NHS Foundation Trust, Wilmslow Road, M20 4BX, UK
- Melanoma Focus, Queen Anne House, Gonville Place, Cambridge, CB1 1ND, UK
| | - Prerana Huddar
- The Christie NHS Foundation Trust, Wilmslow Road, M20 4BX, UK
| | - Rohan Shotton
- The Christie NHS Foundation Trust, Wilmslow Road, M20 4BX, UK
| | - Imogen Cheese
- Division of Cancer Sciences, The University of Manchester, Oxford Road, M13 9PL, UK
- Melanoma Patient Conference, Willow Bank, Prince Crescent, Staunton, Gloucestershire, GL19 3RF, UK
| | - Susanna Daniels
- Melanoma Patient Conference, Willow Bank, Prince Crescent, Staunton, Gloucestershire, GL19 3RF, UK
- Patient Author
| | - Paul Lorigan
- The Christie NHS Foundation Trust, Wilmslow Road, M20 4BX, UK
- Melanoma Focus, Queen Anne House, Gonville Place, Cambridge, CB1 1ND, UK
| | - Rebecca J Lee
- The Christie NHS Foundation Trust, Wilmslow Road, M20 4BX, UK
- Melanoma Focus, Queen Anne House, Gonville Place, Cambridge, CB1 1ND, UK
| |
Collapse
|
12
|
Hedlin H, Garcia A, Weng Y, He Z, Sundaram V, Bunning B, Balasubramanian V, Cunanan K, Kapphahn K, Gummidipundi S, Purington N, Boulos M, Desai M. Clinical trials in a COVID-19 pandemic: Shared infrastructure for continuous learning in a rapidly changing landscape. Clin Trials 2021; 18:324-334. [PMID: 33535821 PMCID: PMC8172421 DOI: 10.1177/1740774520988298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Clinical trials, conducted efficiently and with the utmost integrity, are a key component in identifying effective vaccines, therapies, and other interventions urgently needed to solve the COVID-19 crisis. Yet launching and implementing trials with the rigor necessary to produce convincing results is a complicated and time-consuming process. Balancing rigor and efficiency involves relying on designs that employ flexible features to respond to a fast-changing landscape, measuring valid endpoints that result in translational actions and disseminating findings in a timely manner. We describe the challenges involved in creating infrastructure with potential utility for shared learning. METHODS We have established a shared infrastructure that borrows strength across multiple trials. The infrastructure includes an endpoint registry to aid in selecting appropriate endpoints, a registry to facilitate establishing a Data & Safety Monitoring Board, common data collection instruments, a COVID-19 dedicated design and analysis team, and a pragmatic platform protocol, among other elements. RESULTS The authors have relied on the shared infrastructure for six clinical trials for which they serve as the Data Coordinating Center and have a design and analysis team comprising 15 members who are dedicated to COVID-19. The authors established a pragmatic platform to simultaneously investigate multiple treatments for the outpatient with adaptive features to add or drop treatment arms. CONCLUSION The shared infrastructure provides appealing opportunities to evaluate disease in a more robust manner with fewer resources and is especially valued during a pandemic where efficiency in time and resources is crucial. The most important element of the shared infrastructure is the pragmatic platform. While it may be the most challenging of the elements to establish, it may provide the greatest benefit to both patients and researchers.
Collapse
Affiliation(s)
- Haley Hedlin
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ariadna Garcia
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yingjie Weng
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ziyuan He
- Sean N. Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vandana Sundaram
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Bryan Bunning
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Vidhya Balasubramanian
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kristen Cunanan
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kristopher Kapphahn
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Santosh Gummidipundi
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Natasha Purington
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mary Boulos
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Manisha Desai
- Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
13
|
Quality of Life in Vulnerable Older Patients with Metastatic Colorectal Cancer Receiving Palliative Chemotherapy-The Randomized NORDIC9-Study. Cancers (Basel) 2021; 13:cancers13112604. [PMID: 34073363 PMCID: PMC8198682 DOI: 10.3390/cancers13112604] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 11/17/2022] Open
Abstract
Quality of life data from randomized trials are lacking in older patients with metastatic colorectal cancer (mCRC). In the randomized NORDIC9-study, reduced-dose S1+oxaliplatin (SOx) showed superior efficacy compared to full-dose S1 monotherapy. We hypothesized that treatment with SOx does not result in inferior quality of life. Patients with mCRC aged ≥70 years and that were not a candidate for standard combination chemotherapy were included and randomly assigned to receive either S1 or SOx. The EORTC QLQ-C30 questionnaire was completed at baseline, after 9, and 18 weeks. The primary endpoint was global Quality of Life (QoL) at 9 weeks. For statistical analysis, a non-inferiority design was chosen applying linear mixed effects models for repeated measurements. The results were interpreted according to statistical significance and anchor-based, clinically relevant between-group minimally important differences (MID). A total of 160 patients aged (median (Interquartile range (IQR))) 78 years (76-81) were included. The QLQ-C30 questionnaire was completed by 150, 100, and 60 patients at baseline, at 9, and 18 weeks, respectively. The difference at 9 weeks in global QoL was 6.85 (95%CI-1.94; 15.65) and 7.37 (0.70; 14.05) in the physical functioning domain in favor of SOx exceeding the threshold for MID. At 18 weeks, the between-group MID in physical functioning was preserved. Dose-reduced combination chemotherapy may be recommended in vulnerable older patients with mCRC, rather than full-dose monotherapy.
Collapse
|
14
|
Godman B, Hill A, Simoens S, Selke G, Selke Krulichová I, Zampirolli Dias C, Martin AP, Oortwijn W, Timoney A, Gustafsson LL, Voncina L, Kwon HY, Gulbinovic J, Gotham D, Wale J, Cristina Da Silva W, Bochenek T, Allocati E, Kurdi A, Ogunleye OO, Meyer JC, Hoxha I, Malaj A, Hierländer C, Sauermann R, Hamelinck W, Petrova G, Laius O, Langner I, Yfantopoulos J, Joppi R, Jakupi A, Greiciute-Kuprijanov I, Vella Bonanno P, Piepenbrink JH, de Valk V, Wladysiuk M, Marković-Peković V, Mardare I, Fürst J, Tomek D, Obach Cortadellas M, Zara C, Pontes C, McTaggart S, Laba TL, Melien Ø, Wong-Rieger D, Bae S, Hill R. Potential approaches for the pricing of cancer medicines across Europe to enhance the sustainability of healthcare systems and the implications. Expert Rev Pharmacoecon Outcomes Res 2021; 21:527-540. [PMID: 33535841 DOI: 10.1080/14737167.2021.1884546] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: There are growing concerns among European health authorities regarding increasing prices for new cancer medicines, prices not necessarily linked to health gain and the implications for the sustainability of their healthcare systems.Areas covered: Narrative discussion principally among payers and their advisers regarding potential approaches to the pricing of new cancer medicines.Expert opinion: A number of potential pricing approaches are discussed including minimum effectiveness levels for new cancer medicines, managed entry agreements, multicriteria decision analyses (MCDAs), differential/tiered pricing, fair pricing models, amortization models as well as de-linkage models. We are likely to see a growth in alternative pricing deliberations in view of ongoing challenges. These include the considerable number of new oncology medicines in development including new gene therapies, new oncology medicines being launched with uncertainty regarding their value, and continued high prices coupled with the extent of confidential discounts for reimbursement. However, balanced against the need for new cancer medicines. This will lead to greater scrutiny over the prices of patent oncology medicines as more standard medicines lose their patent, calls for greater transparency as well as new models including amortization models. We will be monitoring these developments.
Collapse
Affiliation(s)
- Brian Godman
- Division of Clinical Pharmacology, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Division of Pharmacoepidemiology, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.,Division of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa.,School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Andrew Hill
- Institute of Translational Medicine, University of Liverpool, UK
| | - Steven Simoens
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Belgium
| | - Gisbert Selke
- Wissenschaftliches Institut Der AOK (WIdO), Berlin, Germany
| | - Iva Selke Krulichová
- Department of Medical Biophysics, Faculty of Medicine in Hradec Králové, Charles University, Hradec, Králové, Czech Republic
| | - Carolina Zampirolli Dias
- Faculty of Pharmacy, Postgraduate Program in Medicines and Pharmaceutical Services, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.,SUS Collaborating Centre for Technology Assessment and Excellence in Health (CCATES), Belo Horizonte, Minas Gerais, Brazil
| | - Antony P Martin
- Faculty of Health and Life Sciences, Liverpool, UK.,QC Medica, Sim Balk Lane, York UK
| | - Wija Oortwijn
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Angela Timoney
- Division of Pharmacoepidemiology, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.,NHS Lothian, Edinburgh, UK
| | - Lars L Gustafsson
- Division of Clinical Pharmacology, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | - Hye-Young Kwon
- Division of Pharmacoepidemiology, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.,Division of Biology & Public Health, Mokwon University, Daejeon, Korea
| | - Jolanta Gulbinovic
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Janet Wale
- Independent Consumer Advocate, Brunswick, Victoria, Australia
| | - Wânia Cristina Da Silva
- Faculty of Pharmacy, Postgraduate Program in Medicines and Pharmaceutical Services, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.,Data and Knowledge Integration Center for Health(CIDACS), Fundação Oswaldo Cruz (FIOCRUZ)/ Salvador, Bahia, Brazil
| | - Tomasz Bochenek
- Department of Nutrition and Drug Research, Faculty of Health Sciences, Jagiellonian University Medical College, Krakow, Poland
| | - Eleonora Allocati
- Istituto Di Ricerche Farmacologiche 'Mario Negri' IRCCS, Milan, Italy
| | - Amanj Kurdi
- Division of Pharmacoepidemiology, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.,Division of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa.,Department of Pharmacology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Olayinka O Ogunleye
- Department of Pharmacology, Therapeutics and Toxicology, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria.,Department of Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Johanna C Meyer
- Division of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - Iris Hoxha
- Department of Pharmacy, Faculty of Medicine, University of Medicine, Tirana, Albania
| | | | - Christian Hierländer
- Department of Pharmaceutical Affairs, Federation of Social Insurances, Vienna, Austria
| | - Robert Sauermann
- Department of Pharmaceutical Affairs, Federation of Social Insurances, Vienna, Austria
| | | | - Guenka Petrova
- Faculty of Pharmacy, Department of Social Pharmacy and Pharmacoeconomics, Medical University of Sofia, Sofia, Bulgaria
| | - Ott Laius
- State Agency of Medicines, Tartu, Estonia
| | - Irene Langner
- Wissenschaftliches Institut Der AOK (WIdO), Berlin, Germany
| | - John Yfantopoulos
- School of National and Kapodistrian University of Athens Athens Greece
| | - Roberta Joppi
- Pharmaceutical Drug Department, Azienda Sanitaria Locale of Verona, Verona, Italy
| | - Arianit Jakupi
- Faculty of Pharmacy, UBT - Higher Education Institution, Prishtina, Kosovo
| | | | - Patricia Vella Bonanno
- Division of Pharmacoepidemiology, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | | | - Vincent de Valk
- National Health Care Institute (ZIN), XH, Diemen, Netherlands
| | | | - Vanda Marković-Peković
- Faculty of Medicine, Department of Social Pharmacy, University of Banja Luka, Banja Luka, Republic of Srpska, Bosnia and Herzegovina
| | - Ileana Mardare
- Faculty of Medicine, Public Health and Management Department, "Carol Davila" University of Medicine and Pharmacy Bucharest, Bucharest, Romania
| | - Jurij Fürst
- Health Insurance Institute, Ljubljana, Slovenia
| | - Dominik Tomek
- Faculty of Medicine, Slovak Medical University in Bratislava, Bratislava, Slovakia
| | | | - Corinne Zara
- Drug Department, Catalan Health Service, Catalan Health Service, Barcelona, Spain
| | - Caridad Pontes
- Drug Department, Catalan Health Service, Catalan Health Service, Barcelona, Spain.,Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma De Barcelona, Barcelona, Spain
| | | | - Tracey-Lea Laba
- Centre for Health Economics Research and Evaluation, University of Technology Sydney, Broadway, Sydney, NSW
| | - Øyvind Melien
- Reviews and Health Technology Assessments, Norwegian Institute of Public Health, Oslo, Norway
| | - Durhane Wong-Rieger
- Health Data Science, Institute of Population Health, Liverpool, Canadian Organization for Rare Disorders, Toronto, Ontario, Canada
| | - SeungJin Bae
- College of Pharmacy, Ewha Woman's University, Seoul, South Korea
| | - Ruaraidh Hill
- Health Data Science, Institute of Population Health Liverpool Reviews and Implementation Group, Whelan Building, University of Liverpool, Liverpool, UK
| |
Collapse
|
15
|
Brundage MD. Revisiting Barriers to Clinical Trials Accrual. J Natl Cancer Inst 2021; 113:219-220. [PMID: 33022708 DOI: 10.1093/jnci/djaa156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 11/14/2022] Open
|
16
|
Saleh RR, Meti N, Ribnikar D, Goldvaser H, Ocana A, Templeton AJ, Seruga B, Amir E. Associations between safety, tolerability, and toxicity and the reporting of health-related quality of life in phase III randomized trials in common solid tumors. Cancer Med 2020; 9:7888-7895. [PMID: 32886422 PMCID: PMC7643655 DOI: 10.1002/cam4.3390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/13/2020] [Accepted: 07/22/2020] [Indexed: 01/09/2023] Open
Abstract
Background Anti‐cancer drugs are approved typically on the basis of efficacy and safety as evaluated in phase III randomized trials (RCTs). Health‐related quality of life (HRQoL) is a direct measure of patient benefit, but is under‐reported. Here we explore associations with reporting of HRQoL data in phase III RCTs in common solid tumors. Methods We searched ClinicalTrials.gov to identify phase III RCTs evaluating new drugs in adults with advanced cancers that completed accrual between January 2005 and October 2016. Data on HRQoL, safety, and tolerability comprising treatment‐related death, treatment discontinuation and commonly reported grade 3 or 4 adverse events (AEs) were extracted. Associations between these measures and reporting of HRQoL data were explored using logistic regression. Results Of 377 phase III RCTs identified initially, 143 studies were analysed and comprised 55% positive trials and 90% industry sponsored trials. HRQoL was listed as an endpoint in 59% trials; and of these, only 65% reported HRQoL data. There were higher odds of reporting HRQoL data for positive trials (OR 2.05, P = .04) and trials published in journals with higher impact factor (OR 1.35, P = .01). Reporting of HRQoL was not associated with treatment‐related death (OR 1.25, P = .40) or treatment discontinuation (OR 1.12, P = .61), but was positively associated with dyspnea and dermatological adverse events. Conclusions HRQoL is reported in only two‐thirds of RCTs that describe collecting such data. Reporting of HRQoL is associated with positive trial outcome and higher journal impact factor, but not associated with overall safety and tolerability of anti‐cancer drugs.
Collapse
Affiliation(s)
- Ramy R Saleh
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Nicholas Meti
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Domen Ribnikar
- Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Hadar Goldvaser
- Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alberto Ocana
- Experimental Therapeutics Unit, Medical Oncology Department, Hospital Clínico San Carlos, and IdISSC, Madrid, Spain.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.,Centro Regional de Investigaciones Biomédicas, Castilla-La Mancha University, Ciudad Real, Spain
| | - Arnoud J Templeton
- Department of Oncology, St. Claraspital, Basel, Switzerland.,Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Bostjan Seruga
- Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Eitan Amir
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
FDA and EMA Approvals of New Breast Cancer Drugs-A Comparative Regulatory Analysis. Cancers (Basel) 2020; 12:cancers12020437. [PMID: 32069837 PMCID: PMC7072445 DOI: 10.3390/cancers12020437] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/07/2020] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide and the solid tumor type for which the highest number of drugs have been approved to date. This study examines new drug approvals for breast cancer by the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA), based on an analysis of regulatory documents from both agencies for the period from 1995 to 2018. Of the 29 breast cancer drugs approved over this time span, 17 received positive decisions from both the FDA and EMA, including all drugs licensed after 2008. Nineteen of the 25 FDA-approved drugs, but none of the EMA approvals, benefited from special regulatory pathways (such as fast track, breakthrough therapy, or priority review). In the U.S.A., four accelerated approvals were granted (of which one, for bevacizumab, was later revoked), while only two drugs received provisional approvals following EMA review. New breast cancer drugs were approved approximately twelve months earlier in the United States than in Europe. These results suggest that a broader use of special regulatory pathways by EMA could help to accelerate access to novel drugs for European breast cancer patients.
Collapse
|