1
|
Mongelli M, De Angelis B, delle Cave V, Greco G, De Arcangelis A, Bernagozzi A, Salvemini C, Calabrese M, Christille JM, Cavalli A, Gustincich S, Monaci MG. The Public Knowledge of Precision Medicine and Genomic Research: A Survey in the Aosta Valley. J Pers Med 2025; 15:80. [PMID: 40137396 PMCID: PMC11943031 DOI: 10.3390/jpm15030080] [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: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Precision medicine (PM) considers the genetic variability of individuals to identify tailored diagnosis and treatments. It relies on the possibility of gathering the widest possible health data and genetic information from individuals to obtain a broad pool of comparative data. To achieve this goal, the Region of Valle d'Aosta, since 2019, has co-financed the research center CMP3VdA, aiming to sequence 5000 genomes of patients with neurodevelopmental, neurodegenerative, oncological, and organ transplantation diseases, and to investigate the genetic variability of the resident population. Methods: This paper presents the results of an online survey of 472 (328F) respondents regarding willingness to participate in the genomic project and awareness, attitudes, and concerns about PM. Results: The main results show that the vast majority (92.6%) would be willing to participate-a higher percentage than in previous studies. Age, education, and prior experience in the healthcare sector are significant factors influencing the awareness of PM. Additionally, subgroups organized by age, gender, and religiosity show significant differences with respect to participants' reasons for participating in research and which types of biological samples they would be willing to donate. Conclusions: Our findings can serve as a guide for stakeholders-particularly policymakers-to target institutional communication and achieve maximum participation in genomic research projects.
Collapse
Affiliation(s)
- Matteo Mongelli
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Biagio De Angelis
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Valeria delle Cave
- Communication and External Relations Directorate, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy (G.G.)
| | - Giuliano Greco
- Communication and External Relations Directorate, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy (G.G.)
| | - Arianna De Arcangelis
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Andrea Bernagozzi
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Chiara Salvemini
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Matteo Calabrese
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Jean Marc Christille
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Andrea Cavalli
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Computational and Chemical Biology, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Stefano Gustincich
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Center for Human Technologies, Non-Coding RNAs and RNA-Based Therapeutics, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152 Genova, Italy
| | - Maria Grazia Monaci
- Department of Human and Social Science, University of Valle d’Aosta, Strada Cappuccini 2A, 11100 Aosta, Italy
| |
Collapse
|
2
|
Jacobs JJL, Beekers I, Verkouter I, Richards LB, Vegelien A, Bloemsma LD, Bongaerts VAMC, Cloos J, Erkens F, Gradowska P, Hort S, Hudecek M, Juan M, Maitland-van der Zee AH, Navarro-Velázquez S, Ngai LL, Rafiq QA, Sanges C, Tettero J, van Os HJA, Vos RC, de Wit Y, van Dijk S. A data management system for precision medicine. PLOS DIGITAL HEALTH 2025; 4:e0000464. [PMID: 39787064 PMCID: PMC11717228 DOI: 10.1371/journal.pdig.0000464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/27/2024] [Indexed: 01/12/2025]
Abstract
Precision, or personalised medicine has advanced requirements for medical data management systems (MedDMSs). MedDMS for precision medicine should be able to process hundreds of parameters from multiple sites, be adaptable while remaining in sync at multiple locations, real-time syncing to analytics and be compliant with international privacy legislation. This paper describes the LogiqSuite software solution, aimed to support a precision medicine solution at the patient care (LogiqCare), research (LogiqScience) and data science (LogiqAnalytics) level. LogiqSuite is certified and compliant with international medical data and privacy legislations. This paper evaluates a MedDMS in five types of use cases for precision medicine, ranging from data collection to algorithm development and from implementation to integration with real-world data. The MedDMS is evaluated in seven precision medicine data science projects in prehospital triage, cardiovascular disease, pulmonology, and oncology. The P4O2 consortium uses the MedDMS as an electronic case report form (eCRF) that allows real-time data management and analytics in long covid and pulmonary diseases. In an acute myeloid leukaemia, study data from different sources were integrated to facilitate easy descriptive analytics for various research questions. In the AIDPATH project, LogiqCare is used to process patient data, while LogiqScience is used for pseudonymous CAR-T cell production for cancer treatment. In both these oncological projects the data in LogiqAnalytics is also used to facilitate machine learning to develop new prediction models for clinical-decision support (CDS). The MedDMS is also evaluated for real-time recording of CDS data from U-Prevent for cardiovascular risk management and from the Stroke Triage App for prehospital triage. The MedDMS is discussed in relation to other solutions for privacy-by-design, integrated data stewardship and real-time data analytics in precision medicine. LogiqSuite is used for multi-centre research study data registrations and monitoring, data analytics in interdisciplinary consortia, design of new machine learning / artificial intelligence (AI) algorithms, development of new or updated prediction models, integration of care with advanced therapy production, and real-world data monitoring in using CDS tools. The integrated MedDMS application supports data management for care and research in precision medicine.
Collapse
Affiliation(s)
| | - Inés Beekers
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| | - Inge Verkouter
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| | - Levi B. Richards
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| | - Alexandra Vegelien
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
- Faculty of Mathematics, VU, Amsterdam, The Netherlands
| | | | - Vera A. M. C. Bongaerts
- Public Health & Primary Care, and Health Campus The Hague, Leiden University Medical Center, The Hague, The Netherlands
| | | | - Frederik Erkens
- Department Production Metrology, Fraunhofer Institute for Production Technology IPT, Aachen, Germany
| | - Patrycja Gradowska
- HOVON Foundation, Rotterdam, The Netherlands; Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Simon Hort
- Adaptive Produktionssteuerung, Fraunhofer Institute for Production Technology IPT, Aachen, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, University Clinic Würzburg, Würzburg, Germany
| | - Manel Juan
- Fundació Clínic per a la Recerca Biomèdica—Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Immunology department, Hospital Clinic of Barcelona, Barcelona, Spain
- HSJD-Clinic Immunotherapy platform, Barcelona, Spain
| | | | - Sergio Navarro-Velázquez
- Fundació Clínic per a la Recerca Biomèdica—Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Immunology department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Lok Lam Ngai
- Department of Haematology, Amsterdam UMC, The Netherlands
| | - Qasim A. Rafiq
- Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Carmen Sanges
- Medizinische Klinik und Poliklinik II, University Clinic Würzburg, Würzburg, Germany
| | - Jesse Tettero
- Department of Haematology, Amsterdam UMC, The Netherlands
| | - Hendrikus J. A. van Os
- Public Health & Primary Care, and Health Campus The Hague, Leiden University Medical Center, The Hague, The Netherlands
- National eHealth Living Lab, Leiden, The Netherlands
| | - Rimke C. Vos
- Public Health & Primary Care, and Health Campus The Hague, Leiden University Medical Center, The Hague, The Netherlands
| | - Yolanda de Wit
- Department of Pulmonary Medicine, Amsterdam UMC, The Netherlands
| | - Steven van Dijk
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| |
Collapse
|
3
|
Vu M, Degeling K, Westerman D, IJzerman MJ. Scenario analysis and multi-criteria decision analysis to explore alternative reimbursement pathways for whole genome sequencing for blood cancer patients. J Cancer Policy 2024; 41:100501. [PMID: 39142605 DOI: 10.1016/j.jcpo.2024.100501] [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: 01/21/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Whole genome sequencing (WGS) has transformative potential for blood cancer management, but reimbursement is hindered by uncertain benefits relative to added costs. This study employed scenario planning and multi-criteria decision analysis (MCDA) to evaluate stakeholders' preferences for alternative reimbursement pathways, informing future health technology assessment (HTA) submission of WGS in blood cancer. METHODS Key factors influencing WGS reimbursement in blood cancers were identified through a literature search. Hypothetical scenarios describing various evidential characteristics of WGS for HTA were developed using the morphological approach. An online survey, incorporating MCDA weights, was designed to gather stakeholder preferences (consumers/patients, clinicians/health professionals, industry representatives, health economists, and HTA committee members) for these scenarios. The survey assessed participants' approval of WGS reimbursement for each scenario, and scenario preferences were determined using the geometric mean method, applying an algorithm to improve reliability and precision by addressing inconsistent responses. RESULTS Nineteen participants provided complete survey responses, primarily clinicians or health professionals (n = 6; 32 %), consumers/patients and industry representatives (both at n = 5; 26 %). "Clinical impact of WGS results on patient care" was the most critical criterion (criteria weight of 0.25), followed by "diagnostic accuracy of WGS" (0.21), "cost-effectiveness of WGS" (0.19), "availability of reimbursed treatment after WGS" (0.16), and "eligibility criteria for reimbursed treatment based on actionable WGS results" and "cost comparison of WGS" (both at 0.09). Participants preferred a scenario with substantial clinical evidence, high access to reimbursed targeted treatment, cost-effectiveness below $50,000 per quality-adjusted life year (QALY) gained, and affordability relative to standard molecular tests. Reimbursement was initially opposed until criteria such as equal cost to standard tests and better treatment accessibility were met. CONCLUSION Payers commonly emphasize acceptable cost-effectiveness, but strong clinical evidence for many variants and comparable costs to standard tests are likely to drive positive reimbursement decisions for WGS.
Collapse
Affiliation(s)
- Martin Vu
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Koen Degeling
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - David Westerman
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia
| | - Maarten J IJzerman
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Erasmus School of Health Policy and Management, Rotterdam, the Netherlands.
| |
Collapse
|
4
|
Chen W, Wang Y, Zemlyanska Y, Butani D, Wong NCB, Virabhak S, Matchar DB, Teerawattananon Y. Evaluating the Value for Money of Precision Medicine from Early Cycle to Market Access: A Comprehensive Review of Approaches and Challenges. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1425-1434. [PMID: 37187236 DOI: 10.1016/j.jval.2023.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 04/05/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVES This study aimed to perform a comprehensive review of modeling approaches and methodological and policy challenges in the economic evaluation (EE) of precision medicine (PM) across clinical stages. METHODS First, a systematic review was performed to assess the approaches of EEs in the past 10 years. Next, a targeted review of methodological articles was conducted for methodological and policy challenges in performing EEs of PM. All findings were synthesized into a structured framework that focused on patient population, Intervention, Comparator, Outcome, Time, Equity and ethics, Adaptability and Modeling aspects, named the "PICOTEAM" framework. Finally, a stakeholder consultation was conducted to understand the major determinants of decision making in PM investment. RESULTS In 39 methodological articles, we identified major challenges to the EE of PM. These challenges include that PM applications involve complex and evolving clinical decision space, that clinical evidence is sparse because of small subgroups and complex pathways in PM settings, a one-time PM application may have lifetime or intergenerational impacts but long-term evidence is often unavailable, and that equity and ethics concerns are exceptional. In 275 EEs of PM, current approaches did not sufficiently capture the value of PM compared with targeted therapies, nor did they differentiate Early EEs from Conventional EEs. Finally, policy makers perceived the budget impact, cost savings, and cost-effectiveness of PM as the most important determinants in decision making. CONCLUSIONS There is an urgent need to modify existing guidelines or develop a new reference case that fits into the new healthcare paradigm of PM to guide decision making in research and development and market access.
Collapse
Affiliation(s)
- Wenjia Chen
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore.
| | - Yi Wang
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Yaroslava Zemlyanska
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Dimple Butani
- Health Intervention and Technology Assessment Program (HITAP), Ministry of Public Health, Thailand
| | | | | | - David Bruce Matchar
- Precision Health Research (PRECISE), Singapore; Health Services and Systems Research, Duke-NUS Medical School, Singapore; Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Yot Teerawattananon
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore; Health Intervention and Technology Assessment Program (HITAP), Ministry of Public Health, Thailand
| |
Collapse
|
5
|
Pollard S, Weymann D, Chan B, Ehman M, Wordsworth S, Buchanan J, Hanna TP, Ho C, Lim HJ, Lorgelly PK, Raymakers AJN, McCabe C, Regier DA. Defining a Core Data Set for the Economic Evaluation of Precision Oncology. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2022; 25:1371-1380. [PMID: 35216902 DOI: 10.1016/j.jval.2022.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/11/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVES Precision oncology is generating vast amounts of multiomic data to improve human health and accelerate research. Existing clinical study designs and attendant data are unable to provide comparative evidence for economic evaluations. This lack of evidence can cause inconsistent and inappropriate reimbursement. Our study defines a core data set to facilitate economic evaluations of precision oncology. METHODS We conducted a literature review of economic evaluations of next-generation sequencing technologies, a common application of precision oncology, published between 2005 and 2018 and indexed in PubMed (MEDLINE). Based on this review, we developed a preliminary core data set for informal expert feedback. We then used a modified-Delphi approach with individuals involved in implementation and evaluation of precision medicine, including 2 survey rounds followed by a final voting conference to refine the data set. RESULTS Two authors determined that variation in published data elements was reached after abstraction of 20 economic evaluations. Expert consultation refined the data set to 83 unique data elements, and a multidisciplinary sample of 46 experts participated in the modified-Delphi process. A total of 68 elements (81%) were selected as required, spanning demographics and clinical characteristics, genomic data, cancer treatment, health and quality of life outcomes, and resource use. CONCLUSIONS Cost-effectiveness analyses will fail to reflect the real-world impacts of precision oncology without data to accurately characterize patient care trajectories and outcomes. Data collection in accordance with the proposed core data set will promote standardization and enable the generation of decision-grade evidence to inform reimbursement.
Collapse
Affiliation(s)
- Samantha Pollard
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer, Vancouver, Canada
| | - Deirdre Weymann
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer, Vancouver, Canada
| | - Brandon Chan
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer, Vancouver, Canada
| | - Morgan Ehman
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer, Vancouver, Canada
| | - Sarah Wordsworth
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, England, UK; Oxford NIHR Biomedical Research Centre, Oxford, England, UK
| | - James Buchanan
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, England, UK; Oxford NIHR Biomedical Research Centre, Oxford, England, UK
| | - Timothy P Hanna
- Department of Oncology, Queen's University, Kingston, Canada
| | - Cheryl Ho
- Division of Medical Oncology, BC Cancer, Vancouver, Canada; Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Howard J Lim
- Division of Medical Oncology, BC Cancer, Vancouver, Canada; Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Paula K Lorgelly
- Department of Applied Health Research, University College London, London, England, UK
| | - Adam J N Raymakers
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer, Vancouver, Canada
| | | | - Dean A Regier
- Canadian Centre for Applied Research in Cancer Control, Cancer Control Research, BC Cancer, Vancouver, Canada; School of Population and Public Health, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
6
|
Van Meenen J, Leysen H, Chen H, Baccarne R, Walter D, Martin B, Maudsley S. Making Biomedical Sciences publications more accessible for machines. MEDICINE, HEALTH CARE, AND PHILOSOPHY 2022; 25:179-190. [PMID: 35039972 DOI: 10.1007/s11019-022-10069-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/08/2022] [Indexed: 06/14/2023]
Abstract
With the rapidly expanding catalogue of scientific publications, especially within the Biomedical Sciences field, it is becoming increasingly difficult for researchers to search for, read or even interpret emerging scientific findings. PubMed, just one of the current biomedical data repositories, comprises over 33 million citations for biomedical research, and over 2500 publications are added each day. To further strengthen the impact biomedical research, we suggest that there should be more synergy between publications and machines. By bringing machines into the realm of research and publication, we can greatly augment the assessment, investigation and cataloging of the biomedical literary corpus. The effective application of machine-based manuscript assessment and interpretation is now crucial, and potentially stands as the most effective way for researchers to comprehend and process the tsunami of biomedical data and literature. Many biomedical manuscripts are currently published online in poorly searchable document types, with figures and data presented in formats that are partially inaccessible to machine-based approaches. The structure and format of biomedical manuscripts should be adapted to facilitate machine-assisted interrogation of this important literary corpus. In this context, it is important to embrace the concept that biomedical scientists should also write manuscripts that can be read by machines. It is likely that an enhanced human-machine synergy in reading biomedical publications will greatly enhance biomedical data retrieval and reveal novel insights into complex datasets.
Collapse
Affiliation(s)
- Joris Van Meenen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Hongyu Chen
- Weill Cornell Medical College, New York, NY, USA
| | - Rudi Baccarne
- Anet Library Automation, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Deborah Walter
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Bronwen Martin
- Faculty of Pharmaceutical, Veterinary and Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium.
| |
Collapse
|
7
|
John Cremin C, Dash S, Huang X. Big Data: Historic Advances and Emerging Trends in Biomedical Research. CURRENT RESEARCH IN BIOTECHNOLOGY 2022. [DOI: 10.1016/j.crbiot.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
8
|
Lu ZK, Xiong X, Lee T, Wu J, Yuan J, Jiang B. Big Data and Real-World Data based Cost-Effectiveness Studies and Decision-making Models: A Systematic Review and Analysis. Front Pharmacol 2021; 12:700012. [PMID: 34737696 PMCID: PMC8562301 DOI: 10.3389/fphar.2021.700012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Background: Big data and real-world data (RWD) have been increasingly used to measure the effectiveness and costs in cost-effectiveness analysis (CEA). However, the characteristics and methodologies of CEA based on big data and RWD remain unknown. The objectives of this study were to review the characteristics and methodologies of the CEA studies based on big data and RWD and to compare the characteristics and methodologies between the CEA studies with or without decision-analytic models. Methods: The literature search was conducted in Medline (Pubmed), Embase, Web of Science, and Cochrane Library (as of June 2020). Full CEA studies with an incremental analysis that used big data and RWD for both effectiveness and costs written in English were included. There were no restrictions regarding publication date. Results: 70 studies on CEA using RWD (37 with decision-analytic models and 33 without) were included. The majority of the studies were published between 2011 and 2020, and the number of CEA based on RWD has been increasing over the years. Few CEA studies used big data. Pharmacological interventions were the most frequently studied intervention, and they were more frequently evaluated by the studies without decision-analytic models, while those with the model focused on treatment regimen. Compared to CEA studies using decision-analytic models, both effectiveness and costs of those using the model were more likely to be obtained from literature review. All the studies using decision-analytic models included sensitivity analyses, while four studies no using the model neither used sensitivity analysis nor controlled for confounders. Conclusion: The review shows that RWD has been increasingly applied in conducting the cost-effectiveness analysis. However, few CEA studies are based on big data. In future CEA studies using big data and RWD, it is encouraged to control confounders and to discount in long-term research when decision-analytic models are not used.
Collapse
Affiliation(s)
- Z Kevin Lu
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, Columbia, SC, United States
| | - Xiaomo Xiong
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, Columbia, SC, United States
| | - Taiying Lee
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, Columbia, SC, United States
| | - Jun Wu
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC, United States
| | - Jing Yuan
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, China
| | - Bin Jiang
- Department of Administrative and Clinical Pharmacy, School of Pharmaceutical Sciences, Health Science Center, Peking University, Beijing, China
| |
Collapse
|
9
|
Mascheroni P, Savvopoulos S, Alfonso JCL, Meyer-Hermann M, Hatzikirou H. Improving personalized tumor growth predictions using a Bayesian combination of mechanistic modeling and machine learning. COMMUNICATIONS MEDICINE 2021; 1:19. [PMID: 35602187 PMCID: PMC9053281 DOI: 10.1038/s43856-021-00020-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In clinical practice, a plethora of medical examinations are conducted to assess the state of a patient's pathology producing a variety of clinical data. However, investigation of these data faces two major challenges. Firstly, we lack the knowledge of the mechanisms involved in regulating these data variables, and secondly, data collection is sparse in time since it relies on patient's clinical presentation. The former limits the predictive accuracy of clinical outcomes for any mechanistic model. The latter restrains any machine learning algorithm to accurately infer the corresponding disease dynamics. METHODS Here, we propose a novel method, based on the Bayesian coupling of mathematical modeling and machine learning, aiming at improving individualized predictions by addressing the aforementioned challenges. RESULTS We evaluate the proposed method on a synthetic dataset for brain tumor growth and analyze its performance in predicting two relevant clinical outputs. The method results in improved predictions in almost all simulated patients, especially for those with a late clinical presentation (>95% patients show improvements compared to standard mathematical modeling). In addition, we test the methodology in two additional settings dealing with real patient cohorts. In both cases, namely cancer growth in chronic lymphocytic leukemia and ovarian cancer, predictions show excellent agreement with reported clinical outcomes (around 60% reduction of mean squared error). CONCLUSIONS We show that the combination of machine learning and mathematical modeling approaches can lead to accurate predictions of clinical outputs in the context of data sparsity and limited knowledge of disease mechanisms.
Collapse
Affiliation(s)
- Pietro Mascheroni
- Braunschweig Integrated Centre of Systems Biology and Helmholtz Centre for Infectious Research, Braunschweig, Germany
| | - Symeon Savvopoulos
- grid.5596.f0000 0001 0668 7884KU Leuven, Department of Chemical Engineering, Leuven, Belgium
| | - Juan Carlos López Alfonso
- Braunschweig Integrated Centre of Systems Biology and Helmholtz Centre for Infectious Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Braunschweig Integrated Centre of Systems Biology and Helmholtz Centre for Infectious Research, Braunschweig, Germany ,Centre for Individualized Infection Medicine, Hannover, Germany ,grid.6738.a0000 0001 1090 0254Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Haralampos Hatzikirou
- grid.440568.b0000 0004 1762 9729Mathematics Department, Khalifa University, Abu Dhabi, UAE ,grid.4488.00000 0001 2111 7257Centre for Information Services and High Performance Computing, TU Dresden, Dresden, Germany
| |
Collapse
|
10
|
van Biesen W, Van Der Straeten C, Sterckx S, Steen J, Diependaele L, Decruyenaere J. The concept of justifiable healthcare and how big data can help us to achieve it. BMC Med Inform Decis Mak 2021; 21:87. [PMID: 33676513 PMCID: PMC7937275 DOI: 10.1186/s12911-021-01444-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/16/2021] [Indexed: 01/08/2023] Open
Abstract
Over the last decades, the face of health care has changed dramatically, with big improvements in what is technically feasible. However, there are indicators that the current approach to evaluating evidence in health care is not holistic and hence in the long run, health care will not be sustainable. New conceptual and normative frameworks for the evaluation of health care need to be developed and investigated. The current paper presents a novel framework of justifiable health care and explores how the use of artificial intelligence and big data can contribute to achieving the goals of this framework.
Collapse
Affiliation(s)
- Wim van Biesen
- Renal Division, 0K12 IA, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Gent, Belgium.
- Consortium for Justifiable Healthcare, Ghent University Hospital, Ghent, Belgium.
| | | | - Sigrid Sterckx
- Consortium for Justifiable Healthcare, Ghent University Hospital, Ghent, Belgium
- Bioethics Institute Ghent, Department of Philosophy and Moral Sciences, Ghent University, Ghent, Belgium
| | - Johan Steen
- Renal Division, 0K12 IA, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Gent, Belgium
- Consortium for Justifiable Healthcare, Ghent University Hospital, Ghent, Belgium
| | - Lisa Diependaele
- Consortium for Justifiable Healthcare, Ghent University Hospital, Ghent, Belgium
- Bioethics Institute Ghent, Department of Philosophy and Moral Sciences, Ghent University, Ghent, Belgium
| | - Johan Decruyenaere
- Consortium for Justifiable Healthcare, Ghent University Hospital, Ghent, Belgium
- Department of Intensive Care, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
11
|
Burnett-Hartman AN, Udaltsova N, Kushi LH, Neslund-Dudas C, Rahm AK, Pawloski PA, Corley DA, Knerr S, Feigelson HS, Hunter JE, Tabano DC, Epstein MM, Honda SA, Ter-Minassian M, Lynch JA, Lu CY. Clinical Molecular Marker Testing Data Capture to Promote Precision Medicine Research Within the Cancer Research Network. JCO Clin Cancer Inform 2020; 3:1-10. [PMID: 31487201 DOI: 10.1200/cci.19.00026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE To evaluate health care systems for the availability of population-level data on the frequency of use and results of clinical molecular marker tests to inform precision cancer care. METHODS We assessed cancer-related molecular marker test data availability across 12 US health care systems in the Cancer Research Network. Overall, these systems provide care to a diverse population of more than 12 million people in the United States. We performed qualitative analyses of test data availability for five blood-based protein, nine germline, and 14 tissue-based tumor marker tests in each health care system's electronic health record and tumor registry using key informants, test code lists, and manual review of data types and output. We then performed quantitative analyses to estimate the proportion of patients with cancer with test utilization data and results for specific molecular marker tests. RESULTS Health systems were able to systematically capture population-level data on all five blood protein markers, six of 14 tissue-based tumor markers, and none of the nine germline markers. Successful, systematic data capture was achievable for tests with electronic data feeds for test results (blood protein markers) or through prior manual abstraction by tumor registrars (select tumor-based markers). For test results stored in scanned image files (particularly germline and tumor marker tests), information on which test was performed and test results was not readily accessible in an electronic format. CONCLUSION Even in health care systems with sophisticated electronic health records, there were few codified data elements available for evaluating precision cancer medicine test use and results at the population level. Health care organizations should establish standards for electronic reporting of precision medicine tests to expedite cancer research and facilitate the implementation of precision medicine approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sarah Knerr
- University of Washington and Kaiser Permanente Washington Health Research Institute, Seattle, WA
| | | | | | - David C Tabano
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO
| | - Mara M Epstein
- University of Massachusetts Medical School, Worcester, MA
| | | | | | - Julie A Lynch
- Department of Veterans Affairs Salt Lake City Health System, Salt Lake City, UT
| | - Christine Y Lu
- Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| |
Collapse
|
12
|
Chen Y, Chirikov VV, Marston XL, Yang J, Qiu H, Xie J, Sun N, Gu C, Dong P, Gao X. Machine Learning for Precision Health Economics and Outcomes Research (P-HEOR): Conceptual Review of Applications and Next Steps. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2020; 7:35-42. [PMID: 32685596 PMCID: PMC7299485 DOI: 10.36469/jheor.2020.12698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 05/15/2023]
Abstract
Precision health economics and outcomes research (P-HEOR) integrates economic and clinical value assessment by explicitly discovering distinct clinical and health care utilization phenotypes among patients. Through a conceptualized example, the objective of this review is to highlight the capabilities and limitations of machine learning (ML) applications to P-HEOR and to contextualize the potential opportunities and challenges for the wide adoption of ML for health economics. We outline a P-HEOR conceptual framework extending the ML methodology to comparatively assess the economic value of treatment regimens. Latest methodology developments on bias and confounding control in ML applications to precision medicine are also summarized.
Collapse
Affiliation(s)
- Yixi Chen
- Pfizer Investment Co. Ltd., Beijing,
China
| | - Viktor V. Chirikov
- Real World Evidence, Pharmerit International, Bethesda, Maryland,
United States
| | - Xiaocong L. Marston
- Real World Evidence, Pharmerit International, Bethesda, Maryland,
United States
- Pharmerit (Shanghai) Company Limited, Shanghai,
China
| | | | - Haibo Qiu
- Zhongda Hospital, Southeast University, Nanjing,
China
| | - Jianfeng Xie
- Zhongda Hospital, Southeast University, Nanjing,
China
| | - Ning Sun
- Easy Visible Sky Tree Technology (Beijing) Co., Ltd., Beijing,
China
| | - Chengming Gu
- Sanofi (China) Investment Co. Ltd., Beijing,
China
| | - Peng Dong
- Pfizer Investment Co. Ltd., Beijing,
China
| | - Xin Gao
- Real World Evidence, Pharmerit International, Bethesda, Maryland,
United States
- Pharmerit (Shanghai) Company Limited, Shanghai,
China
| |
Collapse
|
13
|
Park YJ, Shin MH, Moon SH. Radiogenomics Based on PET Imaging. Nucl Med Mol Imaging 2020; 54:128-138. [PMID: 32582396 DOI: 10.1007/s13139-020-00642-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/02/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Radiogenomics or imaging genomics is a novel omics strategy of associating imaging data with genetic information, which has the potential to advance personalized medicine. Imaging features extracted from PET or PET/CT enable assessment of in vivo functional and physiological activity and provide comprehensive tumor information non-invasively. However, PET features are considered secondary to features on conventional imaging, and there has not yet been a review of the radiogenomic approach using PET features. This review article summarizes the current state of PET-based radiogenomic research for cancer, which discusses some of its limitations and directions for future study.
Collapse
Affiliation(s)
- Yong-Jin Park
- Department of Nuclear Medicine and Molecular Imaging, Samsung Medical Center, Seoul, Republic of Korea
| | - Mu Heon Shin
- Department of Nuclear Medicine and Molecular Imaging, Samsung Medical Center, Seoul, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine and Molecular Imaging, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
14
|
Mackay ZP, Dukhovny D, Phillips KA, Beggs AH, Green RC, Parad RB, Christensen KD. Quantifying Downstream Healthcare Utilization in Studies of Genomic Testing. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:559-565. [PMID: 32389220 PMCID: PMC7293136 DOI: 10.1016/j.jval.2020.01.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/17/2019] [Accepted: 01/26/2020] [Indexed: 05/28/2023]
Abstract
OBJECTIVES The challenges of understanding how interventions influence follow-up medical care are magnified during genomic testing because few patients have received it to date and because the scope of information it provides is complex and often unexpected. We tested a novel strategy for quantifying downstream healthcare utilization after genomic testing to more comprehensively and efficiently identify related services. We also evaluated the effectiveness of different methods for collecting these data. METHODS We developed a risk-based approach for a trial of newborn genomic sequencing in which we defined primary conditions based on existing diagnoses and family histories of disease and defined secondary conditions based on unexpected findings. We then created patient-specific lists of services associated with managing primary and secondary conditions. Services were quantified based on medical record reviews, surveys, and telephone check-ins with parents. RESULTS By focusing on services that genomic testing would most likely influence in the short-term, we reduced the number of services in our analyses by more than 90% compared with analyses of all observed services. We also identified the same services that were ordered in response to unexpected findings as were identified during expert review and by confirming whether recommendations were completed. Data also showed that quantifying healthcare utilization with surveys and telephone check-ins alone would have missed the majority of attributable services. CONCLUSIONS Our risk-based strategy provides an improved approach for assessing the short-term impact of genomic testing and other interventions on healthcare utilization while conforming as much as possible to existing best-practice recommendations.
Collapse
Affiliation(s)
- Zoë P Mackay
- Boston University School of Medicine, Boston, MA, USA
| | - Dmitry Dukhovny
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Kathryn A Phillips
- Center for Translational and Policy Research on Personalized Medicine, Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, USA; Philip R Lee Institute for Health Policy, University of California San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Alan H Beggs
- Harvard Medical School, Boston, MA, USA; Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Robert C Green
- Harvard Medical School, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Partners Healthcare Personalized Medicine, Boston, MA, USA
| | - Richard B Parad
- Harvard Medical School, Boston, MA, USA; Department of Pediatric Newborn Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Kurt D Christensen
- Harvard Medical School, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA, USA.
| |
Collapse
|
15
|
Pontes C, Zara C, Torrent-Farnell J, Obach M, Nadal C, Vella-Bonanno P, Ermisch M, Simoens S, Hauegen RC, Gulbinovic J, Timoney A, Martin AP, Mueller T, Nachtnebel A, Campbell S, Selke G, Bochenek T, Rothe CC, Mardare I, Bennie M, Fürst J, Malmstrom RE, Godman B. Time to Review Authorisation and Funding for New Cancer Medicines in Europe? Inferences from the Case of Olaratumab. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2020; 18:5-16. [PMID: 31696433 DOI: 10.1007/s40258-019-00527-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The potential benefits of early patient access to new medicines in areas of high unmet medical need are recognised, but uncertainties concerning effectiveness, safety and added value when new medicines are authorised, and subsequently funded based on initial preliminary data only, have important implications. In 2016 olaratumab received accelerated conditional approval from both the European Medicines Agency and the US Food and Drug Administration for the treatment of soft-tissue sarcoma, based on the claims of a substantial reduction in the risk of death with an 11.8-month improvement in median overall survival in a phase II trial in combination with doxorubicin vs. doxorubicin alone. The failure to confirm these benefits in the post-authorisation pivotal trial has highlighted key concerns regarding early access and conditional approvals for new medicines. Concerns include potentially considerable clinical and economic costs, so that patients may have received suboptimal treatment and any money spent has foregone the opportunity to improve access to effective treatments. As a result, it seems reasonable to reconsider current marketing authorisation models and approaches. Potential pathways forward include closer collaboration between regulators, pharmaceutical companies and payers to enhance the generation of rapid and comparative confirmatory trials in a safe and fair manner, with minimal patient exposure as required to achieve robust evidence. Additionally, it may be time to review early access systems, and to explore new avenues regarding who should pay or part pay for new treatments whilst information is being collected as part of any obligations for conditional marketing authorisation. Greater co-operation between countries regarding the collection of data in routine clinical care, and further research on post-marketing data analysis and interpretation, may also contribute to improved appraisal and continued access to new innovative cancer treatments.
Collapse
Affiliation(s)
- Caridad Pontes
- Drug Area, Catalan Health Service, Travessera de les Corts 131, Edifici Olimpia, 08028, Barcelona, Spain.
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Corinne Zara
- Drug Area, Catalan Health Service, Travessera de les Corts 131, Edifici Olimpia, 08028, Barcelona, Spain
| | - Josep Torrent-Farnell
- Drug Area, Catalan Health Service, Travessera de les Corts 131, Edifici Olimpia, 08028, Barcelona, Spain
- Department of Pharmacology, Therapeutics and Toxicology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Merce Obach
- Drug Area, Catalan Health Service, Travessera de les Corts 131, Edifici Olimpia, 08028, Barcelona, Spain
| | | | - Patricia Vella-Bonanno
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Michael Ermisch
- Pharmaceutical Department, National Association of Statutory Health Insurance Funds, Berlin, Germany
| | - Steven Simoens
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Renata Curi Hauegen
- National Institute of Science and Technology for Innovation on Diseases of Neglected Populations (INCT-IDPN), Center for Technological Development in Health (CDTS), Osvaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Jolanta Gulbinovic
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Angela Timoney
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- NHS Lothian, Edinburgh, UK
| | - Antony P Martin
- Health Economics Centre, University of Liverpool Management School, Liverpool, UK
| | - Tanja Mueller
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Anna Nachtnebel
- Hauptverband der Österreichischen Sozialversicherungsträger, Vienna, Austria
| | - Stephen Campbell
- Centre for Primary Care, Division of Population Health, Health Services Research and Primary Care, University of Manchester, Manchester, UK
- NIHR Greater Manchester Patient Safety Translational Research Centre, School of Health Sciences, University of Manchester, Manchester, UK
| | - Gisbert Selke
- Wissenschaftliches Institut der AOK (WidO), Berlin, Germany
| | - Tomasz Bochenek
- Department of Drug Management, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Celia C Rothe
- Department of Drug Management, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Ileana Mardare
- Department of Public Health and Management, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Marion Bennie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Jurij Fürst
- Health Insurance Institute, Ljubljana, Slovenia
| | - Rickard E Malmstrom
- Department of Medicine Solna, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Brian Godman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
- Health Economics Centre, University of Liverpool Management School, Liverpool, UK
- Division of Clinical Pharmacology, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
- School of Pharmacy, Sefako Makgatho Health Sciences University, Ga-Rankuwa, South Africa
| |
Collapse
|