1
|
Vidalis A, Dumoulin O, Godbole M, Proenca CC. The role and value of real-world evidence in health technology decision-making in France, Germany, Italy, Spain, and the UK: insights on external control arms. Int J Technol Assess Health Care 2025; 41:e25. [PMID: 40260460 PMCID: PMC12019763 DOI: 10.1017/s0266462324004720] [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: 12/11/2023] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 04/23/2025]
Abstract
Real-world evidence (RWE) is increasingly used and accepted by health technology assessment (HTA) bodies as supportive evidence to inform the approval of new technologies. However, the criteria driving RWE acceptance are often unclear.This study aims to improve understanding of the role and value of RWE in HTA decision-making and outline the best practices in building real-world external control arms (ECAs).A mixed approach of a targeted literature review and HTA expert interviews was applied. The HTA reports of ten selected technologies and the expert interviews from France, Germany, Italy, Spain, and the UK informed the criteria driving the acceptance of RWE. Overall, the UK and Spanish HTA bodies are more receptive to accepting RWE, whereas the French and German are the least accepting. When RWE is used to substantiate efficacy claims, the level of scrutiny from regulators and HTA bodies is considerably higher than when RWE has different intended uses. Representativeness of the data source, overall transparency in the study and robust methodologies are the key criteria driving RWE acceptance across markets.
Collapse
|
2
|
Symma N, Hensel A, Roether B, Steinhoff B, Bauer R. Real-World Data to Document the Use of Herbal Medicinal Products in Children - Report of a Workshop in Krakow. PLANTA MEDICA 2025; 91:167-172. [PMID: 39933713 DOI: 10.1055/a-2523-3856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
The workshop "Real-world data to document the use of phytopharmaceuticals in children" was organized by the GA Foundation Plants for Health in collaboration with multiple international scientific societies on July 14, 2024, during the International Congress on Natural Products Research in Kraków, Poland. The event focused on leveraging real-world data and real-world evidence to support the use of herbal medicinal products (syn. phytopharmaceuticals) in pediatric care. The workshop aimed to discuss the current state and future approaches for utilizing real-world data/real-world evidence in expanding the use of herbal medicinal products in children and adolescents. Therefore, the workshop highlighted the unmet needs and challenges in documenting the effectiveness and safety of herbal medicinal products in children and emphasized the coordinated exchange and collaboration among academia, industry, and regulatory authorities. After an e-symposium in May 2022 with more than 300 participants and an in-person workshop on this topic in Bonn in June 2023, this workshop in Krakow was the third event of its kind hosted by the Foundation Plants for Health. Participants concluded that interdisciplinary collaboration is highly needed to establish qualified methods of data collection and assessment of real-world data, e.g., establishing requirements to incorporate nonprescription herbal medicinal products into electronic patient records and registries that can be accessed by all stakeholders.
Collapse
Affiliation(s)
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Germany
- Foundation Plant for Health, Neumarkt, Germany
| | - Bernd Roether
- Bionorica SE, Neumarkt, Germany
- Foundation Plant for Health, Neumarkt, Germany
| | - Barbara Steinhoff
- Pharma Deutschland e. V., Bonn, Germany
- Foundation Plant for Health, Neumarkt, Germany
| | - Rudolf Bauer
- Institute of Pharmaceutical Sciences - Pharmacognosy, University of Graz, Austria
- Foundation Plant for Health, Neumarkt, Germany
| |
Collapse
|
3
|
Murphy LA, Akehurst R, Cunningham D, de Pouvourville G, Solà-Morales O. Real-world evidence to support health technology assessment and payer decision making: is it now or never? Int J Technol Assess Health Care 2025; 41:e20. [PMID: 40162485 PMCID: PMC12018852 DOI: 10.1017/s0266462325000145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/30/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVES The aim of this policy article is twofold: (i) to provide a summary and update of recent important policy developments, in particular relevant guidance on the use of real-world data/real-world evidence (RWD/RWE) by health technology assessment (HTA) bodies and (ii) to set out our policy recommendations on how the different elements of an "RWE framework" we have previously developed could support, further enhance and facilitate the use of RWE for HTA purposes and by HTA bodies and payers. METHODS We undertook a targeted review and analysis of recent important policy developments. The aim was to build on our recommendations from previous work on the "RWE Framework," and consider how the relevant tools from our Framework can further enhance and facilitate the use of RWE for HTA purposes and by HTA bodies/payers. RESULTS We provide eight conditions that we argue would, in combination, constitute the optimal use and acceptance of RWD/RWE for HTA. We believe that, should the eight conditions hold, RWD/RWE would enable more efficient access to medicines and healthcare technologies for patients. CONCLUSIONS High-quality, fit-for-purpose RWD/RWE can and should be used more frequently in HTA. Multi-stakeholder and cross-geography collaborative partnerships are needed to align on best practices to optimize the evidence that needs to be generated to satisfy all stakeholders' needs.
Collapse
|
4
|
Johnsen SP, Baas P, Sørensen JB, Chouaid C, Griesinger F, Daumont MJ, Rault C, Emanuel G, Penrod JR, Jacobs H, Muwaffak M, Schoemaker MJ, Munro REJ, Baskaran D, Durand-Zaleski I, O’Donnell JC. Advancing real-world research in thoracic malignancies: learnings from the international I-O Optimise initiative. Future Oncol 2025; 21:867-878. [PMID: 39996596 PMCID: PMC11916385 DOI: 10.1080/14796694.2025.2466416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
In recent years, the thoracic malignancies treatment landscape has become more complex with the emergence of novel targeted and immunotherapy-based treatments. Although beneficial to patients and physicians, this fast-paced therapeutic evolution has increased the complexity of clinical decision-making and amplified the importance of real-world evidence to support data from randomized controlled trials. The international I-O Optimise initiative was established in 2016 to provide real-world insights into the thoracic malignancies treatment landscape, and has since collaborated with 14 data sources across Europe and Canada, allowing access to data from ~ 500,000 patients with non-small-cell lung cancer, small-cell lung cancer, and malignant pleural mesothelioma. This article reviews pertinent I-O Optimise research, with discussion of the methodological/data-related learnings and expectations for future insights.
Collapse
Affiliation(s)
- Søren Paaske Johnsen
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University and Aalborg University Hospital, Gistrup, Denmark
| | - Paul Baas
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pulmonary Disease, Leiden University Medical Hospital, Leiden, The Netherlands
| | | | - Christos Chouaid
- Pneumology Unit, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Frank Griesinger
- Department of Haematology & Oncology, University Department Internal Medicine-Oncology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Melinda J Daumont
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Braine-L’Alleud, Belgium
| | | | - Gabrielle Emanuel
- Real-World Data Analytics Markets, Bristol Myers Squibb, Uxbridge, UK
| | - John R Penrod
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | - Isabelle Durand-Zaleski
- AP-HP Health Economics Research Unit, Hôtel-Dieu Hospital, INSERM UMR 1153 CRESS, UPEC, Paris, France
| | - John C O’Donnell
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
5
|
Lin L, Lucassen MJJ, van der Noort V, Egberts TCG, Beijnen JH, Huitema ADR. The feasibility of using real world data as external control arms in oncology trials. Drug Discov Today 2025; 30:104324. [PMID: 40054765 DOI: 10.1016/j.drudis.2025.104324] [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: 12/04/2023] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/12/2025]
Abstract
Before real world data (RWD)-derived external control arms (ECAs) can be applied in clinical trials within the field of oncology, it must be determined whether these ECAs can function as appropriate and reliable control arms. This review provides an overview of studies in which RWD-derived ECAs were constructed and compared to a randomized clinical trial (RCT) arm. RWD-derived ECAs had similar survival outcomes as the RCT arm of comparison in six of the eight included studies. Therefore, when performing RCTs is not feasible, RWD-derived ECAs can act as control arms in single-arm clinical trials, provided that suitable RWD sources are used and suitable methodological decisions are made.
Collapse
Affiliation(s)
- Lishi Lin
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Merel J J Lucassen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Vincent van der Noort
- Department of Biometrics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Toine C G Egberts
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
6
|
Peltner J, Becker C, Wicherski J, Wortberg S, Aborageh M, Costa I, Ehrenstein V, Fernandes J, Heß S, Horváth-Puhó E, Korcinska Handest MR, Lentzen M, Maguire P, Meedom NH, Moore R, Moore V, Nagy D, McNamara H, Paakinaho A, Pfeifer K, Pylkkänen L, Rajamaki B, Reviers E, Röthlein C, Russek M, Silva C, De Valck D, Vo T, Bräuner E, Fröhlich H, Furtado C, Hartikainen S, Kallio A, Tolppanen AM, Haenisch B. The EU project Real4Reg: unlocking real-world data with AI. Health Res Policy Syst 2025; 23:27. [PMID: 40016823 PMCID: PMC11869640 DOI: 10.1186/s12961-025-01287-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND The use of real-world data is established in post-authorization regulatory processes such as pharmacovigilance of drugs and medical devices, but is still frequently challenged in the pre-authorization phase of medicinal products. In addition, the use of real-world data, even in post-authorization steps, is constrained by the availability and heterogeneity of real-world data and by challenges in analysing data from different settings and sources. Moreover, there are emerging opportunities in the use of artificial intelligence in healthcare research, but also a lack of knowledge on its appropriate application to heterogeneous real-world data sources to increase evidentiary value in the regulatory decision-making and health technology assessment context. METHODS The Real4Reg project aims to enable the use of real-world data by developing user-friendly solutions for the data analytical needs of health regulatory and health technology assessment bodies across the European Union. These include artificial intelligence algorithms for the effective analysis of real-world data in regulatory decision-making and health technology assessment. The project aims to investigate the value of real-world data from different sources to generate high-quality, accessible, population-based information relevant along the product life cycle. A total of four use cases are used to provide good practice examples for analyses of real-world data for the evaluation and pre-authorization stage, the improvement of methods for external validity in observational data, for post-authorization safety studies and comparative effectiveness using real-world data. This position paper introduces the objectives and structure of the Real4Reg project and discusses its important role in the context of existing European projects focussing on real-world data. DISCUSSION Real4Reg focusses on the identification and description of benefits and risks of new and optimized methods in real-world data analysis including aspects of safety, effectiveness, interoperability, appropriateness, accessibility, comparative value creation and sustainability. The project's results will support better decision-making about medicines and benefit patients' health. Trial registration Real4Reg is registered in the HMA-EMA Catalogues of real-world data sources and studies (EU PAS number EUPAS105544).
Collapse
Affiliation(s)
- Jonas Peltner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Cornelia Becker
- Research Division, Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Julia Wicherski
- Research Division, Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Silja Wortberg
- Research Division, Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Mohamed Aborageh
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Inês Costa
- INFARMED, National Authority of Medicines and Health Products, I.P., Health Technology Assessment Department (DATS), Lisbon, Portugal
| | - Vera Ehrenstein
- Department of Clinical Medicine, Department of Clinical Epidemiology, and Center for Population Medicine, Aarhus University, Aarhus, Denmark
| | - Joana Fernandes
- INFARMED, National Authority of Medicines and Health Products, I.P., Information and Strategic Planning Department (DIPE), Lisbon, Portugal
| | - Steffen Heß
- Health Data Lab (FDZ), Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Erzsébet Horváth-Puhó
- Department of Clinical Medicine, Department of Clinical Epidemiology, and Center for Population Medicine, Aarhus University, Aarhus, Denmark
| | | | - Manuel Lentzen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Peggy Maguire
- European Institute for Women's Health (EIWH), Dublin, Ireland
| | | | - Rebecca Moore
- European Institute for Women's Health (EIWH), Dublin, Ireland
| | - Vanessa Moore
- European Institute for Women's Health (EIWH), Dublin, Ireland
| | - Dávid Nagy
- Department of Clinical Medicine, Department of Clinical Epidemiology, and Center for Population Medicine, Aarhus University, Aarhus, Denmark
| | | | - Anne Paakinaho
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Kerstin Pfeifer
- Research Division, Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | | | - Blair Rajamaki
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Evy Reviers
- European Association for Professionals and People with ALS (EUpALS), Louvain, Belgium
| | - Christoph Röthlein
- Research Division, Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Martin Russek
- Research Division, Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany
| | - Célia Silva
- INFARMED, National Authority of Medicines and Health Products, I.P., Information and Strategic Planning Department (DIPE), Lisbon, Portugal
| | - Dirk De Valck
- European Association for Professionals and People with ALS (EUpALS), Louvain, Belgium
| | - Thuan Vo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Elvira Bräuner
- Data Analytics Center, Danish Medicines Agency, Copenhagen, Denmark
| | - Holger Fröhlich
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, Bonn, Germany
| | - Cláudia Furtado
- INFARMED, National Authority of Medicines and Health Products, I.P., Information and Strategic Planning Department (DIPE), Lisbon, Portugal
- NOVA National School of Public Health, NOVA University Lisbon, Lisbon, Portugal
| | | | | | | | - Britta Haenisch
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Research Division, Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Germany.
- Center for Translational Medicine, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
7
|
Windfuhr F, de Vries ST, Melinder M, Dahlqvist T, Almeida D, Sepodes B, Torre C, Wettermark B, Mol PGM. Stakeholders' Perspectives Toward the Use of Patient Registry Data for Decision-Making on Medicines: A Cross-Sectional Survey. Drug Saf 2025:10.1007/s40264-025-01528-7. [PMID: 39987538 DOI: 10.1007/s40264-025-01528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The use of patient registries in regulatory, health technology assessment (HTA), and payer decision-making has gained increasing attention in recent years. Stakeholders' perspectives toward the use of registry-based real-world evidence (RWE) are unknown. OBJECTIVES The purpose of this study was to assess stakeholders' perspectives toward the use of RWE from patient registries in decision-making on medicines and explore factors influencing their intention to use registry data in the future. METHODS European regulators, HTA/payers, and other stakeholders (industry, academia, healthcare professionals, patient representatives) were invited by email to participate in a web-based survey. The survey was open between November 2023 and January 2024 and contained 24 questions including demographics and questions about perspectives toward registry-based data for decision-making purposes. The latter consisted of 5-point Likert scale items based on the theory of planned behavior (TPB), i.e., attitudes, subjective norm, perceived behavioral control, and intention. Descriptive analyses and a logistic regression analysis (outcome: intention; determinants: demographics, attitudes, subjective norm, behavioral control) were performed. RESULTS Included were 191 respondents (response rate: 16%), of whom 110 were regulators (58%), 24 HTA/payers (13%), and 54 other stakeholders (28%). Most respondents were between 41 and 50 years old (32%), 65% were women, and 53% had > 10 years work experience. Respondents considered registry data in the medicinal product lifecycle most informative for characterization of disease epidemiology (mean 4.4; 95% confidence interval (CI) 4.2-4.5), and least informative for comparative effectiveness (mean 3.6; 95% CI 3.4-3.7). Reaching the relevant patient population was perceived as the biggest strength (mean 3.6; 95% CI 3.4-3.8), and data quality as the largest weakness of patient registries (mean 2.4; 95% CI 2.2-2.6). Compared with regulators, HTA/payers had a similar intention to use registry data (Odds ratio (OR) 1.56; 95% CI 0.47-5.16), while other stakeholders were more frequently very open (intention) to using registry data in the future (OR 8.48; 95% CI 3.00-23.98). Respondents from organizations in Northern Europe were less often very open to using registry data in the future than respondents from multinational organizations (OR 0.19; 95% CI 0.04-0.85). Finally, respondents with a high perceived behavioral control concerning the use of registry data were more often very open to using registry data in the future than respondents with a neutral or low perceived behavioral control (OR 3.45; 95% CI 1.37-8.64). CONCLUSIONS The participants in our survey were generally open to increasing the use of registry data in the future. Nevertheless, perceived weaknesses such as data quality and accessibility will need to be addressed to align and improve stakeholders' perspectives on the use of patient registries as an evidence basis for medicines decision-making.
Collapse
Affiliation(s)
- Fabian Windfuhr
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Sieta T de Vries
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
| | - Maria Melinder
- Department of Pharmacy, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Tanja Dahlqvist
- Department of Pharmacy, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Diogo Almeida
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Bruno Sepodes
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Carla Torre
- Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Björn Wettermark
- Department of Pharmacy, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Peter G M Mol
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands.
| |
Collapse
|
8
|
Zong J, Rojubally A, Pan X, Wolf B, Greenfeder S, Upton A, Gdovin Bergeson J. A Review and Comparative Case Study Analysis of Real-World Evidence in European Regulatory and Health Technology Assessment Decision Making for Oncology Medicines. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2025; 28:31-41. [PMID: 39393564 DOI: 10.1016/j.jval.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES Real-world evidence (RWE) is valuable in supporting regulatory and health technology assessment (HTA) decisions; however, the actual contribution to approvals remains elusive. This study aimed to review RWE approaches and use in oncology medicine approvals in Europe and understand cohesion and discrepancy in the acceptance of the RWE by the European Medicines Agency (EMA) and European HTA bodies. METHODS This scoping review involved a search of the EMA database, National Institute for Health and Care Excellence (NICE), Gemeinsamer Bundesausschuss (G-BA), and Haute Autorité de Santé (HAS) websites to identify final reports and appraisals for oncology medicines with references to RWE. The selection was guided by research terms associated with RWE study designs, data sources, and outcomes. Qualitative analysis was used to systemize the data. Case studies assessed by more than one agency were selected for comparative assessment of RWE approach, use, and acceptability. RESULTS RWE was mainly leveraged as an external control for indirect treatment comparisons or contextualization to support clinical trial results by the EMA, NICE, G-BA, and HAS. However, this approach was mostly rejected due to methodology biases. Comparative assessment of RWE acceptability for the same oncology medicines across agencies suggests discrepancies between EMA and European HTA bodies and among NICE, G-BA, and HAS. CONCLUSIONS There is diverging acceptance of RWE in EMA and European HTA bodies with no clear consensus on the most effective way to leverage RWE in approvals. With the introduction of the joint European Union Joint Clinical Assessment in 2025, it is crucial for European HTA bodies and EMA to develop synergetic standards for the use of RWE to ensure equitable and timely access to medicines.
Collapse
Affiliation(s)
- Jihong Zong
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| | | | - Xiaoyun Pan
- Bayer Healthcare Pharmaceuticals Inc, Boston, MA, USA
| | | | | | | | | |
Collapse
|
9
|
Verweij S, Weemers J, Jonker C, Pasmooij AMG. Learning from European regulator-initiated studies for regulatory decision making. Drug Discov Today 2025; 30:104256. [PMID: 39626797 DOI: 10.1016/j.drudis.2024.104256] [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: 01/24/2024] [Revised: 11/09/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
Drug developers have displayed a growing interest in using real-word evidence (RWE) in the pre-marketing phase. Together with initiatives like DARWIN EU®, this interest encourages regulators to initiate their own observational clinical studies, which could be included in regulatory decision making. The Regulatory Science Network Netherlands (RSNN) organised an expert meeting in 2022 to discuss scenarios and learnings related to these regulator-initiated studies, of which the main points (e.g., transparency, independency and stakeholder interaction) have been published previously. In this review, the authors add their own views, underlining the importance of reproducibility and stakeholder interaction. Stakeholders should collaborate to embrace regulator-initiated studies in a timely and transparent manner to realise an optimal European framework for generating RWE to be included in the regulatory decision-making process.
Collapse
Affiliation(s)
- Stefan Verweij
- Dutch Medicines Evaluation Board, Utrecht, the Netherlands; Unit of PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, Groningen University, Groningen, the Netherlands.
| | - Just Weemers
- Janssen Biologics BV, Leiden, the Netherlands; Regulatory Science Network Netherlands, Utrecht, the Netherlands
| | - Carla Jonker
- Dutch Medicines Evaluation Board, Utrecht, the Netherlands
| | - Anna Maria Gerdina Pasmooij
- Dutch Medicines Evaluation Board, Utrecht, the Netherlands; Regulatory Science Network Netherlands, Utrecht, the Netherlands
| |
Collapse
|
10
|
Wilson BE, Booth CM. Real-world data: bridging the gap between clinical trials and practice. EClinicalMedicine 2024; 78:102915. [PMID: 39588211 PMCID: PMC11585814 DOI: 10.1016/j.eclinm.2024.102915] [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: 10/16/2023] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024] Open
Abstract
Real-world data (RWD) are rapidly emerging sources of information for patients, clinicians and regulators. While randomized controlled trials (RCTs) reduce bias and confounding through the randomization process and provide the highest quality of evidence regarding drug efficacy, RCTs may be impractical or unfeasible for rare diseases or disease subsets. And yet, studies attempting to replicate clinical trial results using observational datasets have failed. Given the inherent differences between observational data and clinical trial results, this discordance is not surprising. However, RWD may still have independent value as complementary tools to trial results. In this viewpoint, we explore the challenges of RWD and discuss key questions that clinicians, patients, and regulators will need to consider when faced with positive efficacy data from clinical trials, and negative effectiveness data from real world studies. Finally, we explore novel trial designs that might help bridge the gap from RCTs to RWD.
Collapse
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, UNSW 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
|
11
|
Subramaniam D, Anderson-Smits C, Rubinstein R, Thai ST, Purcell R, Girman C. A Framework for the Use and Likelihood of Regulatory Acceptance of Single-Arm Trials. Ther Innov Regul Sci 2024; 58:1214-1232. [PMID: 39285061 PMCID: PMC11530569 DOI: 10.1007/s43441-024-00693-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/23/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Single-arm clinical trials (SAT) are common in drug and biologic submissions for rare or life-threatening conditions, especially when no therapeutic options exist. External control arms (ECAs) improve interpretation of SATs but pose methodological and regulatory challenges. OBJECTIVE Through narrative reviews and expert input, we developed a framework for considerations that might influence regulatory use and likelihood of regulatory acceptance of an SAT, identifying non-oncology first indication approvals as an area of interest. We systematically analyzed FDA and EMA approvals using SATs as pivotal evidence. The framework guided outcome abstraction on regulatory responses. METHODS We examined all non-oncology FDA and EMA drug and biologic approvals for first indications from 2019 to 2022 to identify those with SAT as pivotal safety or efficacy evidence. We abstracted outcomes, key study design features, regulator responses to SAT and (where applicable) ECA design, and product label content. RESULTS Among 20 SAT-based FDA approvals and 17 SAT-based EMA approvals, most common indications were progressive rare diseases with high unmet need/limited therapeutic options and a natural history without spontaneous improvement. Of the types of comparators, most were natural history cohorts (45% FDA; 47% EMA) and baseline controls (40% FDA; 47% EMA). Common critiques were of non-contemporaneous ECAs, subjective endpoints, and baseline covariate imbalance between arms. CONCLUSION Based on recent FDA and EMA approvals, the likelihood of regulatory success for SATs with ECAs depends on many design, analytic, and data quality considerations. Our framework is useful in early drug development when considering SAT strategies for evidence generation.
Collapse
Affiliation(s)
- Disha Subramaniam
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA.
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | | | - Rebecca Rubinstein
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sydney T Thai
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA
| | | | - Cynthia Girman
- Real World Evidence and Patient Outcomes, CERobs Consulting, Wrightsville Beach, NC, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Prilla S, Groeneveld S, Pacurariu A, Restrepo-Méndez MC, Verpillat P, Torre C, Gartner C, Mol PGM, Naumann-Winter F, Breen KC, Gault N, Gross-Martirosyan L, Benchetrit S, Aylward B, Stoyanova-Beninska V, O'Donovan M, Straus S, Kjaer J, Arlett P. Real-World Evidence to Support EU Regulatory Decision Making-Results From a Pilot of Regulatory Use Cases. Clin Pharmacol Ther 2024; 116:1188-1197. [PMID: 38962830 DOI: 10.1002/cpt.3355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/09/2024] [Indexed: 07/05/2024]
Abstract
Studies using real-world data (RWD) can complement evidence from clinical trials and fill evidence gaps during different stages of a medicine's lifecycle. This review presents the experience resulting from the European Medicines Agency (EMA) pilot to generate RWE to support evaluations by EU regulators and down-stream decision makers from September 2021 to February 2023. A total of 61 research topics were identified for RWE generation during this period, covering a wide range of research questions, primarily generating evidence on medicines safety (22, 36%), followed by questions on the design and feasibility of clinical trials (11, 18%), drug utilization (10, 16%), clinical management (10, 16%), and disease epidemiology. A significant number of questions were related to the pediatric population and/or rare diseases. A total of 27 regulatory-led RWD studies have been conducted. Most studies were descriptive and aimed at estimating incidence and prevalence rates of clinical outcomes including adverse events or to evaluate medicines utilization. The review highlights key learnings to guide further efforts to enable the use and establish the value of real-world evidence (RWE) for regulatory decisions. For instance, there is a need to access additional fit-for-purpose and representative data, and to explore further means to provide timely evidence that meets regulatory timelines. The need for early interactions and close collaboration with study requesters, e.g., from the Agency's scientific Committees, to better understand the research question is equally important. Finally, the review provides our perspective on the way forward to maximize the potential of regulatory-led RWE generation.
Collapse
Affiliation(s)
- Stefanie Prilla
- Data Analytic Analytics and Methods Taskforce, European Medicines Agency, Amsterdam, The Netherlands
| | - Sophie Groeneveld
- Data Analytic Analytics and Methods Taskforce, European Medicines Agency, Amsterdam, The Netherlands
| | - Alexandra Pacurariu
- Data Analytic Analytics and Methods Taskforce, European Medicines Agency, Amsterdam, The Netherlands
| | | | - Patrice Verpillat
- Data Analytic Analytics and Methods Taskforce, European Medicines Agency, Amsterdam, The Netherlands
| | - Carla Torre
- Departamento de Farmácia, Farmacologia e Tecnologias em Saúde, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | | | - Peter G M Mol
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, Groningen, The Netherlands
- Dutch Medicines Evaluation Board, CBG-MEB, Utrecht, The Netherlands
| | | | | | - Nathalie Gault
- French National Agency for Medicines and Health Products Safety, Saint-Denis, France
| | | | - Sylvie Benchetrit
- French National Agency for Medicines and Health Products Safety, Saint-Denis, France
| | - Brian Aylward
- Health Products Regulatory Authority, Dublin, Ireland
| | | | | | - Sabine Straus
- Dutch Medicines Evaluation Board, CBG-MEB, Utrecht, The Netherlands
- Department of Medical Informatics, Erasmus Medical Center, Erasmus MC, Rotterdam, The Netherlands
| | - Jesper Kjaer
- Data Analytics Centre, Danish Medicines Agency, Copenhagen, Denmark
| | - Peter Arlett
- Data Analytic Analytics and Methods Taskforce, European Medicines Agency, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Hermans SJF, van der Maas NG, van Norden Y, Dinmohamed AG, Berkx E, Huijgens PC, Rivera DR, de Claro RA, Pignatti F, Versluis J, Cornelissen JJ. Externally Controlled Studies Using Real-World Data in Patients With Hematological Cancers: A Systematic Review. JAMA Oncol 2024; 10:1426-1436. [PMID: 39207765 DOI: 10.1001/jamaoncol.2024.3466] [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: 09/04/2024]
Abstract
Importance The use of real-world data (RWD) external control arms in prospective studies is increasing. The advantages, including the immediate availability of a control population, must be balanced with the requirements of meeting evidentiary standards. Objective To address the question of whether and to what extent the methods of RWD studies compare to standard methods used in randomized clinical trials. Evidence Review A systematic search across 4 electronic databases and Google Scholar was conducted from January 1, 2000, to October 23, 2023. Studies were included in the systematic review if they compared an intervention arm in a clinical trial to an RWD control arm in patients with hematological cancers and if they were published between 2000 and 2023. Findings Thirty-two prospective intervention studies incorporating external control data from RWD sources of patients with hematological cancers were identified. A total of 4306 patients from intervention arms and 10 594 from RWD control arms were included across all studies. Only 2 studies (6%) included prospectively collected RWD. The complete trial inclusion criteria were applied to the RWD cohort in 7 studies (22%). Four studies (13%) published the statistical analysis plan and prespecified use of RWD. A total of 23 studies (72%) applied matching algorithms for trial and RWD cohorts, including matching for demographic, disease, and/or therapy-related characteristics. The end point criteria were the same as the trial in 8 studies (25%). In contrast, 12 studies (38%) used different end points, and 12 (38%) did not provide an end point definition for the RWD. Twelve studies (38%) had a median follow-up difference of less than a year between arms. Eight studies (25%) reported toxic effect data for the trial arm, of which 5 studies reported toxic effect data for the RWD arm. Conclusions and Relevance In this systematic review, limitations were observed in the application of clinical trial eligibility criteria to RWD, statistical rigor and application of matching methods, the definition of end points, follow-up, and reporting of adverse events, which may challenge the conclusions reported in studies using RWD.
Collapse
Affiliation(s)
- Sjoerd J F Hermans
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Niek G van der Maas
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yvette van Norden
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Avinash G Dinmohamed
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Elizabeth Berkx
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Peter C Huijgens
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Donna R Rivera
- US Food and Drug Administration, Silver Spring, Maryland
| | - R Angelo de Claro
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Francesco Pignatti
- Oncology and Hematology Office, European Medicines Agency, Amsterdam, the Netherlands
| | - Jurjen Versluis
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jan J Cornelissen
- Department of Hematology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Oncology and Hematology Office, European Medicines Agency, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Salcher-Konrad M, Nguyen M, Savović J, Higgins JPT, Naci H. Treatment Effects in Randomized and Nonrandomized Studies of Pharmacological Interventions: A Meta-Analysis. JAMA Netw Open 2024; 7:e2436230. [PMID: 39331390 PMCID: PMC11437387 DOI: 10.1001/jamanetworkopen.2024.36230] [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: 06/05/2024] [Accepted: 08/04/2024] [Indexed: 09/28/2024] Open
Abstract
Importance Randomized clinical trials (RCTs) are widely regarded as the methodological benchmark for assessing clinical efficacy and safety of health interventions. There is growing interest in using nonrandomized studies to assess efficacy and safety of new drugs. Objective To determine how treatment effects for the same drug compare when evaluated in nonrandomized vs randomized studies. Data Sources Meta-analyses published between 2009 and 2018 were identified in MEDLINE via PubMed and the Cochrane Database of Systematic Reviews. Data analysis was conducted from October 2019 to July 2024. Study Selection Meta-analyses of pharmacological interventions were eligible for inclusion if both randomized and nonrandomized studies contributed to a single meta-analytic estimate. Data Extraction and Synthesis For this meta-analysis using a meta-epidemiological framework, separate summary effect size estimates were calculated for nonrandomized and randomized studies within each meta-analysis using a random-effects model and then these estimates were compared. The reporting of this study followed the Guidelines for Reporting Meta-Epidemiological Methodology Research and relevant portions of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) reporting guideline. Main Outcome and Measures The primary outcome was discrepancies in treatment effects obtained from nonrandomized and randomized studies, as measured by the proportion of meta-analyses where the 2 study types disagreed about the direction or magnitude of effect, disagreed beyond chance about the effect size estimate, and the summary ratio of odds ratios (ROR) obtained from nonrandomized vs randomized studies combined across all meta-analyses. Results A total of 346 meta-analyses with 2746 studies were included. Statistical conclusions about drug benefits and harms were different for 130 of 346 meta-analyses (37.6%) when focusing solely on either nonrandomized or randomized studies. Disagreements were beyond chance for 54 meta-analyses (15.6%). Across all meta-analyses, there was no strong evidence of consistent differences in treatment effects obtained from nonrandomized vs randomized studies (summary ROR, 0.95; 95% credible interval [CrI], 0.89-1.02). Compared with experimental nonrandomized studies, randomized studies produced on average a 19% smaller treatment effect (ROR, 0.81; 95% CrI, 0.68-0.97). There was increased heterogeneity in effect size estimates obtained from nonrandomized compared with randomized studies. Conclusions and Relevance In this meta-analysis of treatment effects of pharmacological interventions obtained from randomized and nonrandomized studies, there was no overall difference in effect size estimates between study types on average, but nonrandomized studies both overestimated and underestimated treatment effects observed in randomized studies and introduced additional uncertainty. These findings suggest that relying on nonrandomized studies as substitutes for RCTs may introduce additional uncertainty about the therapeutic effects of new drugs.
Collapse
Affiliation(s)
- Maximilian Salcher-Konrad
- Department of Health Policy, London School of Economics and Political Science, London, United Kingdom
- World Health Organization Collaborating Centre for Pharmaceutical Pricing and Reimbursement Policies, Pharmacoeconomics Department, Gesundheit Österreich GmbH (GÖG)/Austrian National Public Health Institute, Vienna, Austria
| | - Mary Nguyen
- Department of Health Policy, London School of Economics and Political Science, London, United Kingdom
- Department of Family and Community Medicine, University of California, San Francisco
| | - Jelena Savović
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- National Institute for Health and Care Research Applied Research Collaboration West, University Hospitals Bristol and Weston National Health Service Foundation Trust, Bristol, United Kingdom
| | - Julian P. T. Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- National Institute for Health and Care Research Applied Research Collaboration West, University Hospitals Bristol and Weston National Health Service Foundation Trust, Bristol, United Kingdom
| | - Huseyin Naci
- Department of Health Policy, London School of Economics and Political Science, London, United Kingdom
| |
Collapse
|
15
|
Gräf DD, Westphal L, Hallgreen CE. The life cycle of vaccines evaluated by the European Medicines Agency. Vaccine 2024; 42:126186. [PMID: 39121512 DOI: 10.1016/j.vaccine.2024.126186] [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: 05/10/2024] [Revised: 06/28/2024] [Accepted: 07/26/2024] [Indexed: 08/11/2024]
Abstract
BACKGROUND vaccines are complex products used in healthy populations. They should be carefully regulated, and benefits should clearly outweigh risks. OBJECTIVES To describe the evidence used to support benefit-risk evaluations of vaccines centrally assessed by the European Medicines Agency (EMA), and to identify if real-world data (RWD) was used throughout the vaccine life cycle. METHODS Cohort study of vaccines approved in the European Union. Inclusion criteria comprised having ATC code J07 and being centrally approved between 2012 and 2022. We collected data from regulatory documents, study protocols, and, when necessary, from scientific publications. Vaccines were followed from initial approval up to March 2023. RESULTS We included 31 vaccines addressing 17 therapeutic areas. More than 390 studies were used in the process of initial marketing authorisation (MA) and monitoring, and 174 studies were listed in initial risk management plans. We also identified 93 studies in the EU PAS register. At MA, all vaccines had at least one pivotal trial and 27 vaccines had at least one supportive study. Most pivotal trials were randomized, double-blinded and active-controlled, with immunogenicity endpoints as primary outcome. RWD was used for extension of indications and monitoring of at least 4 vaccines, and the undertaking of RWE studies was foreseen in the RMP of at least 17 vaccines. DISCUSSION Our study revealed an important reliance on randomized controlled trials with individual-level randomization, and a significant focus on immunogenicity endpoints. The use of RWD in vaccine assessments so far has been restricted to COVID-19, and despite its challenges and limitations, we believe that efforts to expand adoption of RWE in continuous benefit-risk assessments should be made. We further highlight the need to enhance data transparency and reporting standards since heterogeneity among regulatory documents made it difficult to identify all the studies considered in vaccine evaluations.
Collapse
Affiliation(s)
- Débora D Gräf
- Copenhagen Centre for Regulatory Science (CORS), Department of Pharmacy, University of Copenhagen, Denmark.
| | - Lukas Westphal
- Copenhagen Centre for Regulatory Science (CORS), Department of Pharmacy, University of Copenhagen, Denmark
| | - Christine E Hallgreen
- Copenhagen Centre for Regulatory Science (CORS), Department of Pharmacy, University of Copenhagen, Denmark
| |
Collapse
|
16
|
Thokagevistk K, Coppo C, Rey L, Carelli A, Díez V, Vaselenak S, Oliveira L, Patel A, Sicari E, Ramos T, Schach S, Schirghuber E, Simpson A, Choquet R, Le Lay K. Real-World Evidence to Reinforce Clinical Trial Evidence in Health Technology Assessment: A Critical Review of Real-World Evidence Requirements from Seven Countries and Recommendations to Improve Acceptance. JOURNAL OF MARKET ACCESS & HEALTH POLICY 2024; 12:105-117. [PMID: 38808313 PMCID: PMC11130860 DOI: 10.3390/jmahp12020009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/22/2024] [Accepted: 04/18/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Real-world evidence (RWE) can reinforce clinical trial evidence in health technology assessment (HTA). OBJECTIVES Review HTA bodies' (HTAbs) requirements for RWE, real uses, and acceptance across seven countries (Brazil, Canada, France, Germany, Italy, Spain, and the United Kingdom) and outline recommendations that may improve acceptance of RWE in efficacy/effectiveness assessments and appraisals processes. METHODS RWE requirements were summarized based on HTAbs' guidelines. Acceptance by HTAbs was evaluated based on industry experience and case studies. RESULTS As of June 2022, RWE methodological guidelines were in place in three of the seven countries. HTAbs typically requested analyses based on local data sources, but the preferred study design and data sources differed. HTAbs had individual submission, assessment, and appraisal processes; some allowed early meetings for the protocol and/or results validation, though few involved external experts or medical societies to provide input to assessment and appraisal. The extent of submission, assessment, and appraisal requirements did not necessarily reflect the degree of acceptance. CONCLUSION All the countries reviewed face common challenges regarding the use of RWE. Our proposals address the need to facilitate collaboration and communication with industry and regulatory agencies and the need for specific guidelines describing RWE design and criteria of acceptance throughout the assessment and appraisal processes.
Collapse
Affiliation(s)
| | - Céline Coppo
- IQVIA, 17 bis Tsse. des Reflets, 92400 Courbevoie, France
| | - Laetitia Rey
- IQVIA, 17 bis Tsse. des Reflets, 92400 Courbevoie, France
| | - Amanda Carelli
- Roche, Rua Dr. Rubens Gomes Bueno, 691—Várzea de Baixo, São Paulo 04730-903, Brazil
| | - Veronica Díez
- F. Hoffmann-La Roche Ltd., C/Ribera del Loira 50, 28042 Madrid, Spain
| | | | - Liana Oliveira
- Roche, Rua Dr. Rubens Gomes Bueno, 691—Várzea de Baixo, São Paulo 04730-903, Brazil
| | - Ajay Patel
- Roche Products Ltd., Hexagon Place, Shire Park, Falcon Way, Welwyn Garden City AL7 1TW, UK
| | - Emilia Sicari
- Roche SpA, Viale Gian Battista Stucchi, 110, 20900 Monza, Italy
| | - Teresa Ramos
- F. Hoffmann-La Roche Ltd., C/Ribera del Loira 50, 28042 Madrid, Spain
| | - Susanne Schach
- Roche, Emil-Barell-Straße 1, 79639 Grenzach-Wyhlen, Germany
| | - Erika Schirghuber
- F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Alex Simpson
- F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Remy Choquet
- Roche, 4 Cr de l’Île Seguin, 92100 Boulogne-Billancourt, France
| | - Katell Le Lay
- Roche, 4 Cr de l’Île Seguin, 92100 Boulogne-Billancourt, France
| |
Collapse
|
17
|
Verweij S, Ahmed W, Zhou G, Mavridis D, Nikolakopoulos S, Elferink AJ, Rengerink KO, Bijlsma MJ, Mol PGM, Hak E. Do efficacy results obtained from randomized controlled trials translate to effectiveness data from observational studies for relapsing-remitting multiple sclerosis? Pharmacoepidemiol Drug Saf 2024; 33:e5810. [PMID: 38720409 DOI: 10.1002/pds.5810] [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: 08/02/2023] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Randomized controlled trials are considered the gold standard in regulatory decision making, as observational studies are known to have important methodological limitations. However, real-world evidence may be helpful in specific situations. This review investigates how the effect estimates obtained from randomized controlled trials compare to those obtained from observational studies, using drug therapy for relapsing-remitting multiple sclerosis as an example. STUDY DESIGN AND SETTING A systematic review of randomized controlled trials and observational studies was conducted. The primary outcome was the annualized relapse rate. Using (network) meta-analysis together with posterior predictive distributions, the drug-specific rate ratios from the network of randomized controlled trials were compared with those from the network of observational studies. RESULTS Effect estimates from 26 observational studies showed greater magnitudes and were less precise compared to estimates obtained from 21 randomized controlled trials. Twenty of the 28 treatment comparisons between designs had similar rate ratios. Seven inconsistencies in observed rate ratios could be attributed to two specific disease-modifying therapies. CONCLUSION In this case study, estimates from observational studies predominantly agreed with estimates from randomized controlled trials given their posterior predictive distributions. Multiple observational studies together may therefore supplement additional pivotal randomized controlled trials in relapsing-remitting multiple sclerosis, for instance facilitating the extrapolation of trial results to the broader patient population.
Collapse
Affiliation(s)
- Stefan Verweij
- Unit of PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, Groningen, The Netherlands
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
| | - Wouter Ahmed
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Guiling Zhou
- Unit of PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, Groningen, The Netherlands
| | - Dimitris Mavridis
- Department of Primary Education, University of Ioannina, Ioannina, Greece
| | - Stavros Nikolakopoulos
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
- Department of Psychology, University of Ioannina, Ioannina, Greece
- Data Science and Biostatistics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Maarten J Bijlsma
- Unit of PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, Groningen, The Netherlands
- Laboratory of Population Health, Max Planck Institute for Demographic Research, Rostock, Germany
| | - Peter G M Mol
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eelko Hak
- Unit of PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, Groningen, The Netherlands
| |
Collapse
|
18
|
Davenport C, Gravel P, Wang Y, Williams SA, Wieland A, Mitlak B. Real-World Evidence to Support the Registration of a New Osteoporosis Medicinal Product in Europe. Ther Innov Regul Sci 2024; 58:505-518. [PMID: 38341388 PMCID: PMC11043175 DOI: 10.1007/s43441-024-00616-7] [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: 07/21/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Real-World Evidence (RWE), which has historically been used to support post-approval safety studies, has recently gained acceptance for new drug applications as supportive evidence or as new clinical evidence for medicinal products with orphan designation and/or in disease areas with high unmet need. Here, we present a case study for the use of RWE in the approval of abaloparatide in the European Union (EU) under the tradename Eladynos. In addition to data from the pivotal Phase 3 study, the marketing authorization application (MAA) included clinical data from additional interventional and observational studies, as well as post-marketing data obtained from the United States (US) market since approval of abaloparatide by the Food and Drug Administration (FDA) in 2017. The new interventional studies were not designed to assess fracture efficacy and cardiovascular safety which were topics of concern raised by the Committee for Medicinal Products for Human Use (CHMP) during their review of the initial MAA submitted in 2015. However, these studies taken together with the RWE formed the basis for a new MAA. Prior to the planned resubmission in the EU, national Scientific Advice (SA) was sought on the proposed clinical program, specifically on the relevance of Real-World Data (RWD) derived from an observational study to support and complement the efficacy and safety data already available from prospective randomized clinical trials. This case study demonstrates successful use of RWE to address a previously identified gap raised by the CHMP during the review of an earlier MAA, which led to the approval of Eladynos for the treatment of osteoporosis in the EU.
Collapse
Affiliation(s)
- Colleen Davenport
- Radius Health, Inc., 22 Boston Wharf Road, 7th Floor, Boston, MA, 02210, USA.
| | | | - Yamei Wang
- Radius Health, Inc., 22 Boston Wharf Road, 7th Floor, Boston, MA, 02210, USA
| | | | | | - Bruce Mitlak
- Radius Health, Inc., 22 Boston Wharf Road, 7th Floor, Boston, MA, 02210, USA
| |
Collapse
|
19
|
Verkerk K, Voest EE. Generating and using real-world data: A worthwhile uphill battle. Cell 2024; 187:1636-1650. [PMID: 38552611 DOI: 10.1016/j.cell.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/04/2024] [Accepted: 02/09/2024] [Indexed: 04/02/2024]
Abstract
The precision oncology paradigm challenges the feasibility and data generalizability of traditional clinical trials. Consequently, an unmet need exists for practical approaches to test many subgroups, evaluate real-world drug value, and gather comprehensive, accessible datasets to validate novel biomarkers. Real-world data (RWD) are increasingly recognized to have the potential to fill this gap in research methodology. Established applications of RWD include informing disease epidemiology, pharmacovigilance, and healthcare quality assessment. Currently, concerns regarding RWD quality and comprehensiveness, privacy, and biases hamper their broader application. Nonetheless, RWD may play a pivotal role in supplementing clinical trials, enabling conditional reimbursement and accelerated drug access, and innovating trial conduct. Moreover, purpose-built RWD repositories may support the extension or refinement of drug indications and facilitate the discovery and validation of new biomarkers. This perspective explores the potential of leveraging RWD to advance oncology, highlights its benefits and challenges, and suggests a path forward in this evolving field.
Collapse
Affiliation(s)
- K Verkerk
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - E E Voest
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands.
| |
Collapse
|
20
|
Van Spall HGC, Bastien A, Gersh B, Greenberg B, Mohebi R, Min J, Strauss K, Thirstrup S, Zannad F. The role of early-phase trials and real-world evidence in drug development. NATURE CARDIOVASCULAR RESEARCH 2024; 3:110-117. [PMID: 39196202 DOI: 10.1038/s44161-024-00420-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/22/2023] [Indexed: 08/29/2024]
Abstract
Phase 3 randomized controlled trials (RCTs), while the gold standard for treatment efficacy and safety, are not always feasible, are expensive, can be prolonged and can be limited in generalizability. Other under-recognized sources of evidence can also help advance drug development. Basic science, proof-of-concept studies and early-phase RCTs can provide evidence regarding the potential for clinical benefit. Real-world evidence generated from registries or observational datasets can provide insights into the treatment of rare diseases that often pose a challenge for trial recruitment. Pragmatic trials embedded in healthcare systems can assess the treatment effects in clinical settings among patient populations sometimes excluded from trials. This Perspective discusses potential sources of evidence that may be used to complement explanatory phase 3 RCTs and to speed the development of new cardiovascular medications. Content is derived from the 19th Global Cardiovascular Clinical Trialists meeting (December 2022), involving clinical trialists, patients, clinicians, regulators, funders and industry representatives.
Collapse
Affiliation(s)
- Harriette G C Van Spall
- Department of Medicine, Department of Health Research Methods, Evidence, and Impact; Research Institute of St. Joseph's, McMaster University, Hamilton, Ontario, Canada
- Baim Institute for Clinical Research, Boston, MA, USA
| | | | - Bernard Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Barry Greenberg
- Division of Cardiology, UC San Diego Health, San Diego, CA, USA
| | - Reza Mohebi
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Faiez Zannad
- Université de Lorraine, Inserm Clinical Investigation Center at Institut Lorrain du Coeur et des Vaisseaux, University Hospital of Nancy, Nancy, France.
| |
Collapse
|
21
|
Wicherski J, Haenisch B. [The application of real-world evidence in drug regulatory decision-making]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2024; 67:149-154. [PMID: 38214723 PMCID: PMC10834571 DOI: 10.1007/s00103-023-03830-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024]
Abstract
Drug regulation is a system to support and protect public health. Drugs with market access must be effective, safe and of high quality. Therefore, drug regulatory decision-making by the competent authorities is made on a scientific basis. Real-world evidence (RWE) from real-world data (RWD) has so far predominantly been taken into account in a supportive manner in drug regulatory decision-making with regard to drug safety after marketing authorisation. The extensive potential of RWE for regulatory decision-making processes along the entire product life cycle has been increasingly used and further examined in recent years.This article provides an overview of current applications of RWE in drug regulatory decision-making processes. The potentials of RWE along with the hurdles to be addressed are described and examples of current projects on RWE research for drug regulation are given. The work is based on current international literature as well as examples from international and European initiatives and regulatory practice, which aim to support an increased use of RWD/RWE in regulatory decision-making processes. In order to be able to utilise the potential of RWE even more in the future, it is important to make relevant RWD sources more readily available through research projects and initiatives, to further develop evaluative methods and to establish the significance of RWE.
Collapse
Affiliation(s)
- Julia Wicherski
- Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Deutschland
| | - Britta Haenisch
- Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175, Bonn, Deutschland.
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Bonn, Deutschland.
- Zentrum für Translationale Medizin, Universität Bonn, Bonn, Deutschland.
| |
Collapse
|
22
|
Magerl M, Schiffhorst G, Fanter L, Müller G, Hirche C, Berkemeier F, Aygören E. Patient-level indirect treatment comparison of lanadelumab versus pdC1-INH i.v. in hereditary angioedema patients: PATCH study. Allergy 2024; 79:215-224. [PMID: 37641968 DOI: 10.1111/all.15861] [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: 03/16/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Hereditary angioedema (HAE) is an autosomal dominant inherited disease in which patients suffer from local attacks primarily affecting skin and gastrointestinal tract, and sometimes even the upper respiratory tract leading to asphyxiation. Since head-to-head trials between authorized treatments are lacking, this study compares efficacy and safety of lanadelumab and intravenous plasma-derived C1-esterase inhibitor (pdC1-INH i.v.) in HAE patients on long-term prophylaxis by means of an indirect treatment comparison. METHODS Efficacy and safety of lanadelumab against pdC1-INH i.v. were analyzed in a fully prespecified indirect comparison based on individual patient data (n = 231) from the HELP and CHANGE clinical trials. Primary and secondary efficacy endpoints were compared using a generalized linear model for count data. Confounding variables were identified a priori via systematic literature research and validated by clinical experts. Adjustment of confounders was implemented using a conditional regression model. RESULTS Lanadelumab showed a statistically significant improvement in reduction of HAE attack rates compared to pdC1-INH i.v. across multiple endpoints: Monthly attack rate of patients treated with lanadelumab was less than half compared to pdC1-INH i.v. (Rate ratio: 0.486; 95% CI: 0.253, 0.932). Monthly rate of laryngeal attacks was found to be five times lower for lanadelumab (Rate ratio: 0.2; 95% CI: 0.044, 0.915) and monthly rate of acute treated HAE attacks among lanadelumab patients was about one third of the attack rate of pdC1-INH i.v. patients (Rate ratio: 0.366; 95% CI: 0.185, 0.727). CONCLUSION This study contributes to current knowledge in the treatment of HAE by indicating a statistically significant reduction of HAE attacks under lanadelumab compared to pdC1-INH i.v.
Collapse
Affiliation(s)
- Markus Magerl
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology, Berlin, Germany
| | | | - Lena Fanter
- Takeda Pharma Vertrieb GmbH & Co. KG, Berlin, Germany
| | - Gerrit Müller
- Takeda Pharma Vertrieb GmbH & Co. KG, Berlin, Germany
| | | | | | - Emel Aygören
- Universitätsklinikum Frankfurt, Frankfurt, Germany
| |
Collapse
|
23
|
Rahman M, Dal Pan G, Stein P, Levenson M, Kraus S, Chakravarty A, Rivera DR, Forshee R, Concato J. When can real-world data generate real-world evidence? Pharmacoepidemiol Drug Saf 2024; 33:e5715. [PMID: 37855046 DOI: 10.1002/pds.5715] [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] [Received: 06/12/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023]
Affiliation(s)
- Motiur Rahman
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gerald Dal Pan
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Peter Stein
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mark Levenson
- Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Stefanie Kraus
- Office of Regulatory Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Aloka Chakravarty
- Office of the Commissioner, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Donna R Rivera
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Richard Forshee
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - John Concato
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
24
|
Wharton GT, Becker C, Bennett D, Burcu M, Bushnell G, Ferrajolo C, Kaplan S, McMahon AW, Movva N, Raman SR, Scholle O, Suh M, Sun JW, Horton DB. Overview of global real-world data sources for pediatric pharmacoepidemiologic research. Pharmacoepidemiol Drug Saf 2024; 33:e5695. [PMID: 37690792 PMCID: PMC10840986 DOI: 10.1002/pds.5695] [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: 07/10/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023]
Abstract
PURPOSE Given limited information available on real-world data (RWD) sources with pediatric populations, this study describes features of globally available RWD sources for pediatric pharmacoepidemiologic research. METHODS An online questionnaire about pediatric RWD sources and their attributes and capabilities was completed by members and affiliates of the International Society for Pharmacoepidemiology and representatives of nominated databases. All responses were verified by database representatives and summarized. RESULTS Of 93 RWD sources identified, 55 unique pediatric RWD sources were verified, including data from Europe (47%), United States (38%), multiregion (7%), Asia-Pacific (5%), and South America (2%). Most databases had nationwide coverage (82%), contained electronic health/medical records (47%) and/or administrative claims data (42%) and were linkable to other databases (65%). Most (71%) had limited outside access (e.g., by approval or through local collaborators); only 10 (18%) databases were publicly available. Six databases (11%) reported having >20 million pediatric observations. Most (91%) included children of all ages (birth until 18th birthday) and contained outpatient medication data (93%), while half (49%) contained inpatient medication data. Many databases captured vaccine information for children (71%), and one-third had regularly updated data on pediatric height (31%) and weight (33%). Other pediatric data attributes captured include diagnoses and comorbidities (89%), lab results (58%), vital signs (55%), devices (55%), imaging results (42%), narrative patient histories (35%), and genetic/biomarker data (22%). CONCLUSIONS This study provides an overview with key details about diverse databases that allow researchers to identify fit-for-purpose RWD sources suitable for pediatric pharmacoepidemiologic research.
Collapse
Affiliation(s)
- Gerold T Wharton
- Office of Pediatric Therapeutics, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Claudia Becker
- Basel Pharmacoepidemiology Unit, Division of Clinical Pharmacy & Epidemiology, Department of Pharmaceutical Sciences, University Basel, Basel, Switzerland
| | - Dimitri Bennett
- Global Evidence and Outcomes, Safety Pharmacoepidemiology, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mehmet Burcu
- Department of Epidemiology, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Greta Bushnell
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, New Brunswick, NJ, USA; Rutgers Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy and Aging Research, New Brunswick, New Jersey, USA
| | - Carmen Ferrajolo
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Naples, Italy
- Department of Experimental Medicine, Section of Pharmacology, "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Sigal Kaplan
- Department Pharmacoepidemiology, Teva Pharmaceutical Industries Ltd, Netanya, Israel
| | - Ann W McMahon
- Office of Pediatric Therapeutics, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Naimisha Movva
- EpidStrategies, A Division of ToxStrategies Inc, Rockville, Maryland, USA
| | - Sudha R Raman
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Oliver Scholle
- Department of Clinical Epidemiology, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Mina Suh
- EpidStrategies, A Division of ToxStrategies Inc, Rockville, Maryland, USA
| | - Jenny W Sun
- Safety Surveillance Research, Pfizer Inc., New York, New York, USA
| | - Daniel B Horton
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, New Brunswick, NJ, USA; Rutgers Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy and Aging Research, New Brunswick, New Jersey, USA
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
25
|
Macabeo B, Rotrou T, Millier A, François C, Laramée P. The Acceptance of Indirect Treatment Comparison Methods in Oncology by Health Technology Assessment Agencies in England, France, Germany, Italy, and Spain. PHARMACOECONOMICS - OPEN 2024; 8:5-18. [PMID: 38097828 PMCID: PMC10781913 DOI: 10.1007/s41669-023-00455-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/26/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Randomized controlled trials (RCTs) are the gold standard when comparing treatment effectiveness, and Health Technology Assessment (HTA) agencies state a clear preference for such direct comparisons. When these are not available, an indirect treatment comparison (ITC) is an alternative option. The objective of this study was to assess the acceptance of ITC methods by HTA agencies across England, France, Germany, Italy, and Spain, using oncology cases for a homogeneous sample of HTA evaluations. METHODS The study was conducted on the PrismAccess database in May 2021 to retrieve HTA evaluation reports for oncology treatments for solid tumors, in which an ITC was presented. The analysis was restricted to HTA evaluation reports published between April 2018 and April 2021 in England, France, Germany, Italy, and Spain. Identified HTA evaluation reports were screened and reviewed by two independent reviewers. For each ITC presented, the methodology and its acceptance by the HTA agency were analyzed. RESULTS Five hundred and forty-three HTA evaluation reports were identified, of which 120 (22%) presented an ITC. This proportion was the highest in England (51%) and lowest in France (6%). The overall acceptance rate of ITC methods was 30%, with the highest in England (47%) and lowest in France (0%). Network meta-analysis (NMA; 23%) was the most commonly used ITC technique, with a 39% acceptance rate overall, followed by Bucher ITC (19%; 43% acceptance rate) and matching-adjusted indirect comparison (13%; 33% acceptance rate). The most common criticisms of the ITC methods from HTA agencies related to data limitations (heterogeneity and lack of data; 48% and 43%, respectively) and the statistical methods used (41%). CONCLUSIONS The generally low acceptance rate of ITC methods by HTA agencies in oncology suggests that, whilst in the absence of a direct comparison ITCs may provide relevant evidence, this evidence is not widely considered sufficient for the purpose of HTA evaluations. The perception of ITC methods for the purpose of HTA evaluations varies substantially between countries. There is a need for further clarity on the properties of ITC techniques and the assessment of their results as ITC methods continue to evolve quickly and further techniques may become available in the future.
Collapse
Affiliation(s)
- Bérengère Macabeo
- Faculté des sciences Médicales et Paramédicales, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France.
- Pierre Fabre Laboratories, Paris, France.
| | | | | | - Clément François
- Faculté des sciences Médicales et Paramédicales, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France
| | - Philippe Laramée
- Faculté des sciences Médicales et Paramédicales, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France
- Pierre Fabre Laboratories, Paris, France
| |
Collapse
|
26
|
Yuan L, Rahman M, Concato J. Comparison of two assessments of real-world data and real-world evidence for regulatory decision-making. Clin Transl Sci 2024; 17:e13702. [PMID: 38093484 PMCID: PMC10766019 DOI: 10.1111/cts.13702] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Real-world data (RWD) and real-world evidence (RWE) are increasingly used to support regulatory decision making, but regulatory agencies and stakeholders may apply different definitions for RWD and use different criteria to determine when analysis of such data are considered RWE in decisions on drug approvals. To explore this issue, we reviewed two prominent publications that operationalized the definitions of RWD and RWE when describing the use of RWE in drug approvals by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). Both publications considered noninterventional (observational) studies, RWD as a comparator arm for a single-arm trial, product-related literature reviews, and RWD to support clinical trial implementation (e.g., to identify potential participants) as generating RWE. In contrast, inconsistencies were identified regarding types of data sources and study designs that were considered as not generating RWE. For example, a lack of agreement existed regarding whether RWE is generated when RWD describe therapeutic contexts or are used in phase I/II interventional trials, open-label extension studies, or pharmacovigilance activities. These discrepancies highlight opportunities to develop a consistent understanding of the role of RWE in regulatory decision making for drug approvals among regulatory agencies and stakeholders.
Collapse
Affiliation(s)
- Lily Yuan
- Office of Medical Policy, Center for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
- Division of New Technology, Technology Coding and Pricing Group, Center for MedicareCenters for Medicare & Medicaid ServicesWoodlawnMarylandUSA
| | - Motiur Rahman
- Office of Medical Policy, Center for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - John Concato
- Office of Medical Policy, Center for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
27
|
Jandhyala R. Development of a definition for real-world evidence using the Jandhyala method for observing consensus opinion among medical affairs pharmaceutical physicians. Curr Med Res Opin 2023; 39:1551-1558. [PMID: 36710630 DOI: 10.1080/03007995.2023.2172261] [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: 11/30/2022] [Accepted: 01/20/2023] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Low rates of inclusion of real-world evidence (RWE) during regulation may arise from lack of clarity and consensus on its definition. A conceptually mature definition of RWE may have pragmatic utility, increasing its inclusion during regulation. The aim was to develop a definition of RWE to promote inclusion in regulatory submissions and assess its conceptual maturity. METHODS Thirteen medical affairs pharmaceutical physicians completed two qualitative online surveys to generate items needed in a definition of RWE. Items that reached a consensus index of > 50% (CI > = 0.51) were retained in the final definition. The maturity of the definition was assessed using concept analysis. RESULTS After attrition, 11 participants completed the study and generated 18 items to be included in a definition of RWE. All items reached the consensus threshold and were included. The definition was conceptually mature on three of the four dimensions: the potential for a consensual definition across stakeholders, a description of its characteristics and clear preconditions and outcomes. Further research is needed to delineate the boundaries of RWE. CONCLUSIONS A definition of RWE was generated that may increase its inclusion during medicines regulation, especially with further refinement from regulators and other stakeholders.
Collapse
Affiliation(s)
- Ravi Jandhyala
- Medialis Ltd, Wolverton Mill, Milton Keynes, England, UK
- Centre for Pharmaceutical Medicine Research, King's College University, London, UK
| |
Collapse
|
28
|
V Stackelberg A, Jäschke K, Jousseaume E, Templin C, Jeratsch U, Kosmides D, Steffen I, Gökbuget N, Peters C. Tisagenlecleucel vs. historical standard of care in children and young adult patients with relapsed/refractory B-cell precursor acute lymphoblastic leukemia. Leukemia 2023; 37:2346-2355. [PMID: 37880478 PMCID: PMC10681894 DOI: 10.1038/s41375-023-02042-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023]
Abstract
In the absence of randomized controlled trials comparing tisagenlecleucel vs. standard of care (SOC) in pediatric and young adult patients with relapsed or refractory acute lymphoblastic leukemia (r/r ALL), the objective was to compare the efficacy of tisagenlecleucel with historical controls from multiple disease registries using patient-level adjustment of the historical controls. The analysis is based on patient-level data of three tisagenlecleucel studies (ELIANA, ENSIGN and CCTL019B2001X) vs. three registries in Germany/Austria. Statistical analyses were fully pre-specified and propensity score weighting of the historical controls by fine stratification weights was used to adjust for relevant confounders identified by systematic literature review. Results showed high comparability of cohorts after adjustment with absolute SMD ≤ 0.1 for all pre-specified confounders and favorable outcomes for tisagenlecleucel compared to SOC for all examined endpoints. Hazard ratios for OS(Intention to treat)ITT,adjusted, EFS(Full analysis set)FAS,naïve and RFSFAS,naïve were 0.54 (95% CI: 0.41-0.71, p < 0.001), 0.67 (0.52-0.86, p = 0.001) and 0.77 (0.51-1.18, p = 0.233). The OSITT, adjusted, EFSFAS,naïve and RFSFAS,naive survival probability at 2 years was 59.49% for tisagenlecleucel vs. 36.16% for SOC population, 42.31% vs. 30.23% and 59.60% vs. 54.57%, respectively. Odds ratio for ORRITT,adjusted was 1.99 (1.33-2.97, p < 0.001). Results for OS and ORR were statistically significant after adjustment for confounders and provide evidence supporting a superiority of tisagenlecleucel in r/r ALL given the good comparability of cohorts after adjustment for confounders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ingo Steffen
- Charité University Hospital Berlin, Berlin, Germany
| | | | - Christina Peters
- St. Anna Children's Hospital, Children's Cancer Research Institute, University Vienna, Vienna, Austria
| |
Collapse
|
29
|
Acha V, Barefoot B, Juarez Garcia A, Lehner V, Monno R, Sandler S, Spooner A, Verpillat P. Principles for Good Practice in the Conduct of Non-interventional Studies: The View of Industry Researchers. Ther Innov Regul Sci 2023; 57:1199-1208. [PMID: 37460826 PMCID: PMC10579109 DOI: 10.1007/s43441-023-00544-y] [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: 08/05/2022] [Accepted: 06/02/2023] [Indexed: 10/17/2023]
Abstract
This reflection paper presents a consolidated view of EFPIA on the need for principles for good practice in the generation and use of non-interventional studies (NIS), including overarching principles such as the registration of hypothesis evaluating treatment effect (HETE) studies. We first define NIS and the important adjacencies to clinical trials and relationship with real-world evidence (RWE). We then outline the principles for good practice with respect to appropriate research design, study protocol, fit-for-purpose variables and data quality, analytical methods, bias reduction, transparency in conduct and use, privacy management and ethics review. We conclude with recommendations for action for the research community to promote trust and credibility in the use of NIS.
Collapse
|
30
|
Bloomfield-Clagett B, Rahman M, Smith K, Concato J. Use of Real-World Evidence in Neuroscience-Related New Drug and Biologics License Applications for Novel Therapeutics. Clin Pharmacol Ther 2023; 114:1002-1005. [PMID: 37548904 DOI: 10.1002/cpt.3018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
The US Food and Drug Administration (FDA) is evaluating the potential use of real-world evidence (RWE) in regulatory decision making. Some groups have evaluated the use of RWE in regulatory submissions in the United States and abroad, reporting that reliance on RWE to support new product approvals is relatively common. Confusion regarding the use of RWE in drug-approval decisions may arise, however, based on different application of the terms real-world data (RWD) and RWE. We evaluated RWE in new drug applications (NDAs) and biologics license applications (BLAs) from January 2019 to June 2021 for novel drugs and biologics approved by the FDA with indications related to psychiatry, neurology, pain, or sedation (here, termed neuroscience-related). We sought to determine whether the submissions included RWE and to describe the types of data and study designs used. Thirty neuroscience-related NDAs or BLAs were identified for novel drugs and biologics approved during the time-period of interest. Among these approvals, three applications (10%) were adjudicated as containing RWE, one of which included RWE as primary evidence of effectiveness. Our findings highlight how different operational definitions of the terms RWD and RWE can result in demonstrably different reporting of the use of RWE in regulatory decision making for neuroscience-related novel drugs and biologics. A better understanding of this topic, along with awareness of regulatory definitions of RWE, are important factors to promote accurate tracking and reporting of regulatory submissions involving RWE. These factors can also improve awareness among the stakeholder community regarding the role of RWD and RWE in regulatory decision making.
Collapse
Affiliation(s)
| | - Motiur Rahman
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kimberly Smith
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - John Concato
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
- Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
31
|
Micallef B, Dogné JM, Sultana J, Straus SMJM, Nisticò R, Serracino-Inglott A, Borg JJ. An Exploratory Study of the Impact of COVID-19 Vaccine Spontaneous Reporting on Masking Signal Detection in EudraVigilance. Drug Saf 2023; 46:1089-1103. [PMID: 37707778 DOI: 10.1007/s40264-023-01346-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION During the signal detection process, statistical methods are used to identify drug-event combinations (DECs) which are disproportionately reported when compared with other drugs and events in the entire database. We hypothesise that the high volume of COVID-19 vaccine adverse drug reaction (ADR) reports transmitted to EudraVigilance may have affected the performance of disproportionality statistics used in routine signal detection, potentially resulting in signals either being masked, or false associations being flagged as potential signals. OBJECTIVE Our aim was to study the impact of COVID-19 vaccine spontaneous reporting on statistical signal detection in EudraVigilance. METHODS We recalculated the reporting odds ratio (ROR) for signals that were previously discussed at the level of the Pharmacovigilance Risk Assessment Committee, or signals that were retrieved from EudraVigilance, by omitting COVID-19 vaccine reports from the standard ROR calculation and then comparing the lower confidence interval (LCI) of the recalculated ROR to the LCI of the actual ROR in EudraVigilance. RESULTS In total, 52 signals for 38 active substances were reviewed. For 35 signals, the LCI of the recalculated ROR value was lower than the LCI of the actual ROR (suggesting that COVID-19 vaccine ADR reporting had a positive effect on the strength of the signal) while for 15 signals the LCI of the recalculated ROR value was higher than the LCI of the actual ROR (suggesting that COVID-19 vaccine ADR reporting had an attenuating effect on the strength of the signal). For two signals, no change in the ROR was observed. In our analysis, six significant results were found. Five DECs were found to be masked: bleomycin and immune thrombocytopenia (actual ROR LCI = 0.94, recalculated ROR LCI = 1.02), vortioxetine and heavy menstrual bleeding (actual ROR LCI = 0.3, recalculated ROR LCI = 1.06), caplacizumab and heavy menstrual bleeding (actual ROR LCI = 0.98, recalculated ROR LCI = 3.47), ziprasidone and amenorrhoea (actual ROR LCI = 0.84, recalculated ROR LCI = 1.67), and azacitidine and pericarditis (actual ROR LCI = 0.81, recalculated ROR LCI = 2.01). For the DEC of adalimumab and immune reconstitution inflammatory syndrome, the LCI of the actual ROR value was 1.14 and removing COVID-19 vaccine reporting resulted in an LCI of the recalculated ROR value of 0.94 (below threshold). CONCLUSIONS We demonstrated five cases of masking and one case of false-positive association due to the influence of COVID-19 vaccine spontaneous reporting on the ROR. This suggests that the high number of adverse drug reaction reports for COVID-19 vaccines in EudraVigilance has the potential to affect routine statistical signal detection activities. The impact of COVID-19 vaccine ADR reports on current signal detection practices requires further evaluation and solutions to tackle masking issues in EudraVigilance may need to be developed.
Collapse
Affiliation(s)
- Benjamin Micallef
- Medicines Authority, Sir Temi Żammit Buildings, Malta Life Sciences Park, San Ġwann, SĠN 3000, Malta
| | | | - Janet Sultana
- College of Medicine and Health, University of Exeter, Exeter, UK
| | | | - Robert Nisticò
- School of Pharmacy, Department of Biology, University of Tor Vergata, Rome, Italy
| | - Anthony Serracino-Inglott
- Medicines Authority, Sir Temi Żammit Buildings, Malta Life Sciences Park, San Ġwann, SĠN 3000, Malta
- Department of Pharmacy, University of Malta, Msida, Malta
| | - John-Joseph Borg
- Medicines Authority, Sir Temi Żammit Buildings, Malta Life Sciences Park, San Ġwann, SĠN 3000, Malta.
- School of Pharmacy, Department of Biology, University of Tor Vergata, Rome, Italy.
| |
Collapse
|
32
|
Johansson P, Jonéus P, Langenskiöld S. Causal inferences and real-world evidence: A comparative effectiveness evaluation of abiraterone acetate against enzalutamide. PLoS One 2023; 18:e0293000. [PMID: 37883352 PMCID: PMC10602359 DOI: 10.1371/journal.pone.0293000] [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: 03/20/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Regulatory authorities are recognizing the need for real-world evidence (RWE) as a complement to randomized controlled trials in the approval of drugs. However, RWE needs to be fit for regulatory purposes. There is an ongoing discussion regarding whether pre-publication of a protocol on appropriate repositories, e.g. ClinicalTrials.gov, would increase the quality of RWE or not. This paper illustrates that an observational study based on a pre-published protocol can entail the same level of detail as a protocol for a randomized experiment. The strategy is exemplified by designing a comparative effectiveness evaluation of abiraterone acetate against enzalutamide in clinical practice. These two cancer drugs are prescribed to patients with advanced prostate cancer. Two complementary designs, including pre-analysis plans, were published before data on outcomes and proxy-outcomes were obtained. The underlying assumptions are assessed and both analyses show an increased mortality risk from being prescribed abiraterone acetate compared to enzalutamide.
Collapse
Affiliation(s)
- Per Johansson
- Department of Statistics, Uppsala University, Uppsala, Sweden
- Centre for Health-Economic Research, Uppsala University, Uppsala, Sweden
- Institute for Evaluation of Labour Market and Education Policy, Uppsala University, Uppsala, Sweden
- Tsinghua University, Beijing, China
| | - Paulina Jonéus
- Department of Statistics, Uppsala University, Uppsala, Sweden
| | - Sophie Langenskiöld
- Centre for Health-Economic Research, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
33
|
Zhu R, Vora B, Menon S, Younis I, Dwivedi G, Meng Z, Datta-Mannan A, Manchandani P, Nayak S, Tammara BK, Garhyan P, Iqbal S, Dagenais S, Chanu P, Mukherjee A, Ghobadi C. Clinical Pharmacology Applications of Real-World Data and Real-World Evidence in Drug Development and Approval-An Industry Perspective. Clin Pharmacol Ther 2023; 114:751-767. [PMID: 37393555 DOI: 10.1002/cpt.2988] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
Since the 21st Century Cures Act was signed into law in 2016, real-world data (RWD) and real-world evidence (RWE) have attracted great interest from the healthcare ecosystem globally. The potential and capability of RWD/RWE to inform regulatory decisions and clinical drug development have been extensively reviewed and discussed in the literature. However, a comprehensive review of current applications of RWD/RWE in clinical pharmacology, particularly from an industry perspective, is needed to inspire new insights and identify potential future opportunities for clinical pharmacologists to utilize RWD/RWE to address key drug development questions. In this paper, we review the RWD/RWE applications relevant to clinical pharmacology based on recent publications from member companies in the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) RWD Working Group, and discuss the future direction of RWE utilization from a clinical pharmacology perspective. A comprehensive review of RWD/RWE use cases is provided and discussed in the following categories of application: drug-drug interaction assessments, dose recommendation for patients with organ impairment, pediatric plan development and study design, model-informed drug development (e.g., disease progression modeling), prognostic and predictive biomarkers/factors identification, regulatory decisions support (e.g., label expansion), and synthetic/external control generation for rare diseases. Additionally, we describe and discuss common sources of RWD to help guide appropriate data selection to address questions pertaining to clinical pharmacology in drug development and regulatory decision making.
Collapse
Affiliation(s)
- Rui Zhu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Bianca Vora
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Sujatha Menon
- Clinical Pharmacology, Pfizer Inc., Groton, Connecticut, USA
| | - Islam Younis
- Clinical Pharmacology, Gilead Sciences, Inc., Foster City, California, USA
| | - Gaurav Dwivedi
- Quantitative Clinical Pharmacology, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Zhaoling Meng
- R&D Data and Data Science, Clinical Modeling & Evidence Integration, Sanofi, Cambridge, Massachusetts, USA
| | - Amita Datta-Mannan
- Exploratory Medicine & Pharmacology, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Pooja Manchandani
- Clinical Pharmacology and Exploratory Division, Astellas Pharma Global Development, Northbrook, Illinois, USA
| | | | | | - Parag Garhyan
- Global PK/PD/Pharmacometrics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Shahed Iqbal
- Biomarker Sciences, Gilead Sciences, Inc., Foster City, California, USA
| | - Simon Dagenais
- Real World Evidence Center of Excellence, Pfizer, Inc., New York, New York, USA
| | - Pascal Chanu
- Clinical Pharmacology, Genentech/Roche, Inc., Lyon, France
| | - Arnab Mukherjee
- Clinical Pharmacology, Pfizer Inc., Groton, Connecticut, USA
| | - Cyrus Ghobadi
- Exploratory Medicine & Pharmacology, Eli Lilly and Company, Indianapolis, Indiana, USA
| |
Collapse
|
34
|
Morland K, Gerges C, Elwing J, Visovatti SH, Weatherald J, Gillmeyer KR, Sahay S, Mathai SC, Boucly A, Williams PG, Harikrishnan S, Minty EP, Hobohm L, Jose A, Badagliacca R, Lau EMT, Jing Z, Vanderpool RR, Fauvel C, Leonidas Alves J, Strange G, Pulido T, Qian J, Li M, Mercurio V, Zelt JGE, Moles VM, Cirulis MM, Nikkho SM, Benza RL, Elliott CG. Real-world evidence to advance knowledge in pulmonary hypertension: Status, challenges, and opportunities. A consensus statement from the Pulmonary Vascular Research Institute's Innovative Drug Development Initiative's Real-world Evidence Working Group. Pulm Circ 2023; 13:e12317. [PMID: 38144948 PMCID: PMC10739115 DOI: 10.1002/pul2.12317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/26/2023] [Accepted: 11/21/2023] [Indexed: 12/26/2023] Open
Abstract
This manuscript on real-world evidence (RWE) in pulmonary hypertension (PH) incorporates the broad experience of members of the Pulmonary Vascular Research Institute's Innovative Drug Development Initiative Real-World Evidence Working Group. We aim to strengthen the research community's understanding of RWE in PH to facilitate clinical research advances and ultimately improve patient care. Herein, we review real-world data (RWD) sources, discuss challenges and opportunities when using RWD sources to study PH populations, and identify resources needed to support the generation of meaningful RWE for the global PH community.
Collapse
Affiliation(s)
- Kellie Morland
- Global Medical AffairsUnited Therapeutics CorporationResearch Triangle ParkNorth CarolinaUSA
| | - Christian Gerges
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaViennaAustria
| | - Jean Elwing
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Scott H. Visovatti
- Division of Cardiovascular MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Jason Weatherald
- Department of Medicine, Division of Pulmonary MedicineUniversity of AlbertaEdmontonCanada
| | - Kari R. Gillmeyer
- The Pulmonary CenterBoston University Chobian & Avedisian School of MedicineBostonMassachusettsUSA
- Center for Healthcare Organization & Implementation ResearchVA Bedford Healthcare System and VA Boston Healthcare SystemBedfordMassachusettsUSA
| | - Sandeep Sahay
- Division of Pulmonary, Critical Care & Sleep MedicineHouston Methodist HospitalHoustonTexasUSA
| | - Stephen C. Mathai
- Division of Pulmonary and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Athénaïs Boucly
- Faculté de MédecineUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital BicêtreAssistance Publique Hôpitaux de ParisLe Kremlin BicêtreFrance
- National Heart and Lung InstituteImperial CollegeLondonUK
| | - Paul G. Williams
- Center of Chest Diseases & Critical CareMilpark HospitalJohannesburgSouth Africa
| | | | - Evan P. Minty
- Department of Medicine & O'Brien Institute for Public HealthUniversity of CalgaryCalgaryCanada
| | - Lukas Hobohm
- Department of CardiologyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Center for Thrombosis and Hemostasis (CTH)University Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Roberto Badagliacca
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, Sapienza University of RomePoliclinico Umberto IRomeItaly
| | - Edmund M. T. Lau
- Department of Respiratory Medicine, Royal Prince Alfred HospitalUniversity of SydneyCamperdownNew South WalesAustralia
- Faculty of Medicine and HealthUniversity of SydneyCamperdownNew South WalesAustralia
| | - Zhi‐Cheng Jing
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | | | - Charles Fauvel
- Service de Cardiologie, Centre de Compétence en Hypertension Pulmonaire 27/76, Centre Hospitalier Universitaire Charles Nicolle, INSERM EnVI U1096Université de RouenRouenFrance
| | - Jose Leonidas Alves
- Pulmonary Division, Heart InstituteUniversity of São Paulo Medical SchoolSão PauloBrazil
| | - Geoff Strange
- School of MedicineThe University of Notre Dame AustraliaPerthWestern AustraliaAustralia
| | - Tomas Pulido
- Ignacio Chávez National Heart InstituteMéxico CityMexico
| | - Junyan Qian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC‐DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical ImmunologyMinistry of EducationBeijingChina
| | - Valentina Mercurio
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Jason G. E. Zelt
- Department of Medicine, Faculty of MedicineUniversity of OttawaOttawaCanada
| | - Victor M. Moles
- Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Meghan M. Cirulis
- Division of Pulmonary and Critical Care MedicineUniversity of UtahSalt Lake CityUtahUSA
- Department of Pulmonary and Critical Care MedicineIntermountain Medical Center MurraySalt Lake CityUtahUSA
| | | | - Raymond L. Benza
- Mount Sinai HeartIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - C. Gregory Elliott
- Division of Pulmonary and Critical Care MedicineUniversity of UtahSalt Lake CityUtahUSA
- Department of Pulmonary and Critical Care MedicineIntermountain Medical Center MurraySalt Lake CityUtahUSA
| |
Collapse
|
35
|
Welby S, Feng Y, Tang H, Ye C, Cohet C. A feasibility assessment of real-world data capabilities for monitoring vaccine safety and effectiveness in China: Human papillomavirus vaccination in the Yinzhou district as a use case. Pharmacoepidemiol Drug Saf 2023; 32:1131-1141. [PMID: 37228132 DOI: 10.1002/pds.5644] [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: 10/24/2022] [Revised: 04/21/2023] [Accepted: 05/23/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Real-world data (RWD) are increasingly used to generate real-world evidence (RWE) of vaccine safety and effectiveness for regulatory purposes. Assessing feasibility of using RWD sources prior to implementing observational studies is recommended. As a use case, we described the process and findings of a feasibility assessment to identify reliable and relevant data sources for monitoring the safety and effectiveness of the AS04-HPV-16/18 human papillomavirus (HPV) vaccine in China. METHODS Iterative multi-step process: (1) targeted literature review and data source mapping; (2) expert opinion from national RWD experts; (3) survey to evaluate the identified data source operational infrastructure; and (4) continuous appraisal of published studies using the identified data source. RESULTS The Yinzhou Regional Health Information Platform (YRHIP) was identified as a data source of main interest, based on its large population coverage, high cervical cancer screening rates, and availability of adult electronic immunization records. Field meetings with national RWD experts confirmed its suitability for post-authorization vaccine studies. Survey results showed that exposure data and relevant safety and effectiveness endpoints were recorded and linkable at the individual level across the platform. Iterative appraisal of emerging evidence from the literature corroborated these findings. CONCLUSIONS This feasibility assessment indicates that the YRHIP has the capacity to capture demographic, exposure, outcome and other data required to generate RWE on HPV vaccine safety and effectiveness in China. Studies using the YRHIP to monitor the AS04-HPV-16/18 vaccine in routine use building on this feasibility assessment are ongoing.
Collapse
|
36
|
Curtis LH, Sola-Morales O, Heidt J, Saunders-Hastings P, Walsh L, Casso D, Oliveria S, Mercado T, Zusterzeel R, Sobel RE, Jalbert JJ, Mastey V, Harnett J, Quek RGW. Regulatory and HTA Considerations for Development of Real-World Data Derived External Controls. Clin Pharmacol Ther 2023; 114:303-315. [PMID: 37078264 DOI: 10.1002/cpt.2913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Regulators and Health Technology Assessment (HTA) bodies are increasingly familiar with, and publishing guidance on, external controls derived from real-world data (RWD) to generate real-world evidence (RWE). We recently conducted a systematic literature review (SLR) evaluating publicly available information on the use of RWD-derived external controls to contextualize outcomes from uncontrolled trials submitted to the European Medicines Agency (EMA), the US Food and Drug Administration (FDA), and/or select HTA bodies. The review identified several key operational and methodological aspects for which more detailed guidance and alignment within and between regulatory agencies and HTA bodies is necessary. This paper builds on the SLR findings by delineating a set of key takeaways for the responsible generation of fit-for-purpose RWE. Practical methodological and operational guidelines for designing, conducting, and reporting RWD-derived external control studies are explored and discussed. These considerations include: (i) early engagement with regulators and HTA bodies during the study planning phase; (ii) consideration of the appropriateness and comparability of external controls across multiple dimensions, including eligibility criteria, temporality, population representation, and clinical evaluation; (iii) ensuring adequate sample sizes, including hypothesis testing considerations; (iv) implementation of a clear and transparent strategy for assessing and addressing data quality, including data missingness across trials and RWD; (v) selection of comparable and meaningful endpoints that are operationalized and analyzed using appropriate analytic methods; and (vi) conduct of sensitivity analyses to assess the robustness of findings in the context of uncertainty and sources of potential bias.
Collapse
Affiliation(s)
- Lesley H Curtis
- Duke Department of Population Health Sciences and Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Oriol Sola-Morales
- Fundació HiTT and Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Julien Heidt
- IQVIA, Regulatory Science and Strategy, Falls Church, Virginia, USA
| | | | - Laura Walsh
- IQVIA, Epidemiology and Drug Safety Practice, Boston, Massachusetts, USA
| | - Deborah Casso
- IQVIA, Epidemiology and Drug Safety Practice, Seattle, Washington, USA
| | - Susan Oliveria
- IQVIA, Epidemiology and Drug Safety Practice, New York, New York, USA
| | - Tiffany Mercado
- IQVIA, Regulatory Science and Strategy, Falls Church, Virginia, USA
| | | | - Rachel E Sobel
- Regeneron Pharmaceuticals Inc., Pharmacoepidemiology, Tarrytown, New York, USA
| | - Jessica J Jalbert
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| | - Vera Mastey
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| | - James Harnett
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| | - Ruben G W Quek
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| |
Collapse
|
37
|
Moon RJ, Reginster JY, Al-Daghri NM, Thiyagarajan JA, Beaudart C, Bruyère O, Burlet N, Chandran M, da Silva MC, Conaghan PG, Dere WH, Diez-Perez A, Hadji P, Halbout P, Hiligsmann M, Kanis JA, McCloskey EV, Ormarsdottir S, Prieto-Alhambra D, Radermecker RP, Rizzoli R, Al-Saleh Y, Silverman SL, Simon LS, Thomasius F, van Staa T, Laslop A, Cooper C, Harvey NC. Real-world evidence: new opportunities for osteoporosis research. Recommendations from a Working Group from the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO). Osteoporos Int 2023; 34:1283-1299. [PMID: 37351614 PMCID: PMC10382414 DOI: 10.1007/s00198-023-06827-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/28/2023] [Indexed: 06/24/2023]
Abstract
This narrative review summarises the recommendations of a Working Group of the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) for the conduct and reporting of real-world evidence studies with a focus on osteoporosis research. PURPOSE Vast amounts of data are routinely generated at every healthcare contact and activity, and there is increasing recognition that these real-world data can be analysed to generate scientific evidence. Real-world evidence (RWE) is increasingly used to delineate the natural history of disease, assess real-life drug effectiveness, understand adverse events and in health economic analysis. The aim of this work was to understand the benefits and limitations of this type of data and outline approaches to ensure that transparent and high-quality evidence is generated. METHODS A ESCEO Working Group was convened in December 2022 to discuss the applicability of RWE to osteoporosis research and approaches to best practice. RESULTS This narrative review summarises the agreed recommendations for the conduct and reporting of RWE studies with a focus on osteoporosis research. CONCLUSIONS It is imperative that research using real-world data is conducted to the highest standards with close attention to limitations and biases of these data, and with transparency at all stages of study design, data acquisition and curation, analysis and reporting to increase the trustworthiness of RWE study findings.
Collapse
Affiliation(s)
- Rebecca J Moon
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK
- Paediatric Endocrinology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jean-Yves Reginster
- WHO Collaborating Center for Epidemiology of Musculoskeletal Health and Ageing, Liège, Belgium
- Division of Epidemiology, Public Health and Health Economics, University of Liège, Liège, Belgium
| | - Nasser M Al-Daghri
- Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Charlotte Beaudart
- WHO Collaborating Center for Epidemiology of Musculoskeletal Health and Ageing, Liège, Belgium
- Division of Epidemiology, Public Health and Health Economics, University of Liège, Liège, Belgium
| | - Olivier Bruyère
- WHO Collaborating Center for Epidemiology of Musculoskeletal Health and Ageing, Liège, Belgium
- Division of Epidemiology, Public Health and Health Economics, University of Liège, Liège, Belgium
| | - Nansa Burlet
- Division of Epidemiology, Public Health and Health Economics, University of Liège, Liège, Belgium
| | - Manju Chandran
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
| | | | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Willard H Dere
- Department of Internal Medicine, Utah Center for Clinical and Translational Science, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Adolfo Diez-Perez
- Department of Internal Medicine, Hospital del Mar-IMIM, Autonomous University of Barcelona and CIBERFES, Instituto Carlos III, Barcelona, Spain
| | - Peyman Hadji
- Frankfurt Centre for Bone Health, Frankfurt, Germany
- Philipps University of Marburg, Hesse, Germany
| | | | - Mickaël Hiligsmann
- Department of Health Services Research, CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - John A Kanis
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
- Centre for Metabolic Bone Diseases, University of Sheffield, Sheffield, UK
| | - Eugene V McCloskey
- MRC Versus Arthritis Centre for Integrated Research in Musculoskeletal Ageing, University of Sheffield, Sheffield, UK
- Mellanby Centre for Musculoskeletal Research, Department of Oncology & Metabolism, University of Sheffield, Sheffield, UK
| | | | - Daniel Prieto-Alhambra
- Pharmaco- and Device Epidemiology, Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Régis P Radermecker
- Department of Clinical Pharmacology, Diabetes, Nutrition and Metabolic Disorders, CHU Liege, Liege, Belgium
| | - René Rizzoli
- Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Yousef Al-Saleh
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | | | | | | | - Tjeerd van Staa
- Centre for Health Informatics, University of Manchester, Manchester, UK
| | - Andrea Laslop
- Scientific Office, Austrian Medicines and Medical Devices Agency, Vienna, Austria
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
- National Institute for Health Research (NIHR) Musculoskeletal Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK.
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| |
Collapse
|
38
|
Sola-Morales O, Curtis LH, Heidt J, Walsh L, Casso D, Oliveria S, Saunders-Hastings P, Song Y, Mercado T, Zusterzeel R, Mastey V, Harnett J, Quek RGW. Effectively Leveraging RWD for External Controls: A Systematic Literature Review of Regulatory and HTA Decisions. Clin Pharmacol Ther 2023; 114:325-355. [PMID: 37079433 DOI: 10.1002/cpt.2914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/14/2023] [Indexed: 04/21/2023]
Abstract
Real-world data (RWD)-derived external controls can be used to contextualize efficacy findings for investigational therapies evaluated in uncontrolled trials. As the number of submissions to regulatory and health technology assessment (HTA) bodies using external controls rises, and in light of recent regulatory and HTA guidance on the appropriate use of RWD, there is a need to address the operational and methodological challenges impeding the quality of real-world evidence (RWE) generation and the consistency in evaluation of RWE across agencies. This systematic review summarizes publicly available information on the use of external controls to contextualize outcomes from uncontrolled trials for all indications from January 1, 2015, through August 20, 2021, that were submitted to the European Medicines Agency, the US Food and Drug Administration, and/or select major HTA bodies (National Institute for Health and Care Excellence (NICE), Haute Autorité de Santé (HAS), Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen (IQWiG), and Gemeinsamer Bundesausschuss (G-BA)). By systematically reviewing submissions to regulatory and HTA bodies in the context of recent guidance, this study provides quantitative and qualitative insights into how external control design and analytic choices may be viewed by different agencies in practice. The primary operational and methodological aspects identified for discussion include, but are not limited to, engagement of regulators and HTA bodies, approaches to handling missing data (a component of data quality), and selection of real-world endpoints. Continued collaboration and guidance to address these and other aspects will inform and assist stakeholders attempting to generate evidence using external controls.
Collapse
Affiliation(s)
- Oriol Sola-Morales
- Fundació Health Innovation Technology Transfer and International, University of Catalonia, Barcelona, Spain
| | - Lesley H Curtis
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Julien Heidt
- IQVIA, Regulatory Science and Strategy, Falls Church, Virginia, USA
| | - Laura Walsh
- IQVIA, Epidemiology and Drug Safety Practice, Boston, Massachusetts, USA
| | - Deborah Casso
- IQVIA, Epidemiology and Drug Safety Practice, Seattle, Washington, USA
| | - Susan Oliveria
- IQVIA, Epidemiology and Drug Safety Practice, New York, New York, USA
| | | | - Yufei Song
- IQVIA, Epidemiology and Drug Safety Practice, Beijing, China
| | - Tiffany Mercado
- IQVIA, Regulatory Science and Strategy, Falls Church, Virginia, USA
| | | | - Vera Mastey
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| | - James Harnett
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| | - Ruben G W Quek
- Regeneron Pharmaceuticals Inc., Health Economics & Outcomes Research, Tarrytown, New York, USA
| |
Collapse
|
39
|
Graili P, Guertin JR, Chan KKW, Tadrous M. Integration of real-world evidence from different data sources in health technology assessment. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2023; 26:11460. [PMID: 37529633 PMCID: PMC10387532 DOI: 10.3389/jpps.2023.11460] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023]
Abstract
Real-world evidence (RWE) is being increasingly used by a wide range of stakeholders involved in the therapeutic product lifecycle but remains underutilized in the health technology assessment (HTA) process. RWE aims to fill the current evidence gaps, reduce the uncertainty around the benefits of medical technologies, and better understand the long-term impact of health technologies in real-world conditions. Despite the minimal use of RWE in some elements of HTA, there has been a larger push to further utilize RWE in the HTA processes. HTA bodies, as other stakeholders, work towards developing more robust means to leverage RWE from various data sources in the HTA processes. However, these agencies need to overcome important challenges before the broader incorporation of RWE into their routine practice. This paper aims to explore the extensive integration of RWE utilizing diverse sources of RWD. We discuss the utilization of RWE in HTA processes, considering aspects such as when, where, and how RWE can be effectively applied. Additionally, we seek the potential challenges and barriers associated with the utilization of different data sources.
Collapse
Affiliation(s)
- Pooyeh Graili
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
- Quality HTA, Oakville, ON, Canada
| | - Jason R. Guertin
- Axe Santé des Populations et Pratiques Optimales en Santé, Centre de Recherche du CHU de Québec-Université Laval, Laval, QC, Canada
- Department of Social and Preventive Medicine, Université Laval, Laval, QC, Canada
- Tissue Engineering Laboratory (LOEX), Université Laval, Laval, QC, Canada
| | - Kelvin K. W. Chan
- Sunnybrook Health Science Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada
| | - Mina Tadrous
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
- Women’s College Research Institute, Women’s College Hospital, Toronto, ON, Canada
| |
Collapse
|
40
|
Vandenbussche N, Pisarek K, Paemeleire K. Methodological considerations on real-world evidence studies of monoclonal antibodies against the CGRP-pathway for migraine: a systematic review. J Headache Pain 2023; 24:75. [PMID: 37344811 DOI: 10.1186/s10194-023-01611-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/10/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Real-world data are accumulating on the effectiveness, tolerability and safety of anti-calcitonin gene-related peptide pathway monoclonal antibodies for the preventive treatment of migraine. We performed a systematic review of the methodology of prospective, observational, clinic-based real-world evidence studies with these drugs in both episodic and chronic migraine. METHODS The objectives were to evaluate the definitions and reported outcomes used, and to perform a risk of bias assessment for each of the different studies. PubMed and EMBASE were systematically queried for relevant scientific articles. Study quality assessment of the included studies was conducted using the "National Heart, Lung and Blood Institute (NHLBI) Study Quality Assessment Tool for Before-After (Pre-Post) Studies with No Control Group". RESULTS Forty-six studies fitted the criteria for the systematic review and were included in the analysis. Ten studies (21.7%) defined a migraine day for the study, while only 5 studies defined a headache day for the study (10.9%). The most common primary endpoint/objective of the studies was change in monthly migraine days (n = 16, 34.8%), followed by responder rate (n = 15, 32.6%) and change in monthly headache days (n = 5, 10.9%). Eight studies (17.4%) did not define the primary endpoint/objective. Thirty-three studies were graded as "good" quality and 13 studies were graded as "fair". CONCLUSION Our analysis shows rather significant heterogeneity and/or lack of predefined primary outcomes/objectives, definitions of outcomes measures and the use of longitudinal monitoring (e.g. headache diaries). Standardization of terminology, definitions and protocol procedures for real-world evidence studies of preventive treatments for migraine are recommended. TRIAL REGISTRATION This study was registered with PROSPERO with ID CRD42022369366.
Collapse
Affiliation(s)
- Nicolas Vandenbussche
- Department of Neurology, Ghent University Hospital, Corneel Heymanslaan 10, B-9000, Ghent, Belgium.
- Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, B-9000, Ghent, Belgium.
| | - Karolina Pisarek
- Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, B-9000, Ghent, Belgium
| | - Koen Paemeleire
- Department of Neurology, Ghent University Hospital, Corneel Heymanslaan 10, B-9000, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, B-9000, Ghent, Belgium
| |
Collapse
|
41
|
Saesen R, Van Hemelrijck M, Bogaerts J, Booth CM, Cornelissen JJ, Dekker A, Eisenhauer EA, Freitas A, Gronchi A, Hernán MA, Hulstaert F, Ost P, Szturz P, Verkooijen HM, Weller M, Wilson R, Lacombe D, van der Graaf WT. Defining the role of real-world data in cancer clinical research: The position of the European Organisation for Research and Treatment of Cancer. Eur J Cancer 2023; 186:52-61. [PMID: 37030077 DOI: 10.1016/j.ejca.2023.03.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023]
Abstract
The emergence of the precision medicine paradigm in oncology has led to increasing interest in the integration of real-world data (RWD) into cancer clinical research. As sources of real-world evidence (RWE), such data could potentially help address the uncertainties that surround the adoption of novel anticancer therapies into the clinic following their investigation in clinical trials. At present, RWE-generating studies which investigate antitumour interventions seem to primarily focus on collecting and analysing observational RWD, typically forgoing the use of randomisation despite its methodological benefits. This is appropriate in situations where randomised controlled trials (RCTs) are not feasible and non-randomised RWD analyses can offer valuable insights. Nevertheless, depending on how they are designed, RCTs have the potential to produce strong and actionable RWE themselves. The choice of which methodology to employ for RWD studies should be guided by the nature of the research question they are intended to answer. Here, we attempt to define some of the questions that do not necessarily require the conduct of RCTs. Moreover, we outline the strategy of the European Organisation for Research and Treatment of Cancer (EORTC) to contribute to the generation of robust and high-quality RWE by prioritising the execution of pragmatic trials and studies set up according to the trials-within-cohorts approach. If treatment allocation cannot be left up to random chance due to practical or ethical concerns, the EORTC will consider undertaking observational RWD research based on the target trial principle. New EORTC-sponsored RCTs may also feature concurrent prospective cohorts composed of off-trial patients.
Collapse
|
42
|
Oechsle AL, Wiechers C, Abadie V, Abel F, Breugem C, Poets CF. Study protocol for a multicenter, multinational, observational registry of epidemiology, treatment and outcome of patients with Robin sequence. Head Face Med 2023; 19:20. [PMID: 37210548 DOI: 10.1186/s13005-023-00364-3] [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: 12/19/2022] [Accepted: 05/08/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Robin sequence (RS) is a congenital condition characterized by micrognathia, glossoptosis and upper airway obstruction. Diagnosis and treatment are characterized by heterogeneity, resulting in a lack of uniformly collected data. METHODS We have set up a prospective, observational, multicenter, multinational registry aimed at obtaining routine clinical data from RS patients receiving different treatment approaches and enabling an assessment of outcomes obtained through different therapeutic approaches. Patient enrolment has started in January 2022. Disease characteristics, adverse events and complications depending on the different diagnostic and treatment approaches and their effects on neurocognition, growth, speech development and hearing outcome are evaluated using routine clinical data. In addition to characterizing the patient population and comparing outcomes achieved with different treatment approaches, the registry will evolve to focus on endpoints such as quality of life and long-term developmental status. DISCUSSION This registry will provide data on different treatment approaches collected during routine care with diverse framework conditions and will allow assessing diagnostic and therapeutic outcomes of children with RS. These data, urgently demanded by the scientific community, may contribute to refining and personalizing existing therapeutic approaches and increase knowledge about the long-term outcome of children born with this rare condition. TRIAL REGISTRATION DRKS00025365.
Collapse
Affiliation(s)
- Anna-Lisa Oechsle
- Interdisciplinary Center for Cleft Palate & Craniofacial Malformations, Department of Neonatology, Tübingen University Hospital, Calwerstr. 7, 72076, Tübingen, Germany
| | - Cornelia Wiechers
- Interdisciplinary Center for Cleft Palate & Craniofacial Malformations, Department of Neonatology, Tübingen University Hospital, Calwerstr. 7, 72076, Tübingen, Germany
| | - Veronique Abadie
- Department of General Pediatrics, Reference Centre for Rare Diseases 'Pierre Robin sequences and congenital sucking-swallowing troubles', Necker University Hospital, Paris University, &, France
| | - Francois Abel
- Department of Pediatric Respiratory Medicine, Great Ormond Street Hospital, London, UK
| | - Corstiaan Breugem
- Department of Plastic Reconstructive and Hand Surgery, Emma Children's Hospital - Location AMC, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Christian F Poets
- Interdisciplinary Center for Cleft Palate & Craniofacial Malformations, Department of Neonatology, Tübingen University Hospital, Calwerstr. 7, 72076, Tübingen, Germany.
| |
Collapse
|
43
|
Eskola SM, Leufkens HGM, Bate A, Bruin MLD, Gardarsdottir H. The Role of Real-World Data and Evidence in Oncology Medicines Approved in EU in 2018-2019. J Cancer Policy 2023; 36:100424. [PMID: 37116794 DOI: 10.1016/j.jcpo.2023.100424] [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: 02/10/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023]
Abstract
Use of Real-World data (RWD) has gained the interest of different stakeholders in cancer care. The aim of this study was to identify and describe the use of RWD/RWE during the pre-authorisation phase of products authorised by the EMA in 2018 and 2019 (n=111), with the focus on oncology medicines (n=24). Information was extracted from the European Public Assessment Report (EPAR) summaries and recorded for 5 stages (11 categories) of the drug development lifecycle (discovery, early development, clinical development, registration/market launch, lifecycle management). Specific chapters of full EPAR were reviewed to substantiate the findings on RWD/RWE use in clinical trial design, efficacy, safety, and effectiveness evaluation. RWD/RWE is present in all stages of the oncology drug development; 100.0% in discovery, 37.5% early development, 58.3% in clinical development, 62.5% in registration decision and 100.0% in post-authorization lifecycle management. Examples showed that trial design supported by RWD/RWE included use of open label/single arm studies; efficacy was about using either comparison of results to historical controls, supplying survey data obtained outside the clinical trial or utilizing expert panel advice; safety about including literature findings in evidence; and effectiveness on comparison of trial results of the given product to historical data or existing standard of care. The findings of this study provide specific insights into how RWD/RWE is used in development of cancer therapeutics, how it contributes to regulatory decision making and can guide further policy developments in this field.
Collapse
Affiliation(s)
- Sini M Eskola
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands; European Federation of Pharmaceutical Industries and Associations, Brussels, Belgium
| | - Hubertus G M Leufkens
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Andrew Bate
- Global Safety, GSK, Brentford, Middlesex, UK; Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Marie Louise De Bruin
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Helga Gardarsdottir
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Reykjavík, Iceland.
| |
Collapse
|
44
|
Wieseler B, Neyt M, Kaiser T, Hulstaert F, Windeler J. Replacing RCTs with real world data for regulatory decision making: a self-fulfilling prophecy? BMJ 2023; 380:e073100. [PMID: 36863730 DOI: 10.1136/bmj-2022-073100] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Affiliation(s)
- Beate Wieseler
- Institute for Quality and Efficiency in Health Care (IQWiG), Cologne, Germany
| | - Mattias Neyt
- Belgian Health Care Knowledge Centre (KCE), Brussels, Belgium
| | - Thomas Kaiser
- Institute for Quality and Efficiency in Health Care (IQWiG), Cologne, Germany
| | - Frank Hulstaert
- Belgian Health Care Knowledge Centre (KCE), Brussels, Belgium
| | - Jürgen Windeler
- Institute for Quality and Efficiency in Health Care (IQWiG), Cologne, Germany
| |
Collapse
|
45
|
Real-world-Daten in der Arzneimittelregulation – aktuelle Entwicklungen und Ausblick. PRÄVENTION UND GESUNDHEITSFÖRDERUNG 2023. [DOI: 10.1007/s11553-022-01010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Zusammenfassung
Hintergrund
Real-world-Daten (RWD) und die aus ihnen durch statistisch-epidemiologische Analysen abgeleitete Real-world-Evidenz (RWE) spielen eine vielversprechende und zunehmend relevante Rolle bei regulatorischen Entscheidungsfindungen entlang des Produktlebenszyklus von Arzneimitteln.
Ziel der Arbeit
Es wird ein Überblick über den aktuellen Stand, die Entwicklungspotenziale und Initiativen zur verstärkten Nutzung von RWE in der europäischen und internationalen Arzneimittelregulation gegeben.
Material und Methoden
Die Grundlagen für die Übersichtsarbeit sind Originalarbeiten und Reviews aus der aktuellen internationalen Literatur (inklusive eigener Forschungsergebnisse), aktuelle Beispiele aus der regulatorischen Praxis sowie die Einbindung in europäische und internationale Initiativen zur verstärkten Nutzung von RWD/RWE in regulatorischen Entscheidungsprozessen.
Ergebnisse
Aktuell primär zur supportiven Evidenz bei regulatorischen Entscheidungsfindungen wird RWE aus RWD eingesetzt. Neben dem etablierten Einsatz in Phasen nach der Zulassung (z. B. Überwachung der Arzneimittelsicherheit), werden RWD zunehmend auch in der Phase vor der Zulassung und in der Evaluation eingesetzt. Aktuell wird durch verstärkte Vernetzung der Datenquellen auf nationaler und internationaler Ebene eine Gesundheitsdateninfrastruktur aufgebaut, um neue Möglichkeiten zur RWD-Nutzung zu schaffen.
Schlussfolgerung
Neben einer wachsenden Bedeutung von RWD/RWE in der europäischen und internationalen Arzneimittelregulation ergeben sich auch neue Herausforderungen zum Zugang zu und zur Analyse von RWD. Die Variabilität und Heterogenität der RWD-Quellen machen die Entwicklung neuer und optimierter Methoden für RWD-Analysen unerlässlich. Auch neue Leitfaden- und Schulungskonzepte für die beteiligten Stakeholder sind essenziell.
Collapse
|
46
|
Crisafulli S, Khan Z, Karatas Y, Tuccori M, Trifirò G. An overview of methodological flaws of real-world studies investigating drug safety in the post-marketing setting. Expert Opin Drug Saf 2023; 22:373-380. [PMID: 37243676 DOI: 10.1080/14740338.2023.2219892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/07/2023] [Accepted: 05/26/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION The evaluation of the post-marketing safety profile of drugs is a continuous monitoring process for approved and marketed medicines and it is crucial for detecting new adverse drug reactions. As such, real-world studies are essential to complement pre-marketing evidence with information concerning drug risk-benefit profile and use in wider patient populations and they have a great potential to support post-marketing drug safety evaluations. AREAS COVERED A detailed description of the main limitations of real-world data sources (i.e. claims databases, electronic healthcare records, drug/disease registers and spontaneous reporting system databases) and of the main methodological challenges of real-world studies in generating real-world evidence is provided. EXPERT OPINION Real-world evidence biases can be ascribed to both the methodological approach and the specific limitations of the different real-world data sources used to carry out the study. As such, it is crucial to characterize the quality of real-world data, by establishing guidelines and best practices for the assessment of data fitness for purpose. On the other hand, it is important that real-world studies are conducted using a rigorous methodology, aimed at minimizing the risk of bias.
Collapse
Affiliation(s)
| | - Zakir Khan
- Faculty of Medicines, Department of Medical Pharmacology Çukurova University, Sarıçam, Adana, Türkiye
| | - Yusuf Karatas
- Faculty of Medicines, Department of Medical Pharmacology Çukurova University, Sarıçam, Adana, Türkiye
- Pharmacovigilance Specialist, Faculty of Medicines, Balcali Hospital, Sarıçam, Adana, Türkiye
| | - Marco Tuccori
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Unit of Adverse Drug Reaction Monitoring, University Hospital of Pisa, Pisa, Italy
| | - Gianluca Trifirò
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| |
Collapse
|
47
|
Tan GSQ, Sloan EK, Lambert P, Kirkpatrick CMJ, Ilomäki J. Drug repurposing using real-world data. Drug Discov Today 2023; 28:103422. [PMID: 36341896 DOI: 10.1016/j.drudis.2022.103422] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/18/2022] [Accepted: 10/25/2022] [Indexed: 02/02/2023]
Abstract
The use of real-world data in drug repurposing has emerged due to well-established advantages of drug repurposing in supplementing de novo drug discovery and incentives in incorporating real-world evidence in regulatory approvals. We conducted a scoping review to characterize repurposing studies using real-world data and discuss their potential challenges and solutions. A total of 250 studies met the inclusion criteria, of which 36 were original studies on hypothesis generation, 101 on hypothesis validation, and seven on safety assessment. Key challenges that should be addressed for future progress in using real-world data for repurposing include isolated data sources with poor clinical granularity, false-positive signals from data mining, the sensitivity of hypothesis validation to bias and confounding, and the lack of clear regulatory guidance.
Collapse
Affiliation(s)
- George S Q Tan
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Pete Lambert
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Carl M J Kirkpatrick
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia.
| | - Jenni Ilomäki
- Centre for Medicine Use and Safety, Monash University, Parkville, Victoria, Australia.
| |
Collapse
|
48
|
Bakker E, Plueschke K, Jonker CJ, Kurz X, Starokozhko V, Mol PGM. Contribution of Real-World Evidence in European Medicines Agency's Regulatory Decision Making. Clin Pharmacol Ther 2023; 113:135-151. [PMID: 36254408 PMCID: PMC10099093 DOI: 10.1002/cpt.2766] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/10/2022] [Indexed: 12/31/2022]
Abstract
Real-world data/evidence (RWD/RWE) may provide insightful information on medicines' clinical effects to guide regulatory decisions. While its contribution has been recognized for safety monitoring and disease epidemiology across medicines' life cycles, using RWD/RWE to demonstrate efficacy requires further evaluation. This study aimed to (i) characterize RWD/RWE presented by applicants to support claims on medicines' efficacy within initial marketing authorization applications (MAAs) and extension of indication applications (EoIs), and (ii) analyze the contribution of RWD/RWE to regulatory decisions on medicines' benefit-risk profile. RWD/RWE was included to support efficacy in 32 MAAs and 14 EoIs submitted 2018-2019. Of these, RWD/RWE was part of the preauthorization package of 16 MAAs and 10 EoIs, and was (i) considered supporting the regulatory decision in 10 applications (five MAAs, five EoIs), (ii) considered not supporting the regulatory decision in 11 (seven MAAs, four EoIs), and (iii) not addressed at all in the evaluation of 5 applications (four MAAs, one EoI). Common limitations of submitted RWD/RWE included missing data, lack of representativeness of populations, small sample size, absence of an adequate or prespecified analysis plan, and risk of several types of bias. The suitability of RWD/RWE in a given application still requires a case-by-case analysis considering its purpose of use, implying reflection on the data source, together with its assets and limitations, study objectives and designs, and the overall data package issued. Early interactions and continuous dialogues with regulators and relevant stakeholders is key to optimize fit-for-purpose RWE generation, enabling its broader use in medicines development.
Collapse
Affiliation(s)
- Elisabeth Bakker
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Kelly Plueschke
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands
| | - Carla J Jonker
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands.,Dutch Medicines Evaluation Board (MEB), Utrecht, The Netherlands
| | - Xavier Kurz
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands
| | - Viktoriia Starokozhko
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.,Dutch Medicines Evaluation Board (MEB), Utrecht, The Netherlands
| | - Peter G M Mol
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.,Dutch Medicines Evaluation Board (MEB), Utrecht, The Netherlands.,Scientific Advice Working Party of the European Medicines Agency, Amsterdam, The Netherlands
| |
Collapse
|
49
|
Dal-Ré R, Porcher R, Rosendaal FR, Schwarzer-Daum B. Regulatory agencies disregard real-world effectiveness evidence on product labels beyond what is reasonable. J Clin Epidemiol 2023; 153:83-90. [PMID: 36371045 DOI: 10.1016/j.jclinepi.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/19/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Rafael Dal-Ré
- Epidemiology Unit, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Raphaël Porcher
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS-UMR1153), Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
50
|
O’Sullivan JM, McKay RR, Rahbar K, Fizazi K, George DJ, Tombal B, Schmall A, Sandström P, Verholen F, Shore N. Real-world effectiveness, long-term safety and treatment pathway integration of radium-223 therapy in patients with metastatic castration-resistant prostate cancer. Front Med (Lausanne) 2022; 9:fmed-09-1070392. [PMID: 36619649 PMCID: PMC9812947 DOI: 10.3389/fmed.2022.1070392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Radium-223 dichloride (223Ra) is an α-emitter approved for the treatment of metastatic castration-resistant prostate cancer (mCRPC) with bone metastases, but without visceral involvement. Despite being a life-prolonging therapy (LPT), 223Ra remains underutilized. A large body of real-world evidence (RWE) for 223Ra has been published in the decade since the pivotal phase 3 ALSYMPCA study, a period during which the treatment landscape has continued to evolve. How to optimize 223Ra use, including how to integrate it into the mCRPC management pathway amongst other current LPTs (i.e., with respect to timing and concurrent, layered, or sequential use), is therefore of considerable interest. RWE studies lack the conventional restraints of clinical trials and can therefore help to build an understanding of how treatments may be best used in routine practice. Here we review RWE studies investigating the efficacy and safety of 223Ra in mCRPC [including in sequence with the recently approved 177-Lutetium conjugated to the ligand prostate-specific membrane antigen (177Lu-PSMA)], as well as response marker development, imaging techniques, and current clinical practice recommendations.
Collapse
Affiliation(s)
- Joe M. O’Sullivan
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast and Northern Ireland Cancer Centre, Belfast, United Kingdom
| | - Rana R. McKay
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University of Münster Medical Center, Münster, Germany
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris-Saclay, Villejuif, France
| | | | - Bertrand Tombal
- Division of Urology, Institut de Recherche Clinique (IREC), Cliniques Universitaires Saint Luc, Brussels, Belgium
| | | | - Per Sandström
- Bayer HealthCare Pharmaceuticals, Whippany, NJ, United States
| | | | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, United States
| |
Collapse
|