15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Bringing Onco-Innovation to Europe’s Healthcare Systems: The Potential of Biomarker Testing, Real World Evidence, Tumour Agnostic Therapies to Empower Personalised Medicine

      other
      1 , * , 2 , 3 , 4 , 5 , 6 , 7 , 8 , 9 , 10 , 11 , 12 , 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 21 , 8 , 22 , 1 , 2 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , *
      ,
      Cancers
      MDPI
      Europe’s Healthcare Systems, biomarkers, oncogenomics, HRD, cancers

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Simple Summary

          The increasing number of data supporting use of a personalized approach in cancer treatment, is changing the path of patient’s management. In the same time, the availability of technologies should allow patients to receive the best test for the specific individual condition. This is theoretically true, when a specific test is designed for the specific disease condition, while it is difficult to implement in the setting of agnostic therapies. Financial sources availability related to the non homogeneous health systems working in the different countries do not allow for an immediate implementation of the technologies and test commercially available. Future perspectives for targeted oncology include tumor-agnostic drugs, which target a given mutation and could be used in treating cancers from multiple organ types. Therefore, the present paper is aimed to both underline a how much important is this new view and also to sensitize the international bodies that supervise health policies at the decision-making level, with the aim of harmonizing cancer treatment pathways in at least all European countries.

          Abstract

          Rapid and continuing advances in biomarker testing are not being matched by uptake in health systems, and this is hampering both patient care and innovation. It also risks costing health systems the opportunity to make their services more efficient and, over time, more economical. The potential that genomics has brought to biomarker testing in diagnosis, prediction and research is being realised, pre-eminently in many cancers, but also in an ever-wider range of conditions—notably BRCA1/2 testing in ovarian, breast, pancreatic and prostate cancers. Nevertheless, the implementation of genetic testing in clinical routine setting is still challenging. Development is impeded by country-related heterogeneity, data deficiencies, and lack of policy alignment on standards, approval—and the role of real-world evidence in the process—and reimbursement. The acute nature of the problem is compellingly illustrated by the particular challenges facing the development and use of tumour agnostic therapies, where the gaps in preparedness for taking advantage of this innovative approach to cancer therapy are sharply exposed. Europe should already have in place a guarantee of universal access to a minimum suite of biomarker tests and should be planning for an optimum testing scenario with a wider range of biomarker tests integrated into a more sophisticated health system articulated around personalised medicine. Improving healthcare and winning advantages for Europe’s industrial competitiveness and innovation require an appropriate policy framework—starting with an update to outdated recommendations. We show herein the main issues and proposals that emerged during the previous advisory boards organised by the European Alliance for Personalized Medicine which mainly focus on possible scenarios of harmonisation of both oncogenetic testing and management of cancer patients.

          Related collections

          Most cited references39

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Pan-cancer whole-genome analyses of metastatic solid tumours

          Metastatic cancer is a major cause of death and is associated with poor treatment efficacy. A better understanding of the characteristics of late-stage cancer is required to help adapt personalized treatments, reduce overtreatment and improve outcomes. Here we describe the largest, to our knowledge, pan-cancer study of metastatic solid tumour genomes, including whole-genome sequencing data for 2,520 pairs of tumour and normal tissue, analysed at median depths of 106× and 38×, respectively, and surveying more than 70 million somatic variants. The characteristic mutations of metastatic lesions varied widely, with mutations that reflect those of the primary tumour types, and with high rates of whole-genome duplication events (56%). Individual metastatic lesions were relatively homogeneous, with the vast majority (96%) of driver mutations being clonal and up to 80% of tumour-suppressor genes being inactivated bi-allelically by different mutational mechanisms. Although metastatic tumour genomes showed similar mutational landscape and driver genes to primary tumours, we find characteristics that could contribute to responsiveness to therapy or resistance in individual patients. We implement an approach for the review of clinically relevant associations and their potential for actionability. For 62% of patients, we identify genetic variants that may be used to stratify patients towards therapies that either have been approved or are in clinical trials. This demonstrates the importance of comprehensive genomic tumour profiling for precision medicine in cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Interpretation and Impact of Real-World Clinical Data for the Practicing Clinician

            Real-world studies have become increasingly important in providing evidence of treatment effectiveness in clinical practice. While randomized clinical trials (RCTs) are the “gold standard” for evaluating the safety and efficacy of new therapeutic agents, necessarily strict inclusion and exclusion criteria mean that trial populations are often not representative of the patient populations encountered in clinical practice. Real-world studies may use information from electronic health and claims databases, which provide large datasets from diverse patient populations, and/or may be observational, collecting prospective or retrospective data over a long period of time. They can therefore provide information on the long-term safety, particularly pertaining to rare events, and effectiveness of drugs in large heterogeneous populations, as well as information on utilization patterns and health and economic outcomes. This review focuses on how evidence from real-world studies can be utilized to complement data from RCTs to gain a more complete picture of the advantages and disadvantages of medications as they are used in practice. Funding : Sanofi US, Inc.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Precision Medicine: From Science To Value

              Precision medicine is making an impact on patients, health care delivery systems, and research participants in ways that were only imagined fifteen years ago when the human genome was first sequenced. Discovery of disease-causing and drug-response genetic variants has accelerated, while adoption into clinical medicine has lagged. We define precision medicine and the stakeholder community required to enable its integration into research and health care. We explore the intersection of data science, analytics, and precision medicine in the formation of health systems that carry out research in the context of clinical care and that optimize the tools and information used to deliver improved patient outcomes. We provide examples of real-world impact and conclude with a policy and economic agenda necessary for the adoption of this new paradigm of health care both in the United States and globally.
                Bookmark

                Author and article information

                Contributors
                Role: Academic Editor
                Role: Academic Editor
                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                02 February 2021
                February 2021
                : 13
                : 3
                : 583
                Affiliations
                [1 ]European Alliance for Personalized Medicine, Avenue de l’Armee/ Legerlaan 10, 1040 Brussels, Belgium; Chiara.Bernini@ 123456euapm.eu
                [2 ]IRCCS Istituto Nazionale Tumori “Regina Elena”, Via Elio Chianesi, 53, 00128 Rome, Italy; gennaro.ciliberto@ 123456ifo.gov.it (G.C.); simonetta.buglioni@ 123456ifo.gov.it (S.B.)
                [3 ]Dipartimento di Scienze Chirurgiche Oncologiche e Gastroenterologiche, University of Padova, Via Giustiniani 2, 35128 Padova, Italy; pierfranco.conte@ 123456unipd.it
                [4 ]Department of Oncology and Hemato-Oncology, University of Milano and European Institute of Oncology, IRCCS, 20139 Milano, Italy; giuseppe.curigliano@ 123456ieo.it
                [5 ]Pulmonary Department, Clínica Universidad de Navarra, Calle Marquesado de Sta. Marta, 1, 28027 Madrid, Spain; lseijo@ 123456unav.es
                [6 ]Ciber Enfermedades Respiratorias (CIBERES), Av. de Monforte de Lemos, 3-5, 28029 Madrid, Spain
                [7 ]Center for Applied Medical Research (CIMA), Schools of Sciences and Medicine, University of Navarra, Av. de Pío XII, 55, 31008 Pamplona, Spain; lmontuenga@ 123456unav.es
                [8 ]CIBERONC, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain; nmalats@ 123456cnio.es
                [9 ]S.S. Oncologia Medica Toraco Polmonare, Fondazione IRCCS Istituto Nazionale dei Tumori Via Giacomo Venezian, 1, 20133 Milano, Italy; marina.garassino@ 123456istitutotumori.mi.it
                [10 ]Department of Pathology and Tumor Biology, University of Clermont-Auvergne, 49 bd François Mitterrand, 63001 Clermont-Ferrand, France; frederique.penault-llorca@ 123456clermont.unicancer.fr
                [11 ]Associazione aBRCAdaBRA Onlus Via Volontari Italiani del Sangue, 32, 90128 Palermo, Italy; fabrizia.galli@ 123456materdomini.it
                [12 ]Medical Oncology Department, Centre Leon Bérard & Université Claude Bernard Lyon, 69008 Lyon, France; isabelle.ray-coquard@ 123456lyon.unicancer.fr
                [13 ]Surgery Department, Institut Bergonié Cancer Center, Centre Léon Bérard Cheney D- 2 ème étage -28 Rue Laennec, 69373 Lyon, France; denis.querleu@ 123456esgo.org
                [14 ]Department of Pathology, Josephine Nefkens Institute, Erasmus Medical Center, Be 235b, Dr Molwaterplein 50, 3015 Rotterdam, The Netherlands; p.riegman@ 123456erasmusmc.nl
                [15 ]Department of Pathology, University of Aberdeen, King’s College, Aberdeen AB24 3FX, UK; Keith.Kerr@ 123456quinnipiac.edu
                [16 ]Department of Urology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; Hendrik.vanpoppel@ 123456uzleuven.be
                [17 ]Department of Urology, Skane University Hospital, Box 117, 221 00 Lund, Sweden; anders.bjartell@ 123456med.lu.se
                [18 ]Department of Medical and Surgical Sciences and Biotechnology, University of Rome, “la Sapienza”, Piazzale Aldo Moro, 5, 00185 Roma, Italy; Giovanni.Codacci-Pisanelli@ 123456uniroma1.it
                [19 ]Bulgarian Association for Personalised Medicine, 45 Bacho Kiro Str., 1202 Sofia, Bulgaria; office.bappm@ 123456gmail.com
                [20 ]Scientific Directorate, IRCCS Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco, 65, 70124 Bari, Italy; a.paradiso@ 123456oncologico.bari.it
                [21 ]Digestive Cancers Europe, Rue de la Loi 235, 1040 Brussels, Belgium; zorana.maravic@ 123456gmail.com (Z.M.); vfotaki@ 123456ed.ac.uk (V.F.)
                [22 ]Spanish National Cancer Research Centre (CNIO), Calle de Melchor Fernández Almagro, 3, 28029 Madrid, Spain
                [23 ]Independent Patient Advocate, 14 Farthing Road Downham Market, Norfolk PE38 0AF, UK; info@ 123456alastairkent.com
                [24 ]Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milano, Italy; elisabetta.munzone@ 123456ieo.it
                [25 ]KUZ-Coalition of Association in Healthcare, Trpimirova 11, 10000 Zagreb, Croatia; ivica.belina50@ 123456gmail.com
                [26 ]Thoracic Oncology-MOCA, University Hospital Antwerp, Wilrijkstraat 10, 2650 Edegem, Belgium; jan.van.meerbeeck@ 123456uza.be
                [27 ]UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; michael.j.duffy@ 123456ucd.ie
                [28 ]Maria Skłodowska-Curie Institute of Oncology, Wawelska 15 B, 00-001 Warszawa, Poland; beata.jagielska@ 123456pib-nio.pl
                [29 ]Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
                [30 ]CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80131 Napoli, Italy
                Author notes
                Author information
                https://orcid.org/0000-0003-2851-8605
                https://orcid.org/0000-0002-5210-5344
                https://orcid.org/0000-0002-8739-1387
                https://orcid.org/0000-0003-2472-8306
                https://orcid.org/0000-0003-4662-7944
                https://orcid.org/0000-0001-6495-2295
                https://orcid.org/0000-0003-2538-3784
                https://orcid.org/0000-0003-3371-3878
                https://orcid.org/0000-0002-9259-6619
                Article
                cancers-13-00583
                10.3390/cancers13030583
                7867284
                33540773
                bae1086a-11c9-4991-b038-8e5bb9bf027e
                © 2021 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 12 August 2020
                : 15 January 2021
                Categories
                Guidelines

                europe’s healthcare systems,biomarkers,oncogenomics,hrd,cancers

                Comments

                Comment on this article