1
|
Klas K, Strzebonska K, Zaborowska L, Krawczyk T, Włodarczyk A, Bąk-Kuchejda U, Polak M, Van Wambeke S, Waligora M. Risk and Benefit for Basket Trials in Oncology: A Systematic Review and Meta-Analysis. Target Oncol 2024:10.1007/s11523-024-01107-3. [PMID: 39455508 DOI: 10.1007/s11523-024-01107-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Oncology research is increasingly adopting new clinical trial models that implement the concept of precision medicine. One of these is the basket clinical trial design. Basket clinical trials allow new treatments to be evaluated across multiple tumor types. Patients recruited to basket clinical trials share certain molecular characteristics of their cancer that are predictive of clinical benefit from the experimental treatment. OBJECTIVE Our aim was to describe the risks and benefits of basket clinical trials in oncology. METHODS Our study was prospectively registered in PROSPERO (CRD42023406401). We systematically searched PubMed, Embase, and ClinicalTrials.gov for reports of basket clinical trials in oncology published between 1 January, 2001, and 14 June, 2023. We measured the risk by treatment-related adverse events (grades 3, 4, and 5), and the benefit by objective response rate. We also extracted and analyzed data on progression-free survival and overall survival. When possible, data were meta-analyzed. RESULTS We included 126 arms of 75 basket clinical trials accounting for 7659 patients. The pooled objective response rate was 18.0% (95% confidence interval [CI] 14.8-21.1). The rate of treatment-related death was 0.7% (95% CI 0.4-1.0), while 30.4% (95% CI 24.2-36.7) of patients experienced grade 3/4 drug-related toxicity. The median progression-free survival was 3.1 months (95% CI 2.6-3.9), and the median overall survival was 8.9 months (95% CI 6.7-10.2). CONCLUSIONS Our results provide an empirical basis for communicating about the risks and benefits of basket clinical trials and for refining new models of clinical trials applied in precision medicine.
Collapse
Affiliation(s)
- Katarzyna Klas
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Karolina Strzebonska
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
| | - Lucja Zaborowska
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Krawczyk
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Alicja Włodarczyk
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
| | - Urszula Bąk-Kuchejda
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Maciej Polak
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Department of Epidemiology and Population Studies, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | | | - Marcin Waligora
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland.
| |
Collapse
|
2
|
Maniaci A, Giurdanella G, Chiesa Estomba C, Mauramati S, Bertolin A, Lionello M, Mayo-Yanez M, Rizzo PB, Lechien JR, Lentini M. Personalized Treatment Strategies via Integration of Gene Expression Biomarkers in Molecular Profiling of Laryngeal Cancer. J Pers Med 2024; 14:1048. [PMID: 39452555 PMCID: PMC11508418 DOI: 10.3390/jpm14101048] [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: 08/20/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Laryngeal cancer poses a substantial challenge in head and neck oncology, and there is a growing focus on customized medicine techniques. The present state of gene expression indicators in laryngeal cancer and their potential to inform tailored therapy choices are thoroughly examined in this review. We examine significant molecular changes, such as TP53, CDKN2A, PIK3CA, and NOTCH1 mutations, which have been identified as important participants in the development of laryngeal cancer. The study investigates the predictive and prognostic significance of these genetic markers in addition to the function of epigenetic changes such as the methylation of the MGMT promoter. We also go over the importance of cancer stem cell-related gene expression patterns, specifically CD44 and ALDH1A1 expression, in therapy resistance and disease progression. The review focuses on indicators, including PD-L1, CTLA-4, and tumor mutational burden (TMB) in predicting immunotherapy responses, highlighting recent developments in our understanding of the intricate interactions between tumor genetics and the immune milieu. We also investigate the potential for improving prognosis accuracy and treatment selection by the integration of multi-gene expression panels with clinicopathological variables. The necessity for uniform testing and interpretation techniques is one of the difficulties, in implementing these molecular insights into clinical practice, that are discussed. This review seeks to provide a comprehensive framework for promoting personalized cancer therapy by combining the most recent data on gene expression profiling in laryngeal cancer. Molecularly guided treatment options may enhance patient outcomes.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (G.G.); (M.L.)
- ASP Ragusa-Hospital Giovanni Paolo II, 97100 Ragusa, Italy
- Head and Neck Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (C.C.E.); (M.M.-Y.); (J.R.L.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (G.G.); (M.L.)
| | - Carlos Chiesa Estomba
- Head and Neck Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (C.C.E.); (M.M.-Y.); (J.R.L.)
- Department of Otorhinolaryngology-Head and Neck Surgery, Hospital Universitario Donostia, 20003 San Sebastian, Spain
| | - Simone Mauramati
- Department of Otolaryngology Head Neck Surgery, University of Pavia, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Andy Bertolin
- Department Otorhinolaryngology, Vittorio Veneto Hospital (ML, AB), Anesthesia and Intensive Care, Vittorio Veneto Hospital, 31029 Vittorio Veneto, Italy; (A.B.); (M.L.)
| | - Marco Lionello
- Department Otorhinolaryngology, Vittorio Veneto Hospital (ML, AB), Anesthesia and Intensive Care, Vittorio Veneto Hospital, 31029 Vittorio Veneto, Italy; (A.B.); (M.L.)
| | - Miguel Mayo-Yanez
- Head and Neck Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (C.C.E.); (M.M.-Y.); (J.R.L.)
- Department of Otorhinolaryngology-Head and Neck Surgery, Hospital San Rafael (HSR), 15006 A Coruña, Spain
| | - Paolo Boscolo Rizzo
- Department of Medical, Surgical and Health Sciences, Section of Otolaryngology, University of Trieste, 34127 Trieste, Italy;
| | - Jerome R. Lechien
- Head and Neck Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (C.C.E.); (M.M.-Y.); (J.R.L.)
- Department of Otorhinolaryngology and Head and Neck Surgery, CHU de Bruxelles, CHU Saint-Pierre, School of Medicine, 64000 Brussels, Belgium
| | - Mario Lentini
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (G.G.); (M.L.)
| |
Collapse
|
3
|
van Dellen E. Precision psychiatry: predicting predictability. Psychol Med 2024; 54:1500-1509. [PMID: 38497091 DOI: 10.1017/s0033291724000370] [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] [Indexed: 03/19/2024]
Abstract
Precision psychiatry is an emerging field that aims to provide individualized approaches to mental health care. An important strategy to achieve this precision is to reduce uncertainty about prognosis and treatment response. Multivariate analysis and machine learning are used to create outcome prediction models based on clinical data such as demographics, symptom assessments, genetic information, and brain imaging. While much emphasis has been placed on technical innovation, the complex and varied nature of mental health presents significant challenges to the successful implementation of these models. From this perspective, I review ten challenges in the field of precision psychiatry, including the need for studies on real-world populations and realistic clinical outcome definitions, and consideration of treatment-related factors such as placebo effects and non-adherence to prescriptions. Fairness, prospective validation in comparison to current practice and implementation studies of prediction models are other key issues that are currently understudied. A shift is proposed from retrospective studies based on linear and static concepts of disease towards prospective research that considers the importance of contextual factors and the dynamic and complex nature of mental health.
Collapse
Affiliation(s)
- Edwin van Dellen
- Department of Psychiatry and University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
- Department of Neurology, UZ Brussel and Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
4
|
Lemaitre F, Florentin V, Blin O, Bayle A, Benito S, Chauny JV, Galaup A, Korchagina D, Lang M, Le Tourneau C, Pelloux H, Picard N, Guilhaumou R. Can precision medicine be integrated into routine therapeutic decisions at the bedside of patients? Therapie 2024; 79:13-22. [PMID: 38065821 DOI: 10.1016/j.therap.2023.11.007] [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: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 02/27/2024]
Abstract
Therapeutic strategies are shifting from a "one-size-fits-all" population-based approach to a stratified approach targeting groups with similar characteristics, or even individuals, tailoring treatments to the unique characteristics of each patient. Since such strategies rely on increasingly complex knowledge and healthcare technologies, along with an understanding of the tools of precision medicine, the appropriate dissemination and use of these strategies involves a number of challenges for the medical community. Having evaluation methodologies that have been jointly designed with the institutional, industrial, academic stakeholders, and also patients, like streamlining the processes and externally validating performances, could enhance the relevance of the "evaluation" aspect of precision medicine. Creating a network of expert precision-medicine centers and ensuring that precision-medicine procedures are reimbursed by social security would guarantee fair and sustainable access. Finally, training healthcare professionals, creating interfaces between precision-medicine expert centers and primary care professionals as well as patients, and integrating individual patient data into medical records are all key drivers that will enable information from precision-medicine to be made available and guarantee the proper use of these approaches.
Collapse
Affiliation(s)
- Florian Lemaitre
- Inserm, EHESP, Irset (institut de recherche en santé, environnement et travail), UMR S 1085, CHU de Rennes, université Rennes, 35000 Rennes, France.
| | - Virginie Florentin
- Direction médecine personnalisée, Roche SAS, 92600 Boulogne-Billancourt, France
| | - Olivier Blin
- Inserm 1106, service de pharmacologie clinique et pharmacosurveilleance, en delegation Tafalgie Therapeutics, UMR, Aix-Marseille université, AP-HM, 13385 Marseille, France
| | - Arnaud Bayle
- Bureau biostatistique et epidémiologie, Gustave-Roussy, université Paris-Saclay, 94800 Villejuif, France; Inserm, CESP U1018 Oncostat, labelisé Ligue contre le cancer, université Paris-Saclay, 94800 Villejuif, France
| | | | - Jean-Vannak Chauny
- Direction accès au marché & affaires publiques, Amgen, 92100 Boulogne-Billancourt, France
| | | | | | | | - Christophe Le Tourneau
- Institut Curie, Paris, France; Inserm U900, Saint-Cloud, France; Université Paris-Saclay, 75005 Paris, France
| | - Hervé Pelloux
- Parasitology-Mycology laboratory, University Grenoble-Alpes, University Hospital Grenoble-Alpes, 38043 Grenoble, France
| | - Nicolas Picard
- Service de pharmacologie, toxicologie et pharmacovigilance, centre de biologie et de recherche en santé (CBRS), CHU de Limoges, 87042 Limoges, France
| | | |
Collapse
|
5
|
Lemaitre F, Florentin V, Blin O, Bayle A, Benito S, Chauny JV, Galaup A, Korchagina D, Lang M, Le Tourneau C, Pelloux H, Picard N, Guilhaumou R. Comment faire entrer la médecine de précision dans la décision thérapeutique de routine au lit du malade ? Therapie 2024; 79:1-11. [PMID: 38129247 DOI: 10.1016/j.therap.2023.10.014] [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: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 12/23/2023]
Affiliation(s)
- Florian Lemaitre
- Inserm, EHESP, Institut de recherche en santé, environnement et travail (Irset) - UMR S 1085, université de Rennes, CHU de Rennes, 35000 Rennes, France.
| | - Virginie Florentin
- Direction médecine personnalisée, Roche SAS, 92600 Boulogne-Billancourt, France
| | - Olivier Blin
- Inserm 1106, service de pharmacologie clinique et pharmacosurveilleance, en delegation Tafalgie Therapeutics, UMR, Aix-Marseille université, AP-HM, 13385 Marseille, France
| | - Arnaud Bayle
- Bureau biostatistique et epidémiologie, Gustave-Roussy, université Paris-Saclay, 94800 Villejuif, France; Inserm, CESP U1018 Oncostat, labelisé Ligue contre le cancer, université Paris-Saclay, 94800 Villejuif, France
| | | | - Jean-Vannak Chauny
- Direction accès au marché & affaires publiques, Amgen, 92100 Boulogne-Billancourt, France
| | | | | | | | - Christophe Le Tourneau
- Institut Curie, 75000 Paris, France; Inserm U900, 92210 Saint-Cloud, France; Université Paris-Saclay, 75005 Paris, France
| | - Hervé Pelloux
- Parasitology-Mycology laboratory, University Grenoble-Alpes, University Hospital Grenoble-Alpes, 38043 Grenoble, France
| | - Nicolas Picard
- Service de pharmacologie, toxicologie et pharmacovigilance, centre de biologie et de recherche en santé (CBRS), CHU de Limoges, 87042 Limoges, France
| | | |
Collapse
|
6
|
Carels N, Sgariglia D, Junior MGV, Lima CR, Carneiro FRG, da Silva GF, da Silva FAB, Scardini R, Tuszynski JA, de Andrade CV, Monteiro AC, Martins MG, da Silva TG, Ferraz H, Finotelli PV, Balbino TA, Pinto JC. A Strategy Utilizing Protein-Protein Interaction Hubs for the Treatment of Cancer Diseases. Int J Mol Sci 2023; 24:16098. [PMID: 38003288 PMCID: PMC10671768 DOI: 10.3390/ijms242216098] [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: 07/23/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 11/26/2023] Open
Abstract
We describe a strategy for the development of a rational approach of neoplastic disease therapy based on the demonstration that scale-free networks are susceptible to specific attacks directed against its connective hubs. This strategy involves the (i) selection of up-regulated hubs of connectivity in the tumors interactome, (ii) drug repurposing of these hubs, (iii) RNA silencing of non-druggable hubs, (iv) in vitro hub validation, (v) tumor-on-a-chip, (vi) in vivo validation, and (vii) clinical trial. Hubs are protein targets that are assessed as targets for rational therapy of cancer in the context of personalized oncology. We confirmed the existence of a negative correlation between malignant cell aggressivity and the target number needed for specific drugs or RNA interference (RNAi) to maximize the benefit to the patient's overall survival. Interestingly, we found that some additional proteins not generally targeted by drug treatments might justify the addition of inhibitors designed against them in order to improve therapeutic outcomes. However, many proteins are not druggable, or the available pharmacopeia for these targets is limited, which justifies a therapy based on encapsulated RNAi.
Collapse
Affiliation(s)
- Nicolas Carels
- Platform of Biological System Modeling, Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (C.R.L.); (G.F.d.S.)
| | - Domenico Sgariglia
- Engenharia de Sistemas e Computação, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-972, RJ, Brazil;
| | - Marcos Guilherme Vieira Junior
- Computational Modeling of Biological Systems, Scientific Computing Program (PROCC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil or (M.G.V.J.); (F.A.B.d.S.)
| | - Carlyle Ribeiro Lima
- Platform of Biological System Modeling, Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (C.R.L.); (G.F.d.S.)
| | - Flávia Raquel Gonçalves Carneiro
- Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (F.R.G.C.); (R.S.)
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, RJ, Brazil
| | - Gilberto Ferreira da Silva
- Platform of Biological System Modeling, Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (C.R.L.); (G.F.d.S.)
| | - Fabricio Alves Barbosa da Silva
- Computational Modeling of Biological Systems, Scientific Computing Program (PROCC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil or (M.G.V.J.); (F.A.B.d.S.)
| | - Rafaela Scardini
- Center of Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, RJ, Brazil; (F.R.G.C.); (R.S.)
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, RJ, Brazil
- Centro de Ciências Biológicas e da Saúde (CCBS), Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro 22290-255, RJ, Brazil
| | - Jack Adam Tuszynski
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, 10129 Turin, Italy;
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
- Department of Physics, University of Alberta, Edmonton, AB T6G 2J1, Canada
| | - Cecilia Vianna de Andrade
- Department of Pathology, Instituto Fernandes Figueira, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 22250-020, RJ, Brazil;
| | - Ana Carolina Monteiro
- Laboratory of Osteo and Tumor Immunology, Department of Immunobiology, Fluminense Federal University, Rio de Janeiro 24210-201, RJ, Brazil;
| | - Marcel Guimarães Martins
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| | - Talita Goulart da Silva
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| | - Helen Ferraz
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| | - Priscilla Vanessa Finotelli
- Laboratório de Nanotecnologia Biofuncional, Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, RJ, Brazil;
| | - Tiago Albertini Balbino
- Nanotechnology Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil;
| | - José Carlos Pinto
- Chemical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-594, RJ, Brazil; (M.G.M.); (T.G.d.S.); (H.F.); (J.C.P.)
| |
Collapse
|
7
|
Jones RD, Abebe S, Distefano V, Mayer G, Poli I, Silvestri C, Slanzi D. Candidate composite biomarker to inform drug treatments for diabetic kidney disease. Front Med (Lausanne) 2023; 10:1271407. [PMID: 38020124 PMCID: PMC10646536 DOI: 10.3389/fmed.2023.1271407] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Current guidelines recommend renin angiotensin system inhibitors (RASi) as key components of treatment of diabetic kidney disease (DKD). Additional options include sodium-glucose cotransporter-2 inhibitors (SGLT2i), glucagon-like peptide 1 receptor agonists (GLP1a), and mineralocorticoid receptor antagonists (MCRa). The identification of the optimum drug combination for an individual is difficult because of the inter-, and longitudinal intra-individual heterogeneity of response to therapy. Results Using data from a large observational study (PROVALID), we identified a set of parameters that can be combined into a meaningful composite biomarker that appears to be able to identify which of the various treatment options is clinically beneficial for an individual. It uses machine-earning techniques to estimate under what conditions a treatment of RASi plus an additional treatment is different from the treatment with RASi alone. The measure of difference is the annual percent change (ΔeGFR) in the estimated glomerular filtration rate (ΔeGFR). The 1eGFR is estimated for both the RASi-alone treatment and the add-on treatment. Discussion Higher estimated increase of eGFR for add-on patients compared with RASi-alone patients indicates that prognosis may be improved with the add-on treatment. The personalized biomarker value thus identifies which patients may benefit from the additional treatment.
Collapse
Affiliation(s)
- Roger D. Jones
- European Centre for Living Technology, Ca' Foscari University of Venice, Venice, Italy
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Systems Engineering and Research Center, Stevens Institute of Technology, Hoboken, NJ, United States
| | - Seyum Abebe
- European Centre for Living Technology, Ca' Foscari University of Venice, Venice, Italy
| | - Veronica Distefano
- European Centre for Living Technology, Ca' Foscari University of Venice, Venice, Italy
- Department of Economic Sciences, Università del Salento, Salento, Italy
| | - Gert Mayer
- Internal Medicine IV, Medical University Innsbruck, Innsbruck, Austria
| | - Irene Poli
- European Centre for Living Technology, Ca' Foscari University of Venice, Venice, Italy
| | - Claudio Silvestri
- European Centre for Living Technology, Ca' Foscari University of Venice, Venice, Italy
- Department of Environmental Sciences, Informatics and Statistics, Ca' Foscari University of Venice, Venice, Italy
| | - Debora Slanzi
- European Centre for Living Technology, Ca' Foscari University of Venice, Venice, Italy
- Department of Management, Ca' Foscari University of Venice, Venice, Italy
| |
Collapse
|
8
|
Edsjö A, Holmquist L, Geoerger B, Nowak F, Gomon G, Alix-Panabières C, Ploeger C, Lassen U, Le Tourneau C, Lehtiö J, Ott PA, von Deimling A, Fröhling S, Voest E, Klauschen F, Dienstmann R, Alshibany A, Siu LL, Stenzinger A. Precision cancer medicine: Concepts, current practice, and future developments. J Intern Med 2023; 294:455-481. [PMID: 37641393 DOI: 10.1111/joim.13709] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Precision cancer medicine is a multidisciplinary team effort that requires involvement and commitment of many stakeholders including the society at large. Building on the success of significant advances in precision therapy for oncological patients over the last two decades, future developments will be significantly shaped by improvements in scalable molecular diagnostics in which increasingly complex multilayered datasets require transformation into clinically useful information guiding patient management at fast turnaround times. Adaptive profiling strategies involving tissue- and liquid-based testing that account for the immense plasticity of cancer during the patient's journey and also include early detection approaches are already finding their way into clinical routine and will become paramount. A second major driver is the development of smart clinical trials and trial concepts which, complemented by real-world evidence, rapidly broaden the spectrum of therapeutic options. Tight coordination with regulatory agencies and health technology assessment bodies is crucial in this context. Multicentric networks operating nationally and internationally are key in implementing precision oncology in clinical practice and support developing and improving the ecosystem and framework needed to turn invocation into benefits for patients. The review provides an overview of the diagnostic tools, innovative clinical studies, and collaborative efforts needed to realize precision cancer medicine.
Collapse
Affiliation(s)
- Anders Edsjö
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
- Genomic Medicine Sweden (GMS), Kristianstad, Sweden
| | - Louise Holmquist
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Genomic Medicine Sweden (GMS), Kristianstad, Sweden
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | | | - Georgy Gomon
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France
- CREEC, MIVEGEC, University of Montpellier, Montpellier, France
| | - Carolin Ploeger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Ulrik Lassen
- Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- INSERM U900 Research Unit, Saint-Cloud, France
- Faculty of Medicine, Paris-Saclay University, Paris, France
| | - Janne Lehtiö
- Department of Oncology Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas von Deimling
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Emile Voest
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frederick Klauschen
- Institute of Pathology, Charite - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Munich Partner Site, Heidelberg, Germany
| | | | | | - Lillian L Siu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
| |
Collapse
|
9
|
Baumann M, Celis J, Ringborg U, Heitor M, Berns A, Albreht T, Arabadjiev J, Boutros M, Brandenburg M, Canhao H, Carneiro F, Chomienne C, De Lorenzo F, Eggermont AMM, Font A, Garralda E, Goulart M, Henrique R, Lawler M, Maier‐Hein L, Meunier F, Oberst S, Oliveira P, Papatriantafyllou M, Schüz J, Solary E, Valencia A, Vargas R, Weiderpass E, Wilking N. Engaging European society at the forefront of cancer research and care: How discussions at the 5 th Gago Conference on European Science policy led to the Heidelberg Manifesto. Mol Oncol 2023; 17:925-945. [PMID: 36938773 PMCID: PMC10257409 DOI: 10.1002/1878-0261.13423] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/26/2023] [Accepted: 03/17/2023] [Indexed: 03/21/2023] Open
Abstract
European cancer research stakeholders met in October 2022 in Heidelberg, Germany, at the 5th Gago conference on European Cancer Policy, to discuss the current cancer research and cancer care policy landscape in Europe. Meeting participants highlighted gaps in the existing European programmes focusing on cancer research, including Europe's Beating Cancer Plan (EBCP), the Mission on Cancer (MoC), Understanding Cancer (UNCAN.eu), and the joint action CRANE, and put forward the next priorities, in the form of the Heidelberg Manifesto for cancer research. This meeting report presents all discussions that shed light on how infrastructures can be effectively shaped for translational, prevention, clinical and outcomes cancer research, with a focus on implementation and sustainability and while engaging patients and the public. In addition, we summarize recommendations on how to introduce frameworks for the digitalization of European cancer research. Finally, we discuss what structures, commitment, and resources are needed to establish a collaborative cancer research environment in Europe to achieve the scale required for innovation.
Collapse
Affiliation(s)
- Michael Baumann
- German Cancer Research Center (DKFZ)HeidelbergGermany
- European Academy of Cancer SciencesStockholmSweden
| | - Julio Celis
- European Academy of Cancer SciencesStockholmSweden
- Danish Cancer Society Research CenterCopenhagenDenmark
| | - Ulrik Ringborg
- European Academy of Cancer SciencesStockholmSweden
- Cancer Center KarolinskaKarolinska University HospitalStockholmSweden
| | - Manuel Heitor
- European Academy of Cancer SciencesStockholmSweden
- Center for Innovation, Technology and Policy Research, IN+ @ IS TécnicoUniversity of LisbonPortugal
| | - Anton Berns
- European Academy of Cancer SciencesStockholmSweden
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Tit Albreht
- National Institute of Public Health of SloveniaLjubljanaSlovenia
- Faculty of MedicineUniversity of LjubljanaSlovenia
| | - Jeliazko Arabadjiev
- Clinic of Medical OncologyUniversity Hospital Acibadem City Clinic TokudaSofiaBulgaria
- Bulgarian Scientific Society of Immuno‐oncology, and MoC BoardSofiaBulgaria
| | - Michael Boutros
- European Academy of Cancer SciencesStockholmSweden
- Division Signaling and Functional GenomicsGerman Cancer Research Center (DKFZ) and Heidelberg UniversityGermany
- DKFZ‐Hector Cancer Institute at the University Medical Center MannheimGermany
| | | | - Helena Canhao
- Comprehensive Health Research Center (CHRC), NOVA Medical SchoolUniversidade Nova de LisboaPortugal
| | - Fatima Carneiro
- European Academy of Cancer SciencesStockholmSweden
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup)Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Faculdade de Medicina da Universidade do Porto (FMUP)Portugal
- Centro Hospitalar Universitário de São João (CHUSJ)PortoPortugal
| | | | - Francesco De Lorenzo
- European Academy of Cancer SciencesStockholmSweden
- European Cancer Patient CoalitionBrusselsBelgium
| | - Alexander M. M. Eggermont
- European Academy of Cancer SciencesStockholmSweden
- Department Cancer MedicineCSO Princess Máxima Centre Pediatric Oncology, University Medical Center UtrechtThe Netherlands
- Board of the Comprehensive Cancer Center MunichTechnical University MunichGermany
- Ludwig Maximiliaan UniversityMunichGermany
| | | | - Elena Garralda
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
- Cancer Core EuropeAmsterdamThe Netherlands
| | | | - Rui Henrique
- Department of Pathology & Cancer Biology & Epigenetics Group – Research Center of IPO Porto (CI‐IPOP)/RISE@CI‐IPOP (Health Research Network)Portuguese Oncology Institute of Porto (IPO‐Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (P.CCC Raquel Seruca)Portugal
- Department of Pathology and Molecular Immunology, School of Medicine & Biomedical SciencesUniversity of Porto (ICBAS‐UP)Portugal
| | - Mark Lawler
- European Academy of Cancer SciencesStockholmSweden
- FRCPath Patrick G Johnston Centre for Cancer Research, Faculty of Medicine, Health and Life SciencesQueen's University BelfastUK
| | - Lena Maier‐Hein
- Intelligent Medical Systems (IMSY)German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Francoise Meunier
- European Academy of Cancer SciencesStockholmSweden
- Belgian Royal Academy of MedicineBrusselsBelgium
| | - Simon Oberst
- Quality and AccreditationOrganisation of European Cancer InstitutesBrusselsBelgium
| | - Pedro Oliveira
- Nova School of Business and EconomicsCopenhagen Business School & Patient InnovationFrederiksbergDenmark
| | | | - Joachim Schüz
- European Academy of Cancer SciencesStockholmSweden
- International Agency for Research on Cancer (IARC/WHO)LyonFrance
| | - Eric Solary
- European Academy of Cancer SciencesStockholmSweden
- INSERM, U1287 and Department of HematologyGustave Roussy Cancer CenterVillejuifFrance
- Faculté de MédecineUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC)BarcelonaSpain
- ICREABarcelonaSpain
| | | | - Elisabete Weiderpass
- European Academy of Cancer SciencesStockholmSweden
- International Agency for Research on Cancer (IARC/WHO)LyonFrance
| | | |
Collapse
|
10
|
Teuwen LA, Roets E, D’Hoore P, Pauwels P, Prenen H. Comprehensive Genomic Profiling and Therapeutic Implications for Patients with Advanced Cancers: The Experience of an Academic Hospital. Diagnostics (Basel) 2023; 13:1619. [PMID: 37175010 PMCID: PMC10177779 DOI: 10.3390/diagnostics13091619] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Next-generation sequencing (NGS) can be used to detect tumor-specific genomic alterations. This retrospective single-center study aims to assess the application of an extensive NGS panel to identify actionable alterations and initiate matched targeted treatment for patients with advanced cancer. We analyzed genomic alterations in solid tumor biopsies from 464 patients with advanced cancer with the Foundation Medicine assay (FoundationOne®CDx). Therapeutic implications were determined using the Memorial Sloan Kettering Precision Oncology Knowledge Base (OncoKB) classification. The FoundationOne®CDx was successfully applied in 464/521 patients (89%). The most common altered genes were TP53 (61%), KRAS (20%), CDKN2A (20%), TERT (16%), and APC (16%). Among the 419 patients with successfully analyzed tumor mutational burden (TMB), 43 patients presented with a high TMB (≥10 mutations/megabase). Out of the 126 patients with an actionable target, 40 patients received matched treatment (32%) of which 17 were within a clinical trial. This study shows that the application of NGS is feasible in an academic center and increases the detection of actionable alterations and identification of patients eligible for targeted treatment or immunotherapy regardless of tumor histology. Strategies such as early referral for NGS, inclusion in clinical (basket) trials, and the development of new targeted drugs are necessary to improve the matched treatment rate.
Collapse
Affiliation(s)
- Laure-Anne Teuwen
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Evelyne Roets
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Pieter D’Hoore
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
| | - Patrick Pauwels
- Department of Pathology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium;
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Hans Prenen
- Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650 Edegem, Belgium; (L.-A.T.); (E.R.); (P.D.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
11
|
Spencer EA, Agrawal M, Jess T. Prognostication in inflammatory bowel disease. Front Med (Lausanne) 2022; 9:1025375. [PMID: 36275829 PMCID: PMC9582521 DOI: 10.3389/fmed.2022.1025375] [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] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Personalized care in inflammatory bowel diseases (IBD) hinges on parsing the heterogeneity of IBD patients through prognostication of their disease course and therapeutic response to allow for tailor-made treatment and monitoring strategies to optimize care. Herein we review the currently available predictors of outcomes in IBD and those on the both near and far horizons. We additionally discuss the importance of worldwide collaborative efforts and tools to support clinical use of these prognostication tools.
Collapse
Affiliation(s)
- Elizabeth A. Spencer
- Division of Pediatric Gastroenterology and Nutrition, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, United States
| | - Manasi Agrawal
- Division of Gastroenterology, Icahn School of Medicine, Mount Sinai Hospital, New York, NY, United States
- Center for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Aalborg University, Aalborg, Denmark
| | - Tine Jess
- Center for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Aalborg University, Aalborg, Denmark
- Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
12
|
Klein H, Mazor T, Siegel E, Trukhanov P, Ovalle A, Vecchio Fitz CD, Zwiesler Z, Kumari P, Van Der Veen B, Marriott E, Hansel J, Yu J, Albayrak A, Barry S, Keller RB, MacConaill LE, Lindeman N, Johnson BE, Rollins BJ, Do KT, Beardslee B, Shapiro G, Hector-Barry S, Methot J, Sholl L, Lindsay J, Hassett MJ, Cerami E. MatchMiner: an open-source platform for cancer precision medicine. NPJ Precis Oncol 2022; 6:69. [PMID: 36202909 PMCID: PMC9537311 DOI: 10.1038/s41698-022-00312-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Widespread, comprehensive sequencing of patient tumors has facilitated the usage of precision medicine (PM) drugs to target specific genomic alterations. Therapeutic clinical trials are necessary to test new PM drugs to advance precision medicine, however, the abundance of patient sequencing data coupled with complex clinical trial eligibility has made it challenging to match patients to PM trials. To facilitate enrollment onto PM trials, we developed MatchMiner, an open-source platform to computationally match genomically profiled cancer patients to PM trials. Here, we describe MatchMiner’s capabilities, outline its deployment at Dana-Farber Cancer Institute (DFCI), and characterize its impact on PM trial enrollment. MatchMiner’s primary goals are to facilitate PM trial options for all patients and accelerate trial enrollment onto PM trials. MatchMiner can help clinicians find trial options for an individual patient or provide trial teams with candidate patients matching their trial’s eligibility criteria. From March 2016 through March 2021, we curated 354 PM trials containing a broad range of genomic and clinical eligibility criteria and MatchMiner facilitated 166 trial consents (MatchMiner consents, MMC) for 159 patients. To quantify MatchMiner’s impact on trial consent, we measured time from genomic sequencing report date to trial consent date for the 166 MMC compared to trial consents not facilitated by MatchMiner (non-MMC). We found MMC consented to trials 55 days (22%) earlier than non-MMC. MatchMiner has enabled our clinicians to match patients to PM trials and accelerated the trial enrollment process.
Collapse
Affiliation(s)
- Harry Klein
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA.
| | - Tali Mazor
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA.
| | - Ethan Siegel
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Pavel Trukhanov
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Andrea Ovalle
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | - Zachary Zwiesler
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Priti Kumari
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | - Eric Marriott
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Jason Hansel
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Joyce Yu
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Adem Albayrak
- Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan Barry
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rachel B Keller
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Neal Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Bruce E Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Barrett J Rollins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Khanh T Do
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian Beardslee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Geoffrey Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - John Methot
- Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - James Lindsay
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | - Michael J Hassett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ethan Cerami
- Department of Data Science, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| |
Collapse
|
13
|
Molecular oncology: what is needed to speed access to innovative therapies in clinical research? Curr Opin Oncol 2022; 34:575-578. [PMID: 35855513 DOI: 10.1097/cco.0000000000000880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW A better understanding of the biology of cancer cells has led in the past 20 years to more and more molecular and immunological driven treatment strategies impacting both clinical trials and day-to-day practice. The aim of this review is to describe new approaches to conduct clinical trials in this area to speed up drug development and increase access to innovation for cancer patients. RECENT FINDINGS The design of an early phase trial has an impact on its clinical benefit. Trials deriving from a specific biomarker or histologic characteristic (also known as enrichment design) are more likely to demonstrate benefit than trials based on a more conventional design. However, the increase of low incidence cancer molecular subtypes poses a major hurdle in the clinical management and drug development research for cancer patients. SUMMARY With the identification of news targets and the subsequent introduction of precision medicine, new strategies and tools are needed to provide access to biomarker identification and target-oriented clinical trials to all cancer patients. We propose to set up a new patient-centered model to conduct clinical trials allowing simply to 'bring the trial to the patient'.
Collapse
|
14
|
Barber FD, Campbell E, Yamamura Y, Patterson CJ, Hartnett AC, Kinahan H, Miller VA, Brink AL, Poullard A, Urschel GE, Brantley A, Cepeda IG, Goswami P, Charles S, Philip S, Bresser S, Musekiwa-Adjei S, Perez N, Le H, Phillips P, Subbiah V, Meric-Bernstam F, Dumbrava EE. Management of Adverse Events in Early Clinical Trials by Advanced Practice Providers in the Outpatient Setting: The University of Texas MD Anderson Cancer Center Experience. J Adv Pract Oncol 2022; 13:664-672. [PMID: 36199491 PMCID: PMC9514129 DOI: 10.6004/jadpro.2022.13.7.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Advanced practice providers (APPs) play important roles in enrolling, educating, and caring for patients in clinical trials. However, much remains unknown about the role of APPs in managing adverse events (AEs) in early (phase I to II) clinical trials. In this study, we assessed the outpatient management of grade 3 to 4 AEs by APPs in early trials and characterized the workflow of our APP Phase I to II Fast Track (FT) Clinic. Patients and Methods We retrospectively reviewed records of patients with advanced or metastatic solid tumors enrolled in phase I to II clinical trials who were seen by APPs from September 2017 to August 2018 in the APP phase I to II FT clinic in the Department of Investigational Cancer Therapeutics. Results A total of 808 patients enrolled in 159 clinical trials were seen in 2,697 visits (median 3 visits per patient; range 1-28) by 10 APPs. Treatment was interrupted in 6.9% of visits, and grade 3 to 4 AEs were seen in 5.4% of visits; however, patients from 1.4% of visits were sent to the emergency center (EC) and/or admitted. Patients referred to the EC and/or admitted were more likely to have baseline hypoalbuminemia, high lactate dehydrogenase, and poor Eastern Cooperative Oncology Group performance status (i.e., ECOG > 1; p < .001). There were no associations between EC referral and gender, APP years of experience, or type of treatment. Conclusions The APP Phase I to II FT Clinic has an important role in the management of AEs by APPs in early clinical trials in the outpatient setting, potentially avoiding EC visits and admissions.
Collapse
Affiliation(s)
- Fedricker D. Barber
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Erick Campbell
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuko Yamamura
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Clover J. Patterson
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Audrey C. Hartnett
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Holly Kinahan
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Victoria A. Miller
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amanda L. Brink
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anna Poullard
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriele E. Urschel
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andre Brantley
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Isabel G. Cepeda
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Poonam Goswami
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sheena Charles
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shincy Philip
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sara Bresser
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra Musekiwa-Adjei
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nageli Perez
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hung Le
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Penny Phillips
- Clinical Center for Targeted Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ecaterina E. Dumbrava
- From Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Mohamed L, Manjrekar S, Ng DP, Walsh A, Lopes G, Parker JL. The Effect of Biomarker Use on the Speed and Duration of Clinical Trials for Cancer Drugs. Oncologist 2022; 27:849-856. [PMID: 35993585 PMCID: PMC9526484 DOI: 10.1093/oncolo/oyac130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Background The purpose of this study was to explore the effects biomarkers have on the duration and speed of clinical trials in oncology. Materials and Methods Clinical trial data was pooled from www.clinicaltrials.gov within the 4 cancer indications of non-small cell lung cancer, breast cancer, melanoma, and colorectal cancer. Heatmaps of clinical timelines were used to display differences in the frequency and timing of clinical trials across trials that used or did not use biomarkers, for all 4 indications. Results Screening of 8630 clinical trials across the 4 indications yielded 671 unique drugs corresponding to 1224 eligible trials used in our analysis. The constructed heatmaps visually represented that biomarkers did not have an effect on the time gap between trial phases for non-small cell lung cancer and melanoma but did for colorectal and breast cancer trials, reducing the speed of trial timelines. It was also observed that biomarker trials were more often concurrent over shorter periods of time and began later in the timeline for non-small cell lung and colorectal cancers. Conclusion The novel visualization method revealed longer gaps between trial phases, later clinical trial start times, and shorter periods of concurrently run trials for drugs that used biomarkers. The study highlights that biomarker-driven trials might impact drug approval timelines and need to be considered carefully in clinical development plan.
Collapse
Affiliation(s)
- Luqmaan Mohamed
- Master of Biotechnology Program, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Siddhi Manjrekar
- Master of Biotechnology Program, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Derek P Ng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Alec Walsh
- Cheriton School of Computer Science, University of Waterloo, Waterloo, ON, Canada
| | - Gilberto Lopes
- University of Miami, Miller School of Medicine, Coral Gables, FL, USA
| | - Jayson L Parker
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| |
Collapse
|
16
|
New Drug Development and Clinical Trial Design by Applying Genomic Information Management. Pharmaceutics 2022; 14:pharmaceutics14081539. [PMID: 35893795 PMCID: PMC9330622 DOI: 10.3390/pharmaceutics14081539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
Depending on the patients’ genotype, the same drug may have different efficacies or side effects. With the cost of genomic analysis decreasing and reliability of analysis methods improving, vast amount of genomic information has been made available. Several studies in pharmacology have been based on genomic information to select the optimal drug, determine the dose, predict efficacy, and prevent side effects. This paper reviews the tissue specificity and genomic information of cancer. If the tissue specificity of cancer is low, cancer is induced in various organs based on a single gene mutation. Basket trials can be performed for carcinomas with low tissue specificity, confirming the efficacy of one drug for a single gene mutation in various carcinomas. Conversely, if the tissue specificity of cancer is high, cancer is induced in only one organ based on a single gene mutation. An umbrella trial can be performed for carcinomas with a high tissue specificity. Some drugs are effective for patients with a specific genotype. A companion diagnostic strategy that prescribes a specific drug for patients selected with a specific genotype is also reviewed. Genomic information is used in pharmacometrics to identify the relationship among pharmacokinetics, pharmacodynamics, and biomarkers of disease treatment effects. Utilizing genomic information, sophisticated clinical trials can be designed that will be better suited to the patients of specific genotypes. Genomic information also provides prospects for innovative drug development. Through proper genomic information management, factors relating to drug response and effects can be determined by selecting the appropriate data for analysis and by understanding the structure of the data. Selecting pre-processing and appropriate machine-learning libraries for use as machine-learning input features is also necessary. Professional curation of the output result is also required. Personalized medicine can be realized using a genome-based customized clinical trial design.
Collapse
|
17
|
Outcomes and endpoints in clinical trials supporting the marketing authorisation of treatments in paediatric acute lymphoblastic leukaemia. Drug Discov Today 2022; 27:2440-2466. [PMID: 35597514 DOI: 10.1016/j.drudis.2022.05.015] [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: 12/09/2021] [Revised: 04/04/2022] [Accepted: 05/16/2022] [Indexed: 11/23/2022]
Abstract
The improvement in acute lymphoblastic leukaemia (ALL) treatment has led research efforts to focus on the unmet medical needs of an increasingly smaller patient cohort with resistant leukaemia and to develop more-targeted agents. Survival and response rates remain the most-prevalent endpoints in paediatric ALL research, but other intermediate clinical endpoints and molecular biomarkers for efficacy and mid- and long-term safety endpoints are also being investigated. The success of current ALL treatment appears to be driving new paradigms to optimise clinical drug development, while at the same time, regulatory tools in place are supporting meaningful drug development in the area.
Collapse
|
18
|
Snowdon C, Kernaghan S, Moretti L, Turner NC, Ring A, Wilkinson K, Martin S, Foster S, Kilburn LS, Bliss JM. Operational complexity versus design efficiency: challenges of implementing a phase IIa multiple parallel cohort targeted treatment platform trial in advanced breast cancer. Trials 2022; 23:372. [PMID: 35526005 PMCID: PMC9077636 DOI: 10.1186/s13063-022-06312-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/23/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Platform trial designs are used increasingly in cancer clinical research and are considered an efficient model for evaluating multiple compounds within a single disease or disease subtype. However, these trial designs can be challenging to operationalise. The use of platform trials in oncology clinical research has increased considerably in recent years as advances in molecular biology enable molecularly defined stratification of patient populations and targeted therapy evaluation. Whereas multiple separate trials may be deemed infeasible, platform designs allow efficient, parallel evaluation of multiple targeted therapies in relatively small biologically defined patient sub-populations with the promise of increased molecular screening efficiency and reduced time for drug evaluation. Whilst the theoretical efficiencies are widely reported, the operational challenges associated with these designs (complexity, cost, regulatory, resource) are not always well understood. MAIN: In this commentary, we describe our practical experience of the implementation and delivery of the UK plasmaMATCH trial, a platform trial in advanced breast cancer, comprising an integrated screening component and multiple parallel downstream mutation-directed therapeutic cohorts. plasmaMATCH reported its primary results within 3 years of opening to recruitment. We reflect on the operational challenges encountered and share lessons learnt to inform the successful conduct of future trials. Key to the success of the plasmaMATCH trial was well co-ordinated stakeholder engagement by an experienced clinical trials unit with expert methodology and trial management expertise, a federated model of clinical leadership, a well-written protocol integrating screening and treatment components and including justification for the chosen structure and intentions for future adaptions, and an integrated funding model with streamlined contractual arrangements across multiple partners. Findings based on our practical experience include the importance of early engagement with the regulators and consideration of a flexible resource infrastructure to allow adequate resource allocation to support concurrent trial activities as adaptions are implemented in parallel to the continued management of patient safety and data quality of the ongoing trial cohorts. CONCLUSION Platform trial designs allow the efficient reporting of multiple treatment cohorts. Operational challenges can be overcome through multidisciplinary engagement, streamlined contracting processes, rationalised protocol and database design and appropriate resourcing.
Collapse
Affiliation(s)
- Claire Snowdon
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK.
- The Institute of Cancer Research Clinical Trials and Statistics Unit, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.
| | - Sarah Kernaghan
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Laura Moretti
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Nicholas C Turner
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
- Breast Unit, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Alistair Ring
- Breast Unit, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Katie Wilkinson
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Sue Martin
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Stephanie Foster
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Lucy S Kilburn
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Judith M Bliss
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| |
Collapse
|
19
|
The Promise of Nanotechnology in Personalized Medicine. J Pers Med 2022; 12:jpm12050673. [PMID: 35629095 PMCID: PMC9142986 DOI: 10.3390/jpm12050673] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Both personalized medicine and nanomedicine are new to medical practice. Nanomedicine is an application of the advances of nanotechnology in medicine and is being integrated into diagnostic and therapeutic tools to manage an array of medical conditions. On the other hand, personalized medicine, which is also referred to as precision medicine, is a novel concept that aims to individualize/customize therapeutic management based on the personal attributes of the patient to overcome blanket treatment that is only efficient in a subset of patients, leaving others with either ineffective treatment or treatment that results in significant toxicity. Novel nanomedicines have been employed in the treatment of several diseases, which can be adapted to each patient-specific case according to their genetic profiles. In this review, we discuss both areas and the intersection between the two emerging scientific domains. The review focuses on the current situation in personalized medicine, the advantages that can be offered by nanomedicine to personalized medicine, and the application of nanoconstructs in the diagnosis of genetic variability that can identify the right drug for the right patient. Finally, we touch upon the challenges in both fields towards the translation of nano-personalized medicine.
Collapse
|
20
|
Rios JD, Velummailum R, Bennett J, Nobre L, Tsang DS, Bouffet E, Hawkins C, Tabori U, Denburg A, Pechlivanoglou P. Clinical and economic impact of molecular testing for BRAF fusion in pediatric low-grade Glioma. BMC Pediatr 2022; 22:13. [PMID: 34980048 PMCID: PMC8722113 DOI: 10.1186/s12887-021-03069-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/07/2021] [Indexed: 11/10/2022] Open
Abstract
Background Treatment personalization via tumor molecular testing holds promise for improving outcomes for patients with pediatric low-grade glioma (PLGG). We evaluate the health economic impact of employing tumor molecular testing to guide treatment for patients diagnosed with PLGG, particularly the avoidance of radiation therapy (RT) for patients with BRAF-fusion. Methods We performed a model-based cost-utility analysis comparing two strategies: molecular testing to determine BRAF fusion status at diagnosis against no molecular testing. We developed a microsimulation to model the lifetime health and cost outcomes (in quality-adjusted life years (QALYs) and 2018 CAD, respectively) for a simulated cohort of 100,000 patients newly diagnosed with PLGG after their initial surgery. Results The life expectancy after diagnosis for individuals who did not receive molecular testing was 39.01 (95% Confidence Intervals (CI): 32.94;44.38) years and 40.08 (95% CI: 33.19;45.76) years for those who received testing. Our findings indicate that patients who received molecular testing at diagnosis experienced a 0.38 (95% CI: 0.08;0.77) gain in QALYs and $1384 (95% CI: $-3486; $1204) reduction in costs over their lifetime. Cost and QALY benefits were driven primarily by the avoidance of long-term adverse events (stroke, secondary neoplasms) associated with unnecessary use of radiation. Conclusions We demonstrate the clinical benefit and cost-effectiveness of molecular testing in guiding the decision to provide RT in PLGG. While our results do not consider the impact of targeted therapies, this work is an example of the value of simulation modeling in assessing the long-term costs and benefits of precision oncology interventions for childhood cancer, which can aid decision-making about health system reimbursement. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-021-03069-1.
Collapse
Affiliation(s)
- Juan David Rios
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, 11th Floor - L4 East, Toronto, ON, M5G 0A4, Canada
| | - Russanthy Velummailum
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, 11th Floor - L4 East, Toronto, ON, M5G 0A4, Canada
| | - Julie Bennett
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Liana Nobre
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Derek S Tsang
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada.,Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Eric Bouffet
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- Department of Pathology, Hospital for Sick Children, Toronto, ON, Canada
| | - Uri Tabori
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada
| | - Avram Denburg
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, 11th Floor - L4 East, Toronto, ON, M5G 0A4, Canada.,Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada.,Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Petros Pechlivanoglou
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, 11th Floor - L4 East, Toronto, ON, M5G 0A4, Canada. .,Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada.
| |
Collapse
|
21
|
Botham J, Shilling V, Jones J. Patient and public understanding of the concept of 'personalised medicine' in relation to cancer treatment: a systematic review. Future Healthc J 2021; 8:e703-e708. [PMID: 34888471 DOI: 10.7861/fhj.2021-0063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Personalised medicine (PM) is becoming increasingly integrated into standard clinical practice for treating numerous diseases, including cancer. Implementing PM into healthcare systems will only be successful with the acceptance and input of both patients' and public opinion. This review, therefore, aimed to identify both patients' and public understanding, and perceived benefits and concerns of PM in cancer treatment. A literature search was conducted using MEDLINE, EMBASE, PsycINFO and CINAHL databases. The eligibility criteria specified that papers must explore the public or patients' understanding of PM or pharmacogenomic (PGx) testing in relation to cancer treatment. Patients have a greater understanding of, and trust in, PM compared with members of the public, but often misunderstand how genomic testing in PM works. Key areas that can be targeted to inform future health literacy interventions include genetic literacy for the public and understanding of how PM testing and treatment works for patients.
Collapse
Affiliation(s)
- Jed Botham
- Brighton and Sussex Medical School, Brighton, UK
| | - Valerie Shilling
- Sussex Health Outcomes Research and Education in Cancer (SHORE-C), Brighton, UK
| | - John Jones
- Brighton and Sussex Medical School, Brighton, UK
| |
Collapse
|
22
|
Ring A, Lang JE. ASO Author Reflections: Advancing Circulating Tumor Cells as Liquid Biopsies to Efficiently Target Metastatic Breast Cancer. Ann Surg Oncol 2021; 29:2895-2896. [PMID: 34853938 DOI: 10.1245/s10434-021-11139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 11/18/2022]
Affiliation(s)
- Alexander Ring
- Division of Surgical Oncology, Department of Surgery and University of Southern California Norris Cancer Center, University of Southern California, Los Angeles, CA, USA.,Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Julie E Lang
- Division of Surgical Oncology, Department of Surgery and University of Southern California Norris Cancer Center, University of Southern California, Los Angeles, CA, USA. .,Cleveland Clinic Breast Cancer Program, Division of Breast Services, Department of General Surgery, Cleveland, OH, USA.
| |
Collapse
|
23
|
Lévy V. Of some innovations in clinical trial design in hematology and oncology. Therapie 2021; 77:191-195. [PMID: 34922739 DOI: 10.1016/j.therap.2021.10.011] [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: 08/24/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022]
Abstract
The design of clinical trials, formalized in the immediate post-war period, has undergone major changes due to therapeutic innovations, particularly the arrival of targeted therapies in onco-hematology. The traditional phase I-II-III regimen is regularly questioned and multiple adaptations are proposed. This article proposes to expose some of these modifications and the issues they lead to.
Collapse
Affiliation(s)
- Vincent Lévy
- Département de recherche clinique, hôpital Avicenne, université Sorbonne Paris Nord, AP-HP, 93000 Bobigny, France.
| |
Collapse
|
24
|
Qiu T, Liang S, Wang Y, Dussart C, Borissov B, Toumi M. Reinforcing Collaboration and Harmonization to Unlock the Potentials of Advanced Therapy Medical Products: Future Efforts Are Awaited From Manufacturers and Decision-Makers. Front Public Health 2021; 9:754482. [PMID: 34900902 PMCID: PMC8655837 DOI: 10.3389/fpubh.2021.754482] [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: 08/06/2021] [Accepted: 10/22/2021] [Indexed: 11/29/2022] Open
Abstract
Some advanced therapy medicinal products (ATMPs) hold great promises for life-threatening diseases with high unmet needs. However, ATMPs are also associated with significant challenges in market access, which necessitates the joint efforts between all relevant stakeholders to navigate. In this review, we will elaborate on the importance of collaborations and harmonization across different stakeholders, to expedite the market access of promising ATMPs. Manufacturers of ATMPs should proactively establish collaborations with other stakeholders throughout the whole lifecycle of ATMPs, from early research to post-market activities. This covered engagements with (1) external developers (i.e., not-for-profit organizations and commercial players) to obtain complementary knowledge, technology, or infrastructures, (2) patient groups and healthcare providers to highlight their roles as active contributors, and (3) decision-makers, such as regulators, health technology assessment (HTA) agencies, and payers, to communicate the uncertainties in evidence package, where parallel consultation will be a powerful strategy. Harmonization between decision-makers is desired at (1) regulatory level, in terms of strengthening the international standardization of regulatory framework to minimize discrepancies in evidence requirements for market authorization, and (2) HTA level, in terms of enhancing alignments between regional and national HTA agencies to narrow inequity in patient access, and cross-border HTA cooperation to improve the quality and efficiency of HTA process. In conclusion, manufacturers and decision-makers shared the common goals to safeguard timely patient access to ATMPs. Collaboration and harmonization will be increasingly leveraged to enable the value delivery of ATMPs to all stakeholders.
Collapse
Affiliation(s)
- Tingting Qiu
- Département de Santé Publique, Aix-Marseille Université, Marseille, France
| | - Shuyao Liang
- Département de Santé Publique, Aix-Marseille Université, Marseille, France
| | - Yitong Wang
- Département de Santé Publique, Aix-Marseille Université, Marseille, France
| | - Claude Dussart
- Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Mondher Toumi
- Département de Santé Publique, Aix-Marseille Université, Marseille, France
| |
Collapse
|
25
|
Di Liello R, Piccirillo MC, Arenare L, Gargiulo P, Schettino C, Gravina A, Perrone F. Master Protocols for Precision Medicine in Oncology: Overcoming Methodology of Randomized Clinical Trials. Life (Basel) 2021; 11:1253. [PMID: 34833129 PMCID: PMC8618758 DOI: 10.3390/life11111253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/04/2021] [Accepted: 11/16/2021] [Indexed: 01/06/2023] Open
Abstract
Randomized clinical trials are considered the milestones of clinical research in oncology, and guided the development and approval of new compounds so far. In the last few years, however, molecular and genomic profiling led to a change of paradigm in therapeutic algorithms of many cancer types, with the spread of different biomarker-driven therapies (or targeted therapies). This scenario of "personalized medicine" revolutionized therapeutic strategies and the methodology of the supporting clinical research. New clinical trial designs are emerging to answer to the unmet clinical needs related to the development of these targeted therapies, overcoming the "classical" structure of randomized studies. Innovative trial designs able to evaluate more than one treatment in the same group of patients or many groups of patients with the same treatment (or both) are emerging as a possible future standard in clinical trial methodology. These are identified as "master protocols", and include umbrella, basket and platform trials. In this review, we described the main characteristics of these new trial designs, focusing on the opportunities and limitations of their use in the era of personalized medicine.
Collapse
Affiliation(s)
- Raimondo Di Liello
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Via S. Pansini 5, 80131 Napoli, Italy;
| | - Maria Carmela Piccirillo
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori—IRCCS Fondazione G. Pascale, Via M. Semmola, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (A.G.); (F.P.)
| | - Laura Arenare
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori—IRCCS Fondazione G. Pascale, Via M. Semmola, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (A.G.); (F.P.)
| | - Piera Gargiulo
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori—IRCCS Fondazione G. Pascale, Via M. Semmola, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (A.G.); (F.P.)
| | - Clorinda Schettino
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori—IRCCS Fondazione G. Pascale, Via M. Semmola, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (A.G.); (F.P.)
| | - Adriano Gravina
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori—IRCCS Fondazione G. Pascale, Via M. Semmola, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (A.G.); (F.P.)
| | - Francesco Perrone
- Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori—IRCCS Fondazione G. Pascale, Via M. Semmola, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (A.G.); (F.P.)
| |
Collapse
|
26
|
Werutsky G, Barrios CH, Cardona AF, Albergaria A, Valencia A, Ferreira CG, Rolfo C, de Azambuja E, Rabinovich GA, Sposetti G, Arrieta O, Dienstmann R, Rebelatto TF, Denninghoff V, Aran V, Cazap E. Perspectives on emerging technologies, personalised medicine, and clinical research for cancer control in Latin America and the Caribbean. Lancet Oncol 2021; 22:e488-e500. [PMID: 34735818 DOI: 10.1016/s1470-2045(21)00523-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022]
Abstract
Challenges of health systems in Latin America and the Caribbean include accessibility, inequity, segmentation, and poverty. These challenges are similar in different countries of the region and transcend national borders. The increasing digital transformation of health care holds promise of more precise interventions, improved health outcomes, increased efficiency, and ultimately reduced health-care costs. In Latin America and the Caribbean, the adoption of digital health tools is in early stages and the quality of cancer registries, electronic health records, and structured databases are problematic. Cancer research and innovation in the region are limited due to inadequate academic resources and translational research is almost fully dependent on public funding. Regulatory complexity and extended timelines jeopardise the potential improvement in participation in international studies. Emerging technologies, artificial intelligence, big data, and cancer research represent an opportunity to address the health-care challenges in Latin America and the Caribbean collectively, by optimising national capacities, sharing and comparing best practices, and transferring scientific and technical capabilities.
Collapse
Affiliation(s)
- Gustavo Werutsky
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil.
| | - Carlos H Barrios
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil; Oncology Department, Rio de Janeiro, Brazil
| | - Andres F Cardona
- Thoracic and Brain Tumor Unit, Clinical and Translational Oncology Group, Clínica del Country, Bogotá, Colombia; Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia; Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá, Colombia
| | - André Albergaria
- Translational Research & Industry Partnerships Unit, Instituto de Inovação em Saúde (i3S), Porto, Portugal
| | - Alfonso Valencia
- Institución Catalana de Investigación y Estudios Avanzados (ICREA) and Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Christian Rolfo
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Evandro de Azambuja
- Medical Oncology Department, Institut Jules Bordet and l'Université Libre de Bruxelles, Brussels, Belgium
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, and School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Georgina Sposetti
- Instituto de Investigaciones Clinicas Mar del Plata, Buenos Aires, Argentina; Un Ensayo para Mi, Buenos Aires, Argentina
| | - Oscar Arrieta
- Department of Thoracic Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Rodrigo Dienstmann
- Oncoclínicas Precision Medicine and Big Data Initiative, Rio de Janeiro, Brazil
| | | | - Valeria Denninghoff
- University of Buenos Aires - National Council for Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Veronica Aran
- Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Eduardo Cazap
- Latin American and Caribbean Society of Medical Oncology (SLACOM), Buenos Aires, Argentina
| |
Collapse
|
27
|
Ringborg U, Berns A, Celis JE, Heitor M, Tabernero J, Schüz J, Baumann M, Henrique R, Aapro M, Basu P, Beets‐Tan R, Besse B, Cardoso F, Carneiro F, van den Eede G, Eggermont A, Fröhling S, Galbraith S, Garralda E, Hanahan D, Hofmarcher T, Jönsson B, Kallioniemi O, Kásler M, Kondorosi E, Korbel J, Lacombe D, Carlos Machado J, Martin‐Moreno JM, Meunier F, Nagy P, Nuciforo P, Oberst S, Oliveiera J, Papatriantafyllou M, Ricciardi W, Roediger A, Ryll B, Schilsky R, Scocca G, Seruca R, Soares M, Steindorf K, Valentini V, Voest E, Weiderpass E, Wilking N, Wren A, Zitvogel L. The Porto European Cancer Research Summit 2021. Mol Oncol 2021; 15:2507-2543. [PMID: 34515408 PMCID: PMC8486569 DOI: 10.1002/1878-0261.13078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 01/22/2023] Open
Abstract
Key stakeholders from the cancer research continuum met in May 2021 at the European Cancer Research Summit in Porto to discuss priorities and specific action points required for the successful implementation of the European Cancer Mission and Europe's Beating Cancer Plan (EBCP). Speakers presented a unified view about the need to establish high-quality, networked infrastructures to decrease cancer incidence, increase the cure rate, improve patient's survival and quality of life, and deal with research and care inequalities across the European Union (EU). These infrastructures, featuring Comprehensive Cancer Centres (CCCs) as key components, will integrate care, prevention and research across the entire cancer continuum to support the development of personalized/precision cancer medicine in Europe. The three pillars of the recommended European infrastructures - namely translational research, clinical/prevention trials and outcomes research - were pondered at length. Speakers addressing the future needs of translational research focused on the prospects of multiomics assisted preclinical research, progress in Molecular and Digital Pathology, immunotherapy, liquid biopsy and science data. The clinical/prevention trial session presented the requirements for next-generation, multicentric trials entailing unified strategies for patient stratification, imaging, and biospecimen acquisition and storage. The third session highlighted the need for establishing outcomes research infrastructures to cover primary prevention, early detection, clinical effectiveness of innovations, health-related quality-of-life assessment, survivorship research and health economics. An important outcome of the Summit was the presentation of the Porto Declaration, which called for a collective and committed action throughout Europe to develop the cancer research infrastructures indispensable for fostering innovation and decreasing inequalities within and between member states. Moreover, the Summit guidelines will assist decision making in the context of a unique EU-wide cancer initiative that, if expertly implemented, will decrease the cancer death toll and improve the quality of life of those confronted with cancer, and this is carried out at an affordable cost.
Collapse
|
28
|
Salvo M, González-Feliú E, Toro J, Gallegos I, Maureira I, Miranda-González N, Barajas O, Bustamante E, Ahumada M, Colombo A, Armisén R, Villamán C, Ibañez C, Bravo ML, Sanhueza V, Spencer ML, de Toro G, Morales E, Bizama C, García P, Carrasco AM, Gutiérrez L, Bermejo JL, Verdugo RA, Marcelain K. Validation of an NGS Panel Designed for Detection of Actionable Mutations in Tumors Common in Latin America. J Pers Med 2021; 11:jpm11090899. [PMID: 34575676 PMCID: PMC8472524 DOI: 10.3390/jpm11090899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/29/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022] Open
Abstract
Next-generation sequencing (NGS) is progressively being used in clinical practice. However, several barriers preclude using this technology for precision oncology in most Latin American countries. To overcome some of these barriers, we have designed a 25-gene panel that contains predictive biomarkers for most current and near-future available therapies in Chile and Latin America. Library preparation was optimized to account for low DNA integrity observed in formalin-fixed paraffin-embedded tissue. The workflow includes an automated bioinformatic pipeline that accounts for the underrepresentation of Latin Americans in genome databases. The panel detected small insertions, deletions, and single nucleotide variants down to allelic frequencies of 0.05 with high sensitivity, specificity, and reproducibility. The workflow was validated in 272 clinical samples from several solid tumor types, including gallbladder (GBC). More than 50 biomarkers were detected in these samples, mainly in BRCA1/2, KRAS, and PIK3CA genes. In GBC, biomarkers for PARP, EGFR, PIK3CA, mTOR, and Hedgehog signaling inhibitors were found. Thus, this small NGS panel is an accurate and sensitive method that may constitute a more cost-efficient alternative to multiple non-NGS assays and costly, large NGS panels. This kind of streamlined assay with automated bioinformatics analysis may facilitate the implementation of precision medicine in Latin America.
Collapse
Affiliation(s)
- Mauricio Salvo
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
| | - Evelin González-Feliú
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
| | - Jessica Toro
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
| | - Iván Gallegos
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
- Department of Pathology, Hospital Clínico de la Universidad de Chile, Santiago 8380456, Chile
| | - Ignacio Maureira
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
- Department of Medical Technology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile
| | - Nicolás Miranda-González
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
| | - Olga Barajas
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
- Department of Internal Medicine, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
- Fundación Arturo López Pérez, Santiago 7500921, Chile; (E.B.); (A.M.C.)
| | - Eva Bustamante
- Fundación Arturo López Pérez, Santiago 7500921, Chile; (E.B.); (A.M.C.)
| | - Mónica Ahumada
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
- Department of Internal Medicine, Hospital Clínico Universidad de Chile, Santiago 8380456, Chile
| | - Alicia Colombo
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
- Department of Pathology, Hospital Clínico de la Universidad de Chile, Santiago 8380456, Chile
| | - Ricardo Armisén
- Center for Genetics and Genomics, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Camilo Villamán
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
| | - Carolina Ibañez
- Department of Hematology & Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile (PUC), Santiago 3580000, Chile; (C.I.); (M.L.B.)
| | - María Loreto Bravo
- Department of Hematology & Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile (PUC), Santiago 3580000, Chile; (C.I.); (M.L.B.)
| | - Verónica Sanhueza
- Department of Pathology, Hospital Padre Hurtado, Santiago 8710022, Chile;
| | - M. Loreto Spencer
- Department of Pathology, Hospital Clínico Regional Guillermo Grant Benavente, Concepción 4070038, Chile;
| | - Gonzalo de Toro
- School of Medical Technology, Universidad Austral de Chile at Puerto Montt, Puerto Montt 5110566, Chile;
| | - Erik Morales
- Department of Pathology, Hospital Regional de Talca, Talca 3460000, Chile;
- Department of Preclinical Sciences, Faculty of Medicine, Universidad Católica del Maule, Talca 3460000, Chile
| | - Carolina Bizama
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile; (C.B.); (P.G.)
| | - Patricia García
- Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile; (C.B.); (P.G.)
| | | | - Lorena Gutiérrez
- Department of Pathology, Hospital San Juan de Dios, Santiago 8320000, Chile;
| | | | - Ricardo A. Verdugo
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
- Human Genetics Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile
- Correspondence: (R.A.V.); (K.M.); Tel.: +56-22978-9527 (R.A.V.); +56-22978-9562 (K.M.)
| | - Katherine Marcelain
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago 8330015, Chile; (M.S.); (E.G.-F.); (J.T.); (I.G.); (I.M.); (N.M.-G.); (O.B.); (M.A.); (A.C.); (C.V.)
- Correspondence: (R.A.V.); (K.M.); Tel.: +56-22978-9527 (R.A.V.); +56-22978-9562 (K.M.)
| |
Collapse
|
29
|
Pierobon M, Robert NJ, Northfelt DW, Jahanzeb M, Wong S, Hodge KA, Baldelli E, Aldrich J, Craig DW, Liotta LA, Avramovic S, Wojtusiak J, Alemi F, Wulfkuhle JD, Bellos A, Gallagher RI, Arguello D, Conrad A, Kemkes A, Loesch DM, Vocila L, Dunetz B, Carpten JD, Petricoin EF, Anthony SP. Multi-omic molecular profiling guide's efficacious treatment selection in refractory metastatic breast cancer: a prospective phase II clinical trial. Mol Oncol 2021; 16:104-115. [PMID: 34437759 PMCID: PMC8732340 DOI: 10.1002/1878-0261.13091] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 11/22/2022] Open
Abstract
This prospective phase II clinical trial (Side Out 2) explored the clinical benefits of treatment selection informed by multi‐omic molecular profiling (MoMP) in refractory metastatic breast cancers (MBCs). Core needle biopsies were collected from 32 patients with MBC at trial enrollment. Patients had received an average of 3.94 previous lines of treatment in the metastatic setting before enrollment in this study. Samples underwent MoMP, including exome sequencing, RNA sequencing (RNA‐Seq), immunohistochemistry, and quantitative protein pathway activation mapping by Reverse Phase Protein Microarray (RPPA). Clinical benefit was assessed using the previously published growth modulation index (GMI) under the hypothesis that MoMP‐selected therapy would warrant further investigation for GMI ≥ 1.3 in ≥ 35% of the patients. Of the 32 patients enrolled, 29 received treatment based on their MoMP and 25 met the follow‐up criteria established by the trial protocol. Molecular information was delivered to the tumor board in a median time frame of 14 days (11–22 days), and targetable alterations for commercially available agents were found in 23/25 patients (92%). Of the 25 patients, 14 (56%) reached GMI ≥ 1.3. A high level of DNA topoisomerase I (TOPO1) led to the selection of irinotecan‐based treatments in 48% (12/25) of the patients. A pooled analysis suggested clinical benefit in patients with high TOPO1 expression receiving irinotecan‐based regimens (GMI ≥ 1.3 in 66.7% of cases). These results confirmed previous observations that MoMP increases the frequency of identifiable actionable alterations (92% of patients). The MoMP proposed allows the identification of biomarkers that are frequently expressed in MBCs and the evaluation of their role as predictors of response to commercially available agents. Lastly, this study confirmed the role of MoMP for informing treatment selection in refractory MBC patients: more than half of the enrolled patients reached a GMI ≥ 1.3 even after multiple lines of previous therapies for metastatic disease.
Collapse
Affiliation(s)
| | | | | | - Mohammad Jahanzeb
- A Division of 21st Century Oncology, Florida Precision Oncology, Raton, FL, USA
| | - Shukmei Wong
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | | | | - David W Craig
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | - Sanja Avramovic
- Department of Health Administration and Policy, George Mason University, Fairfax, VA, USA
| | - Janusz Wojtusiak
- Department of Health Administration and Policy, George Mason University, Fairfax, VA, USA
| | - Farrokh Alemi
- Department of Health Administration and Policy, George Mason University, Fairfax, VA, USA
| | | | | | | | | | | | | | | | - Linda Vocila
- Translational Drug Development (TD2), Scottsdale, AZ, USA
| | | | - John D Carpten
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | |
Collapse
|
30
|
Davies-Teye BB, Medeiros M, Chauhan C, Baquet CR, Mullins CD. Pragmatic patient engagement in designing pragmatic oncology clinical trials. Future Oncol 2021; 17:3691-3704. [PMID: 34337970 DOI: 10.2217/fon-2021-0556] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Oncology trials are the cornerstone of effective and safe therapeutic discoveries. However, there is increasing demand for pragmatism and patient engagement in the design, implementation and dissemination of oncology trials. Many researchers are uncertain about making trials more practical and even less knowledgeable about how to meaningfully engage patients without compromising scientific rigor to meet regulatory requirements. The present work provides practical guidance for addressing both pragmaticism and meaningful patient engagement. Applying evidence-based approaches like PRECIS-2-tool and the 10-Step Engagement Framework offer practical guidance to make future trials in oncology truly pragmatic and patient-centered. Consequently, such patient-centered trials have improved participation, faster recruitment and greater retention, and uptake of innovative technologies in community-based care.
Collapse
Affiliation(s)
- Bernard Bright Davies-Teye
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.,The PATIENTS Program, University of Maryland, Baltimore, MD 21201, USA
| | - Michelle Medeiros
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.,The PATIENTS Program, University of Maryland, Baltimore, MD 21201, USA
| | - Cynthia Chauhan
- The PATIENTS Program, University of Maryland, Baltimore, MD 21201, USA
| | - Claudia Rose Baquet
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.,The PATIENTS Program, University of Maryland, Baltimore, MD 21201, USA
| | - C Daniel Mullins
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.,The PATIENTS Program, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
31
|
Perrone ME, Alvarez R, Vo TT, Chung MW, Chhieng DC, Paulson VA, Colbert BG, Q Konnick E, Huang EC. Validating cell-free DNA from supernatant for molecular diagnostics on cytology specimens. Cancer Cytopathol 2021; 129:956-965. [PMID: 34265180 DOI: 10.1002/cncy.22491] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cytology specimens are often used for biomarker testing in the setting of neoplasia. On occasion, formalin-fixed paraffin-embedded (FFPE) cell blocks unfortunately may not yield sufficient material for testing. Recent studies have suggested that residual supernatant fluid from cell block preparation is a valuable source of DNA: both cellular and cell-free DNA (cfDNA). In the present study, the use of cfDNA from supernatant is compared against DNA from FFPE materials. METHODS cfDNA was extracted prospectively from residual supernatants of 30 cytology samples (29 neoplastic cases and 1 benign ascitic fluid from a patient with a history of melanoma). Samples were tested using clinically validated next-generation-sequencing platforms and the results were compared with data from paired FFPE cell blocks in a real-time prospective clinical setting. Thirteen samples were tested on an amplicon-based assay (Solid Tumor Hotspot), and 17 samples were tested using a comprehensive capture-based assay (UW-Oncoplex). RESULTS Neoplastic content was estimated by mutational variant allele fraction, with a mean content of 24.0% and 25.8% in supernatant and FFPE, respectively. The variant concordance between paired samples was 90%, and identical results were detected in both supernatant and FFPE samples in 74% of cases. CONCLUSIONS This study confirmed that cfDNA from supernatant is a viable alternative to FFPE cell blocks for molecular biomarker testing using both amplicon-based and capture-based assays with potential for decreasing additional tissue sampling and faster turnaround time.
Collapse
Affiliation(s)
- Marie E Perrone
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Rebeca Alvarez
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Tawnie T Vo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Moon-Wook Chung
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - David C Chhieng
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Vera A Paulson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Brice G Colbert
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Eric Q Konnick
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Eric C Huang
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
32
|
Vellekoop H, Huygens S, Versteegh M, Szilberhorn L, Zelei T, Nagy B, Koleva-Kolarova R, Tsiachristas A, Wordsworth S, Rutten-van Mölken M. Guidance for the Harmonisation and Improvement of Economic Evaluations of Personalised Medicine. PHARMACOECONOMICS 2021; 39:771-788. [PMID: 33860928 PMCID: PMC8200346 DOI: 10.1007/s40273-021-01010-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 05/02/2023]
Abstract
OBJECTIVE The objective of this study was to develop guidance contributing to improved consistency and quality in economic evaluations of personalised medicine (PM), given current ambiguity about how to measure the value of PM as well as considerable variation in the methodology and reporting in economic evaluations of PM. METHODS A targeted literature review of methodological papers was performed for an overview of modelling challenges in PM. Expert interviews were held to discuss best modelling practice. A systematic literature review of economic evaluations of PM was conducted to gain insight into current modelling practice. The findings were synthesised and used to develop a set of draft recommendations. The draft recommendations were discussed at a stakeholder workshop and subsequently finalised. RESULTS Twenty-two methodological papers were identified. Some argued that the challenges in modelling PM can be addressed within existing methodological frameworks, others disagreed. Eighteen experts were interviewed. They believed large uncertainty to be a key concern. Out of 195 economic evaluations of PM identified, 56% addressed none of the identified modelling challenges. A set of 23 recommendations was developed. Eight recommendations focus on the modelling of test-treatment pathways. The use of non-randomised controlled trial data is discouraged but several recommendations are provided in case randomised controlled trial data are unavailable. The parameterisation of structural uncertainty is recommended. Other recommendations consider perspective and discounting; premature survival data; additional value elements; patient and clinician compliance; and managed entry agreements. CONCLUSIONS This study provides a comprehensive list of recommendations to modellers of PM and to evaluators and reviewers of PM models.
Collapse
Affiliation(s)
- Heleen Vellekoop
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands.
| | - Simone Huygens
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
| | - Matthijs Versteegh
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
| | | | - Tamás Zelei
- Syreon Research Institute, Budapest, Hungary
| | - Balázs Nagy
- Syreon Research Institute, Budapest, Hungary
| | | | | | - Sarah Wordsworth
- Health Economics Research Centre, University of Oxford, Oxford, UK
| | - Maureen Rutten-van Mölken
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Sampath V, Ramchandran R. The Yin and the Yang of Transformative Research During the COVID-19 Pandemic-A Perspective. Front Pediatr 2021; 9:650302. [PMID: 34249804 PMCID: PMC8264183 DOI: 10.3389/fped.2021.650302] [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: 01/06/2021] [Accepted: 05/10/2021] [Indexed: 12/05/2022] Open
Abstract
The COVID-19 pandemic has highlighted the necessity for scientists from diverse disciplines to collaboratively mitigate the singular calamity facing humanity this century. The ability of researchers to combine exponential advances in technology and scientific acumen has resulted in landmark discoveries in pediatric research and is surmounting the COVID-19 challenge. Several of these discoveries exist in a realm of research that is not classically "basic" or "clinical." Translational research characterizes this domain partially, but does not fully capture the integrated research approaches that have spurred these discoveries. Herein, we share our perspective on the common themes underpinning the basic and clinical research. We also highlight major differences in the scope, emphasis, approach, and limitations of basic and clinical research that impede multi-disciplinary approaches that facilitate truly transformative research. These differences in research thinking and methodology are ingrained during training wherein the limitations of the chosen discipline, and strengths of alternate disciplines are not adequately explored. Insular approaches are particularly limited in impacting complex diseases pathophysiology in the era of precision medicine. We propose that integration of -omics technologies, systems biology, adaptive clinical trial designs, humanized animal models, and precision pre-clinical model systems must be incorporated into research training of future scientists. Several initiatives from the NIH and other institutions are facilitating such broad-based "research without frontiers" training that paves the way for seamless, multi-disciplinary, research. Such efforts become "transformative" when scientific challenges are tackled in partnership with a willingness to share ideas, tackle challenges, and develop tools/models from the very beginning.
Collapse
Affiliation(s)
- Venkatesh Sampath
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, United States
- Department of Pediatrics, University of Missouri at Kansas City, Kansas City, MO, United States
| | - Ramani Ramchandran
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
34
|
Boniolo F, Dorigatti E, Ohnmacht AJ, Saur D, Schubert B, Menden MP. Artificial intelligence in early drug discovery enabling precision medicine. Expert Opin Drug Discov 2021; 16:991-1007. [PMID: 34075855 DOI: 10.1080/17460441.2021.1918096] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Precision medicine is the concept of treating diseases based on environmental factors, lifestyles, and molecular profiles of patients. This approach has been found to increase success rates of clinical trials and accelerate drug approvals. However, current precision medicine applications in early drug discovery use only a handful of molecular biomarkers to make decisions, whilst clinics gear up to capture the full molecular landscape of patients in the near future. This deep multi-omics characterization demands new analysis strategies to identify appropriate treatment regimens, which we envision will be pioneered by artificial intelligence.Areas covered: In this review, the authors discuss the current state of drug discovery in precision medicine and present our vision of how artificial intelligence will impact biomarker discovery and drug design.Expert opinion: Precision medicine is expected to revolutionize modern medicine; however, its traditional form is focusing on a few biomarkers, thus not equipped to leverage the full power of molecular landscapes. For learning how the development of drugs can be tailored to the heterogeneity of patients across their molecular profiles, artificial intelligence algorithms are the next frontier in precision medicine and will enable a fully personalized approach in drug design, and thus ultimately impacting clinical practice.
Collapse
Affiliation(s)
- Fabio Boniolo
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,School of Medicine, Chair of Translational Cancer Research and Institute for Experimental Cancer Therapy, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Emilio Dorigatti
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Statistical Learning and Data Science, Department of Statistics, Ludwig Maximilian Universität München, Munich, Germany
| | - Alexander J Ohnmacht
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Dieter Saur
- School of Medicine, Chair of Translational Cancer Research and Institute for Experimental Cancer Therapy, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Benjamin Schubert
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Mathematics, Technical University of Munich, Garching, Germany
| | - Michael P Menden
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Munich, Germany.,Department of Biology, Ludwig-Maximilians University Munich, Martinsried, Germany.,German Centre for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
35
|
Su M, Zhang Z, Zhou L, Han C, Huang C, Nice EC. Proteomics, Personalized Medicine and Cancer. Cancers (Basel) 2021; 13:2512. [PMID: 34063807 PMCID: PMC8196570 DOI: 10.3390/cancers13112512] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023] Open
Abstract
As of 2020 the human genome and proteome are both at >90% completion based on high stringency analyses. This has been largely achieved by major technological advances over the last 20 years and has enlarged our understanding of human health and disease, including cancer, and is supporting the current trend towards personalized/precision medicine. This is due to improved screening, novel therapeutic approaches and an increased understanding of underlying cancer biology. However, cancer is a complex, heterogeneous disease modulated by genetic, molecular, cellular, tissue, population, environmental and socioeconomic factors, which evolve with time. In spite of recent advances in treatment that have resulted in improved patient outcomes, prognosis is still poor for many patients with certain cancers (e.g., mesothelioma, pancreatic and brain cancer) with a high death rate associated with late diagnosis. In this review we overview key hallmarks of cancer (e.g., autophagy, the role of redox signaling), current unmet clinical needs, the requirement for sensitive and specific biomarkers for early detection, surveillance, prognosis and drug monitoring, the role of the microbiome and the goals of personalized/precision medicine, discussing how emerging omics technologies can further inform on these areas. Exemplars from recent onco-proteogenomic-related publications will be given. Finally, we will address future perspectives, not only from the standpoint of perceived advances in treatment, but also from the hurdles that have to be overcome.
Collapse
Affiliation(s)
- Miao Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (M.S.); (Z.Z.); (L.Z.); (C.H.)
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (M.S.); (Z.Z.); (L.Z.); (C.H.)
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (M.S.); (Z.Z.); (L.Z.); (C.H.)
| | - Chao Han
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (M.S.); (Z.Z.); (L.Z.); (C.H.)
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China; (M.S.); (Z.Z.); (L.Z.); (C.H.)
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
36
|
Coart E, Saad ED. Considerations on the mechanics and sample sizes for early trials of targeted agents and immunotherapy in oncology. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1915693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Elisabeth Coart
- Consulting Services & Research, International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium
| | - Everardo D. Saad
- Consulting Services & Research, International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium
| |
Collapse
|
37
|
Issa AM, Carleton B, Gerhard T, Filipski KK, Freedman AN, Kimmel S, Liu G, Longo C, Maitland-van der Zee AH, Sansbury L, Zhou W, Bartlett G. Pharmacoepidemiology: A time for a new multidisciplinary approach to precision medicine. Pharmacoepidemiol Drug Saf 2021; 30:985-992. [PMID: 33715268 DOI: 10.1002/pds.5226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 03/09/2021] [Indexed: 11/06/2022]
Abstract
The advent of the genomic age has created a rapid increase in complexity for the development and selection of drug treatments. A key component of precision medicine is the use of genetic information to improve therapeutic effectiveness of drugs and prevent potential adverse drug reactions. Pharmacoepidemiology, as a field, uses observational methods to evaluate the safety and effectiveness of drug treatments in populations. Pharmacoepidemiology by virtue of its focus, tradition, and research orientation can provide appropriate study designs and analysis methods for precision medicine. The objective of this manuscript is to demonstrate how pharmacoepidemiology can impact and shape precision medicine and serve as a reference for pharmacoepidemiologists interested in contributing to the science of precision medicine. This paper depicts the state of the science with respect to the need for pharmacoepidemiology and pharmacoepidemiological methods, tools and approaches for precision medicine; the need for and how pharmacoepidemiologists use their skills to engage with the precision medicine community; and recommendations for moving the science of precision medicine pharmacoepidemiology forward. We propose a new integrated multidisciplinary approach dedicated to the emerging science of precision medicine pharmacoepidemiology.
Collapse
Affiliation(s)
- Amalia M Issa
- Personalized Precision Medicine & Targeted Therapeutics, Springfield, Pennsylvania, USA.,'Pharmaceutical Sciences' and 'Health Policy', University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA.,'Family Medicine' and `Centre of Genomics & Policy'; Faculty of Medicine & Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Bruce Carleton
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, and BC Children's Hospital and Research Institute, Vancouver, British Columbia, Canada
| | - Tobias Gerhard
- Center for Pharmacoepidemiology and Treatment Science, Rutgers University, New Brunswick, New Jersey, USA
| | - Kelly K Filipski
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland, USA
| | - Andrew N Freedman
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, Maryland, USA
| | - Stephen Kimmel
- 'College of Public Health & Health Professions' and 'College of Medicine', University of Florida, Gainesville, Florida, USA
| | - Geoffrey Liu
- Epidemiology; Dalla Lana School of Public Health, Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | - Cristina Longo
- Respiratory Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anke H Maitland-van der Zee
- Respiratory Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Leah Sansbury
- Epidemiology, Value Evidence and Outcomes, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Wei Zhou
- Center for Observational and Real-world Evidence, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Gillian Bartlett
- School of Medicine, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
38
|
Grisafi D, Ceschi A, Avalos Clerici V, Scaglione F. The Contribution of Clinical Pharmacologists in Precision Medicine: An Opportunity for Health Care Improvement. Curr Ther Res Clin Exp 2021; 94:100628. [PMID: 34306268 PMCID: PMC8296076 DOI: 10.1016/j.curtheres.2021.100628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/16/2021] [Indexed: 12/02/2022] Open
Abstract
Background Clinical pharmacologists play an important role and have professional value in the field, especially regarding their role within precision medicine (PM) and personalized therapies. Objective In this work, we sought to stimulate debate on the role of clinical pharmacologists. Methods A literature review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement, through electronic consultation of 2 databases, PubMed/Medline and Embase, and Google Scholar with manual research taking into account the peer-reviewed literature such as observational studies, reviews, original research articles, comments, mini-reviews, and opinion papers published in English between 2010 and February 2020. Titles and abstracts were screened by 1 author, and studies identified for full-text analysis and selected according to inclusion criteria were agreed on by 2 reviewers. Results We identified a total of 535 peer-reviewed articles and the number of full texts eligible for the project was 43. Several publications highlight the clinical value of pharmacologists in highly complex hospitals, where the strategies of PM are implemented. Although there are still no studies measuring the clinical efficiency and the efficacy of clinical pharmacology services, and the applicability of PM protocols, this review shows the considerable debate around the future mission of clinical pharmacology services as a bridging discipline capable of combining the complex knowledge and different professional skills needed to fully implement PM. Conclusions Various strategies have been conceived and planned to facilitate the transition from mainstream medicine to PM, which will enable patients to be treated more accurately, with significant advantages in terms of safety and effectiveness of treatments. Therefore, in the future, to ensure that the evolutionary process of medicine can involve as many patients and caregivers as possible, infrastructures capable of bringing together different multidisciplinary skills among health professionals will have to be implemented. Clinical pharmacologists could be the main drivers of this strategy because they already, with their multidisciplinary training, operate in a series of services in high-level hospitals, facilitating the clinical governance of the most challenging patients. The implementation of these strategies will lastly allow national health organizations to adequately address the management and therapeutic challenges related to the advent of new drugs and cell and gene therapies by facilitating the removal of economic and organizational barriers to ensure equitable access to PM. (Curr Ther Res Clin Exp. 2021; 82:XXX–XXX)
Collapse
Affiliation(s)
- Davide Grisafi
- Department of Biotechnology and Translational Medicine, University of Milano, Via Vanvitelli, 32 20129 MILANO (MI), Milan, Italy
| | - Alessandro Ceschi
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland.,Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland.,Faculty of Biomedical Sciences, University of Southern Switzerland, Lugano, Switzerland
| | | | - Francesco Scaglione
- Department of Biotechnology and Translational Medicine, University of Milano, Via Vanvitelli, 32 20129 MILANO (MI), Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milano, Via Vanvitelli, 32 20129 MILANO (MI), Milan, Italy
| |
Collapse
|
39
|
Advani D, Sharma S, Kumari S, Ambasta RK, Kumar P. Precision Oncology, Signaling and Anticancer Agents in Cancer Therapeutics. Anticancer Agents Med Chem 2021; 22:433-468. [PMID: 33687887 DOI: 10.2174/1871520621666210308101029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The global alliance for genomics and healthcare facilities provides innovational solutions to expedite research and clinical practices for complex and incurable health conditions. Precision oncology is an emerging field explicitly tailored to facilitate cancer diagnosis, prevention and treatment based on patients' genetic profile. Advancements in "omics" techniques, next-generation sequencing, artificial intelligence and clinical trial designs provide a platform for assessing the efficacy and safety of combination therapies and diagnostic procedures. METHOD Data were collected from Pubmed and Google scholar using keywords: "Precision medicine", "precision medicine and cancer", "anticancer agents in precision medicine" and reviewed comprehensively. RESULTS Personalized therapeutics including immunotherapy, cancer vaccines, serve as a groundbreaking solution for cancer treatment. Herein, we take a measurable view of precision therapies and novel diagnostic approaches targeting cancer treatment. The contemporary applications of precision medicine have also been described along with various hurdles identified in the successful establishment of precision therapeutics. CONCLUSION This review highlights the key breakthroughs related to immunotherapies, targeted anticancer agents, and target interventions related to cancer signaling mechanisms. The success story of this field in context to drug resistance, safety, patient survival and in improving quality of life is yet to be elucidated. We conclude that, in the near future, the field of individualized treatments may truly revolutionize the nature of cancer patient care.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Sudhanshu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| |
Collapse
|
40
|
Bergamino Sirvén M, Pernas S, Cheang MCU. Lights and Shadows in Immuno-Oncology Drug Development. Cancers (Basel) 2021; 13:691. [PMID: 33572060 PMCID: PMC7915946 DOI: 10.3390/cancers13040691] [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] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 11/16/2022] Open
Abstract
The rapidly evolving landscape of immuno-oncology (IO) is redefining the treatment of a number of cancer types. IO treatments are becoming increasingly complex, with different types of drugs emerging beyond checkpoint inhibitors. However, many of the new drugs either do not progress from phase I-II clinical trials or even fail in late-phase trials. We have identified at least five areas in the development of promising IO treatments that should be redefined for more efficient designs and accelerated approvals. Here we review those critical aspects of IO drug development that could be optimized for more successful outcome rates in all cancer types. It is important to focus our efforts on the mechanisms of action, types of response and adverse events of these novel agents. The use of appropriate clinical trial designs with robust biomarkers of response and surrogate endpoints will undoubtedly facilitate the development and subsequent approval of these drugs. Further research is also needed to establish biomarker-driven strategies to select which patients may benefit from immunotherapy and identify potential mechanisms of resistance.
Collapse
Affiliation(s)
- Milana Bergamino Sirvén
- Clinical Studies and Clinical Trials and Statistics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| | - Sonia Pernas
- Department of Medical Oncology, Catalan Institute of Oncology—ICO, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
- Breast Cancer Group, Institut d’Investigacio Biomedica de Bellvitge—IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Maggie C. U. Cheang
- Clinical Studies and Clinical Trials and Statistics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| |
Collapse
|
41
|
Kappelmann N, Müller-Myhsok B, Kopf-Beck J. Adapting the randomised controlled trial (RCT) for precision medicine: introducing the nested-precision RCT (npRCT). Trials 2021; 22:13. [PMID: 33407710 PMCID: PMC7788903 DOI: 10.1186/s13063-020-04965-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 12/10/2020] [Indexed: 12/01/2022] Open
Abstract
Adaptations to the gold standard randomised controlled trial (RCT) have been introduced to decrease trial costs and avoid high sample sizes. To facilitate development of precision medicine algorithms that aim to optimise treatment allocation for individual patients, we propose a new RCT adaptation termed the nested-precision RCT (npRCT). The npRCT combines a traditional RCT (intervention A versus B) with a precision RCT (stratified versus randomised allocation to A or B). This combination allows online development of a precision algorithm, thus providing an integrated platform for algorithm development and its testing. Moreover, as both the traditional and the precision RCT include participants randomised to interventions of interest, data from these participants can be jointly analysed to determine the comparative effectiveness of intervention A versus B, thus increasing statistical power. We quantify savings of the npRCT compared to two independent RCTs by highlighting sample size requirements for different target effect sizes and by introducing an open-source power calculation app. We describe important practical considerations such as blinding issues and potential biases that need to be considered when designing an npRCT. We also highlight limitations and research contexts that are less suited for an npRCT. In conclusion, we introduce the npRCT as a novel precision medicine trial design strategy which may provide one opportunity to efficiently combine traditional and precision RCTs.
Collapse
Affiliation(s)
- Nils Kappelmann
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80804, Munich, Germany. .,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany.
| | - Bertram Müller-Myhsok
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80804, Munich, Germany.,Department of Health Data Science, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
42
|
Lenze EJ, Nicol GE, Barbour DL, Kannampallil T, Wong AWK, Piccirillo J, Drysdale AT, Sylvester CM, Haddad R, Miller JP, Low CA, Lenze SN, Freedland KE, Rodebaugh TL. Precision clinical trials: a framework for getting to precision medicine for neurobehavioural disorders. J Psychiatry Neurosci 2021; 46:E97-E110. [PMID: 33206039 PMCID: PMC7955843 DOI: 10.1503/jpn.200042] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The goal of precision medicine (individually tailored treatments) is not being achieved for neurobehavioural conditions such as psychiatric disorders. Traditional randomized clinical trial methods are insufficient for advancing precision medicine because of the dynamic complexity of these conditions. We present a pragmatic solution: the precision clinical trial framework, encompassing methods for individually tailored treatments. This framework includes the following: (1) treatment-targeted enrichment, which involves measuring patients' response after a brief bout of an intervention, and then randomizing patients to a full course of treatment, using the acute response to predict long-term outcomes; (2) adaptive treatments, which involve adjusting treatment parameters during the trial to individually optimize the treatment; and (3) precise measurement, which involves measuring predictor and outcome variables with high accuracy and reliability using techniques such as ecological momentary assessment. This review summarizes precision clinical trials and provides a research agenda, including new biomarkers such as precision neuroimaging, transcranial magnetic stimulation-electroencephalogram digital phenotyping and advances in statistical and machine-learning models. Validation of these approaches - and then widespread incorporation of the precision clinical trial framework - could help achieve the vision of precision medicine for neurobehavioural conditions.
Collapse
Affiliation(s)
- Eric J Lenze
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Ginger E Nicol
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Dennis L Barbour
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Thomas Kannampallil
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Alex W K Wong
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Jay Piccirillo
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Andrew T Drysdale
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Chad M Sylvester
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Rita Haddad
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - J Philip Miller
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Carissa A Low
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Shannon N Lenze
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Kenneth E Freedland
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| | - Thomas L Rodebaugh
- From the Washington University School of Medicine, St. Louis, Missouri (Lenze, Nicol, Kannampallil Wong, Piccirillo, Drysdale, Sylvester, Haddad, Miller, Lenze, Freedland); the Washington University McKelvey School of Engineering, St. Louis, MO (Barbour); the University of Pittsburgh, Pittsburgh, PA (Low); and the Washington University School of Arts & Sciences, St. Louis, MO (Rodebaugh)
| |
Collapse
|
43
|
Serra O, Smyth EC, Lordick F. Progress and challenges in gastroesophageal cancer. Curr Probl Cancer 2020; 44:100590. [DOI: 10.1016/j.currproblcancer.2020.100590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022]
|
44
|
Lin R, Thall PF, Yuan Y. BAGS: A Bayesian Adaptive Group Sequential Trial Design With Subgroup-Specific Survival Comparisons. J Am Stat Assoc 2020; 116:322-334. [DOI: 10.1080/01621459.2020.1837142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Peter F. Thall
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
45
|
Basket trials: From tumour gnostic to tumour agnostic drug development. Cancer Treat Rev 2020; 90:102082. [DOI: 10.1016/j.ctrv.2020.102082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
|
46
|
Brierley CK, Zabor EC, Komrokji RS, DeZern AE, Roboz GJ, Brunner AM, Stone RM, Sekeres MA, Steensma DP. Low participation rates and disparities in participation in interventional clinical trials for myelodysplastic syndromes. Cancer 2020; 126:4735-4743. [PMID: 32767690 DOI: 10.1002/cncr.33105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND The development of novel therapies for the myelodysplastic syndromes (MDS) is hampered by inadequate trial recruitment. Factors contributing to low trial accrual are incompletely understood. METHODS This study analyzed a pooled patient database from institutions of the US MDS Clinical Research Consortium to compare the characteristics of participants in interventional trials with those of patients who did not enroll in a trial. RESULTS Data were identified for 1919 patients with MDS, and 449 of these patients (23%) participated in an interventional clinical trial. The median age of all patients was 68 years, and 64% were male. Patients who participated in trials were significantly younger than nonparticipants (P = .014), and men were more likely to participate in a trial (71% of trial participants were male, whereas 61% of nonparticipants were; P < .001). Race and ethnicity were not associated with trial enrollment. Patients in more affluent ZIP codes had a higher participation rate (P < .001). Patients with intermediate- and high-risk disease according to the revised International Prognostic Scoring System were overrepresented (P = .004), and trial participants less frequently had treatment-related disease (P < .001). In multivariable analyses, participation in a clinical trial was associated with a reduced hazard of death (P = .004). Even at large referral centers, only a minority of patients with MDS enrolled in interventional trials. CONCLUSIONS Restrictive trial eligibility criteria that exclude patients with MDS on account of age, comorbidities, or a history of another cancer are limit enrollment of MDS patients to clinical trials. Gaining insight into the barriers to trial accrual may help investigators and study sponsors to design trials that will accrue more rapidly and augment treatment options for patients with MDS.
Collapse
Affiliation(s)
| | - Emily C Zabor
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Rami S Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Gail J Roboz
- Weill Cornell Medical College, New York, New York
| | - Andrew M Brunner
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard M Stone
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mikkael A Sekeres
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - David P Steensma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Wang Y, Park JYP, Pacis A, Denroche RE, Jang GH, Zhang A, Cuggia A, Domecq C, Monlong J, Raitses-Gurevich M, Grant RC, Borgida A, Holter S, Stossel C, Bu S, Masoomian M, Lungu IM, Bartlett JM, Wilson JM, Gao ZH, Riazalhosseini Y, Asselah J, Bouganim N, Cabrera T, Boucher LM, Valenti D, Biagi J, Greenwood CM, Polak P, Foulkes WD, Golan T, O'Kane GM, Fischer SE, Knox JJ, Gallinger S, Zogopoulos G. A Preclinical Trial and Molecularly Annotated Patient Cohort Identify Predictive Biomarkers in Homologous Recombination–deficient Pancreatic Cancer. Clin Cancer Res 2020; 26:5462-5476. [DOI: 10.1158/1078-0432.ccr-20-1439] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/24/2020] [Accepted: 08/03/2020] [Indexed: 12/27/2022]
Abstract
Abstract
Purpose:
Pancreatic ductal adenocarcinoma (PDAC) arising in patients with a germline BRCA1 or BRCA2 (gBRCA) mutation may be sensitive to platinum and PARP inhibitors (PARPi). However, treatment stratification based on gBRCA mutational status alone is associated with heterogeneous responses.
Experimental Design:
We performed a seven-arm preclinical trial consisting of 471 mice, representing 12 unique PDAC patient-derived xenografts, of which nine were gBRCA mutated. From 179 patients whose PDAC was whole-genome and transcriptome sequenced, we identified 21 cases with homologous recombination deficiency (HRD), and investigated prognostic biomarkers.
Results:
We found that biallelic inactivation of BRCA1/BRCA2 is associated with genomic hallmarks of HRD and required for cisplatin and talazoparib (PARPi) sensitivity. However, HRD genomic hallmarks persisted in xenografts despite the emergence of therapy resistance, indicating the presence of a genomic scar. We identified tumor polyploidy and a low Ki67 index as predictors of poor cisplatin and talazoparib response. In patients with HRD PDAC, tumor polyploidy and a basal-like transcriptomic subtype were independent predictors of shorter survival. To facilitate clinical assignment of transcriptomic subtype, we developed a novel pragmatic two-marker assay (GATA6:KRT17).
Conclusions:
In summary, we propose a predictive and prognostic model of gBRCA-mutated PDAC on the basis of HRD genomic hallmarks, Ki67 index, tumor ploidy, and transcriptomic subtype.
Collapse
Affiliation(s)
- Yifan Wang
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jin Yong Patrick Park
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alain Pacis
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 3Canadian Centre for Computational Genomics, McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | | | - Gun Ho Jang
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Amy Zhang
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Adeline Cuggia
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Celine Domecq
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jean Monlong
- 5Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Maria Raitses-Gurevich
- 6Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Robert C. Grant
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Ayelet Borgida
- 8Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Spring Holter
- 8Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Chani Stossel
- 6Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- 9Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Simeng Bu
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Mehdi Masoomian
- 10Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ilinca M. Lungu
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - John M.S. Bartlett
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 10Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Julie M. Wilson
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Zu-Hua Gao
- 11Department of Pathology, McGill University, Montreal, Quebec, Canada
| | | | - Jamil Asselah
- 12Department of Oncology, McGill University, Montreal, Quebec, Canada
| | | | - Tatiana Cabrera
- 13Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - Louis-Martin Boucher
- 13Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - David Valenti
- 13Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - James Biagi
- 14Department of Oncology, Queen's University, Kingston, Ontario, Canada
| | - Celia M.T. Greenwood
- 5Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- 12Department of Oncology, McGill University, Montreal, Quebec, Canada
- 15Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
- 16Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Paz Polak
- 17Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - William D. Foulkes
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- 5Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Talia Golan
- 6Pancreatic Cancer Translational Research Laboratory, Oncology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- 9Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Grainne M. O'Kane
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Sandra E. Fischer
- 10Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer J. Knox
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Steven Gallinger
- 4Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- 7Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - George Zogopoulos
- 1Rosalind and Morris Goodman Cancer Research Centre of McGill University, Montreal, Quebec, Canada
- 2Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
48
|
Parasrampuria DA, Bandekar R, Puchalski TA. Scientific diligence for oncology drugs: a pharmacology, translational medicine and clinical perspective. Drug Discov Today 2020; 25:1855-1864. [DOI: 10.1016/j.drudis.2020.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/02/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
|
49
|
Patterson C, Barber FD. Clinical Trial Subinvestigator: An Emerging Role for Oncology Nurse Practitioners. Clin J Oncol Nurs 2020; 24:479-481. [PMID: 32945784 DOI: 10.1188/20.cjon.479-481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Phase 1 clinical trials are essential to improving outcomes in cancer care. The investigational agents in these trials may be associated with adverse events that can contribute to symptom burden and declining performance status for trial participants. The emerging role for oncology nurse practitioners (ONPs) as subinvestigators offers a unique practice setting for advanced practice nurses. In this role, ONPs provide expert oncology care, are responsible for swift recognition and management of adverse events, and ensure adherence to the clinical trial protocol.
Collapse
|
50
|
Wallerstedt SM, Nilsson Ek A, Olofsson Bagge R, Kovács A, Strandell A, Linderholm B. Personalised medicine and the decision to withhold chemotherapy in early breast cancer with intermediate risk of recurrence - a systematic review and meta-analysis. Eur J Clin Pharmacol 2020; 76:1199-1211. [PMID: 32504183 PMCID: PMC7419442 DOI: 10.1007/s00228-020-02914-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE To assess the evidence for decision making, at the health care and the patient levels, regarding the use of gene expression assays to inform chemotherapy decisions in breast cancer patients with intermediate clinical risk of recurrence. METHODS Systematic literature searches were performed (January 2002-April 2020) in Medline, Embase, PubMed, Cochrane Library, PsycINFO and HTA databases. INCLUSION CRITERIA patients (P) were individuals with post-surgical breast cancer at intermediate clinical risk of recurrence; intervention (I)/comparison (C) was (i) use of, versus no use of, a gene expression assay and (ii) withholding versus providing chemotherapy; outcomes (O) were overall survival (OS), health-related quality of life (HRQL), and recurrence. Randomised controlled trials (RCTs) and non-RCTs were included. Random-effects meta-analyses were performed where possible. RESULTS Three inconclusive non-RCTs, respectively, compared OS and recurrence with and without a gene expression assay. No studies investigated HRQL. Regarding the comparison withholding versus providing chemotherapy based on a gene expression assay, one RCT and four non-RCTs evaluated OS. In the RCT, 93.9% (I) versus 93.8% (C) were alive at 9 years. Three RCTs and seven non-RCTs evaluated recurrence. Three RCTs could be pooled regarding distant recurrence; 4.29% versus 3.88% had such an event (risk ratio: 1.12 (95% confidence interval: 0.90 to 1.39). CONCLUSION Regarding the use of gene expression assays in breast cancer, evidence on patient effects, informing patient-level chemotherapy decision making, is available. However, evidence for prioritisation at the overall health care level, i.e. use of, versus no use of, such assays, is largely lacking.
Collapse
Affiliation(s)
- Susanna M Wallerstedt
- HTA-centrum, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Pharmacology, Sahlgrenska Academy, University of Gothenburg, Box 431, SE-405 30, Gothenburg, Sweden.
| | - Astrid Nilsson Ek
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Sahlgrenska Cancer Center, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Annika Strandell
- HTA-centrum, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Barbro Linderholm
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|