1
|
Hu X, Duan Z, Li X, Cao H, Zhao H, Zhang Y. Symptom and functional networks of patients with cancer in different latent risk subgroups based on patient-reported outcomes. BMC Cancer 2025; 25:872. [PMID: 40369473 PMCID: PMC12079923 DOI: 10.1186/s12885-025-14256-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Currently, risk stratification and effective management of heterogeneous patients with cancer based on patient-reported outcomes (PROs), used to evaluate clinical efficacy and outcomes, are relatively rare and urgently needed. We aimed to explore latent risk subgroups and delineate multidimensional networks of symptoms and functions based on PROs in this study. METHODS Patients with cancer were recruited from eight hospitals in two Provinces in China. The PROs measure for patients with cancer (CA-PROM) was used to measure patients' HRQoL, symptoms, and functions. Latent profile analysis (LPA) was used to explore latent risk subgroups using four fitting indicators on the patients' HRQoL. Network model (NM) of multidimensional symptoms and functions was applied at the item level of the CA-PROM. The expected influence (EI), bridge EI, and predictability of each node were used to evaluate the centrality and predictability of NM. Network accuracy and stability were tested using a case-dropping bootstrap procedure. Finally, a network comparison test (NCT) was conducted to examine whether network characteristics differed among the various risk subgroups. RESULTS In total, 1,404 valid questionnaires were collected. Three latent risk subgroups were determined based on the four fitting indicators. Considering the mean difference in HRQoL, subgroups 1, 2, and 3 were indicated as high-risk (n = 196), low-risk (n = 716), and medium-risk (n = 492) subgroups, respectively. There were statistically significant differences in most demographic data, disease conditions, and treatment among three latent risk subgroups. Network analysis revealed that some symptoms and functions (e.g., despair, gastrointestinal abnormalities, care and support from their families and friends, appetite, and so on) played more important roles in the heterogeneity of HRQoL for Chinese patients w ith cancer. But the performance of these symptoms and functions reported by patients varied among three subgroups. Network accuracy and stability basically met the preset criteria. NCT results showed that edge differences were observed in five nodes, and seven nodes with different EI values could be informative for targeted support for the patients of different clusters. CONCLUSION Different central and bridge symptoms or functions in multidimensional networks of PROs may serve as potential targets for personalized interventions among patients with cancer who are at different risk levels of HRQoL.
Collapse
Affiliation(s)
- Xiaojuan Hu
- Department of Medical Information Technology, School of Management, Shanxi Medical University, No. 56 South XinJian Road, Taiyuan, Shanxi Province, 030001, China
- Shanxi Provincial Key Laboratory of Major Diseases Risk Assessment, Taiyuan, China
| | - Zhihui Duan
- Department of Thoracic Surgery, Shanxi Cancer Hospital, No. 3 Workers' New Village, Taiyuan, Shanxi Province, 030013, China
| | - Xiaokun Li
- Department of Neurosurgery, Shanxi Cancer Hospital, No. 3 Workers' New Village, Taiyuan, Shanxi Province, 030013, China
| | - Hongyan Cao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, No. 56 South XinJian Road, Taiyuan, Shanxi Province, 030001, China
| | - Hui Zhao
- Department of Respiratory Medicine, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi Province, 030001, China
| | - Yanbo Zhang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, No. 56 South XinJian Road, Taiyuan, Shanxi Province, 030001, China.
- Shanxi Provincial Key Laboratory of Major Diseases Risk Assessment, Taiyuan, China.
| |
Collapse
|
2
|
Garcia-Foncillas J, Bayle A, Arnold D, Avouac B, Awada A, de la Cruz-Merino L, Helland Å, Lassen U, Laurent-Puig P, Normanno N, Rohrberg K, Taieb J, Stenzinger A. Overcoming barriers to advanced biomolecular technologies that inform treatment of solid tumors: a roadmap to access. Future Oncol 2025:1-8. [PMID: 40340714 DOI: 10.1080/14796694.2025.2501523] [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: 08/09/2024] [Accepted: 04/30/2025] [Indexed: 05/10/2025] Open
Abstract
The advent of advanced biomolecular technologies for detecting molecular and genomic signatures of individual tumors has transformed oncology care, introducing proven methodologies that can inform treatment with matched targeted therapies and predict response at the individual patient level. However, access to these technologies has been hampered by multiple barriers, most notably price and obtainability. Other barriers include lack of knowledge of available technologies, concerns about value, and outdated infrastructures that impede critical operations within the clinic or laboratory. Accessibility barriers to advanced biomolecular testing are critically important to patient care, as new technological advances in molecular medicine continue to outpace the implementation of solutions. Given the proven evidence for improved patient outcomes with precision oncology medicines, it is imperative to understand the value afforded by these technologies. The purpose of this narrative review is to describe existing and emerging barriers to access and present a "roadmap to access" that will facilitate the urgently needed discussions to identify solutions for improving access. Implementation of these solutions will raise awareness of available technologies and treatments and their prognostic significance, improve evidence collection for demonstration of value, and fortify clinical and laboratory infrastructure and operations.
Collapse
Affiliation(s)
- Jesús Garcia-Foncillas
- University Cancer Institute & The Department of Oncology, University Hospital Fundacion Jimenez Diaz, Autonomous University, Madrid, Spain
| | - Arnaud Bayle
- Bureau Biostatistique et Epidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Gustave Roussy, INSERM, Université Paris-Saclay, CESP U1018 Oncostat, Labelisé Ligue Contre le Cancer, Villejuif, France
| | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| | | | - Ahmad Awada
- Oncology Medicine Department, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Luis de la Cruz-Merino
- Cancer Immunotherapy, Biomedicine Institute of Seville (IBIS)/CSIC, Clinical Oncology Department, University Hospital Virgen Macarena and School of Medicine, University of Seville, Seville, Spain
| | - Åslaug Helland
- Division of Cancer Medicine, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Ulrik Lassen
- Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Nicola Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | - Julien Taieb
- GI Oncology Department, Georges Pompidou European Hospital SIRIC-CARPEM, Université Paris Cité, Paris, France
| | - Albrecht Stenzinger
- Institute of Pathology and Center for Personalized Medicine (ZPM), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
3
|
Passiglia F, Listì A, Bironzo P, Merlini A, Benso F, Napoli F, Barbu FA, Zambelli V, Tabbò F, Reale ML, Sini C, Roca E, Taveggia PA, Simionato F, Buffoni L, Mazilu L, Barbieri V, Pignataro D, Araujo A, Paz-Ares L, Felip E, Secen N, Comanescu A, Ramizi KM, Bettini AC, Scotti V, Linardou H, Mohorcic K, Meoni G, Giannarelli D, Volante M, Malapelle U, Vallone S, Scagliotti G, Righi L, Novello S. Actionable NSCLC Mutation Identification by Comprehensive Genomic Profiling for Clinical Trial Enrollment: The European Program for the Routine Testing of Patients With Advanced Lung Cancer (EPROPA). J Thorac Oncol 2025; 20:614-624. [PMID: 39694416 DOI: 10.1016/j.jtho.2024.12.010] [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: 06/03/2024] [Revised: 11/15/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION The advocacy Women Against Lung Cancer in Europe (WALCE) promoted the European Program for the Routine Testing of Patients With Advanced Lung Cancer (EPROPA) and provided a free-of-charge molecular profiling platform for NSCLC sample characterization with the aim of increasing the detection of targetable drivers and improving patients' access to clinical trials in Europe. METHODS From January 2021 to December 2023, 20 centers located at five different European countries (Greece, Slovenia, Romania, Albania, and Italy) joined EPROPA, with 555 patients with advanced NSCLC registered to the program. Anonymized patients' clinical-pathological data were shared through the EPROPA web platform and tissue samples were collected at the Molecular Pathology Unit of the Reference Center (University of Turin) for molecular analyses. A comprehensive genomic profiling by a targeted next-generation sequencing approach has been performed and molecular reports have been discussed within the molecular tumor board to assess patients' eligibility for clinical trials. RESULTS The average turnaround time was eight days, with only 30 out of 555 tissue samples (6%) not suitable for molecular analysis. In the 525 analyzed samples, a total of 570 molecular alterations have been identified, including 264 pathogenic targetable oncogenic alterations and 113 cases with co-occurring mutations. A total of 18 molecular alterations with potential germline and hereditary cancer syndrome implications have been reported. The identification of a clinical trial was considered for 205 patients. After molecular tumor board discussion, 30 patients were enrolled and treated in clinical studies available in Europe. Survival outcomes were significantly improved in patients with targetable molecular alterations receiving a matched targeted therapy. CONCLUSION This data confirmed the feasibility and usefulness of the program in the real-world practice scenario, supporting the implementation of next-generation sequencing-based molecular characterization of NSCLC samples, to reduce the unequal access to tests, drugs, and clinical trials in Europe.
Collapse
Affiliation(s)
- Francesco Passiglia
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Angela Listì
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Paolo Bironzo
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Federica Benso
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Francesca Napoli
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Francesca Alice Barbu
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Vanessa Zambelli
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Fabrizio Tabbò
- Medical Oncology, ASLCN2 Alba e Bra, Michele e Pietro Ferrero Hospital, Verduno, Cuneo, Italy
| | | | - Claudio Sini
- Medical Oncology and CPDO, Giovanni Paolo II Hospital, Olbia, Italy
| | - Elisa Roca
- Thoracic Oncology - Lung Unit P. Pederzoli Hospital, Peschiera d/G, Verona, Italy
| | | | | | - Lucio Buffoni
- Medical Oncology, Humanitas Gradenigo Hospital, Torino, Italy
| | - Laura Mazilu
- Medical Oncology, Ovidius Clinical Hospital, Ovidius University of Constanta, Romania
| | - Vito Barbieri
- Medical Oncology, Renato Dulbecco University Hospital, Catanzaro, Italy
| | | | - Antonio Araujo
- Department of Medical Oncology, Centro Hospitalar Universitário de Santo António, Portugal
| | - Luis Paz-Ares
- Medical Oncology Department, University Hospital 12 De Octubre, Madrid, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Nevena Secen
- Pulmonology-Oncology Department, AcibademBelmedic, Belgrade, Serbia
| | | | | | | | - Vieri Scotti
- Department of Radiotherapy, AOU Careggi, Firenze, Italy
| | - Helena Linardou
- Fourth Oncology Department and Comprehensive Clinical Trials Center, Metropolitan Hospital, Athens, Greece
| | - Katja Mohorcic
- Medical Oncology Department, University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | - Giulia Meoni
- SOS Medical Oncology, San Giovanni di Dio Hospital, Firenze, Italy
| | - Diana Giannarelli
- Fondazione Policlinico Universitario A. Gemelli, IRCCS - Facility of Epidemiology and Biostatistics, Rome, Italy
| | - Marco Volante
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | | | - Giorgio Scagliotti
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Luisella Righi
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy.
| |
Collapse
|
4
|
Serna-Blasco R, Mediavilla-Medel P, Medina K, Sala MÁ, Aguiar D, Díaz-Serrano A, Antoñanzas M, Ocaña J, Mielgo X, Fernández I, López-Castro R, Cobo M, Martínez M, Villa JC, Rosado P, López A, Guirado M, Viteri S, Rodríguez D, García F, Simón S, Moreno MÁ, Catot S, González Larriba JL, Salas C, Calvo V, Romero A, Provencio M. Comprehensive molecular profiling of advanced NSCLC using NGS: Prevalence of druggable mutations and clinical trial opportunities in the ATLAS study. Lung Cancer 2025; 204:108550. [PMID: 40300279 DOI: 10.1016/j.lungcan.2025.108550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND In Spain, next-generation sequencing (NGS) is currently available in a limited number of specialized centers and remains inaccessible to a significant proportion of patients. The ATLAS study aims to explore the tumor molecular profile beyond known EGFR mutations and ALK translocations using NGS on tumor biopsy samples. METHODS Patients with EGFR-sensitizing mutations or ALK translocations were excluded. A total of 455 patients with advanced non-small cell lung cancer (NSCLC) were enrolled from 22 Spanish hospitals. DNA and RNA were extracted from formalin-fixed paraffin-embedded samples, and libraries were prepared using the Oncomine Focus Assay. The Trialing app was used to identify active clinical trials in Spain. RESULTS Mutations were detected in 65.7 % of the cases. Local pathology assessments detected druggable mutations in only 7.9 % of cases, while centralized NGS testing increased this detection rate to 25.9 %. The most prevalent druggable alteration was KRAS G12C (53.6 %), followed by MET amplification (8.1 %) and MET exon 14 skipping (7.3 %). Additionally, 34.5 % of patients had molecular alterations matching clinical trials, offering potential treatment opportunities. Women had a significantly higher probability of harbouring druggable mutations (36 % vs. 20.3 %, p < 0.001), including the KRAS G12C which was significantly more frequent in females (22.6 % vs. 10 %). KRAS mutations were more common in adenocarcinomas and increased with tumor differentiation grade (p < 0.001 and p = 0.049, respectively). Likewise, ALK translocations, EGFR mutations, BRAF V600E, MET exon 14 skipping, and RET/ROS1 fusions were mainly found in adenocarcinomas whereas copy number variations were more frequent in squamous carcinomas (28.6 % vs. 15.1 %; p = 0.003) and in men (22 % vs. 11.6 %; p = 0.008). CONCLUSION The ATLAS study demonstrates the utility of comprehensive NGS testing, which detects clinically relevant mutations in more than one-third of patients and may extend therapy options.
Collapse
Affiliation(s)
- Roberto Serna-Blasco
- Liquid Biopsy Laboratory, Medical Oncology Department. Instituto de Investigación Biomédica Puerta de Hierro - Segovia de Arana, Majadahonda, Comunidad de Madrid, Spain
| | - Pilar Mediavilla-Medel
- Liquid Biopsy Laboratory, Medical Oncology Department. Instituto de Investigación Biomédica Puerta de Hierro - Segovia de Arana, Majadahonda, Comunidad de Madrid, Spain
| | - Karla Medina
- Department of Oncology, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Islas Canarias, Spain
| | - María Ángeles Sala
- Department of Oncology, Hospital Universitario Basurto, Bilbao, País Vasco, Spain
| | - David Aguiar
- Department of Oncology, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Islas Canarias, Spain
| | - Asunción Díaz-Serrano
- Complejo Público Asistencial de Zamora, Hospital Virgen de la Concha, Zamora, Castilla y León, Spain
| | - Mónica Antoñanzas
- Department of Oncology, Hospital Universitario Clínico San Carlos, Madrid, Comunidad de Madrid, Spain
| | - Julio Ocaña
- Department of Oncology, Hospital Sanitas CIMA, Barcelona, Cataluña, Spain
| | - Xabier Mielgo
- Department of Oncology, Hospital Universitario Fundación Alcorcón, Alcorcón, Comunidad de Madrid, Spain
| | - Inmaculada Fernández
- Department of Oncology, Hospital Universitario Virgen Macarena, Sevilla, Andalucía, Spain
| | - Rafael López-Castro
- Department of Oncology, Hospital Clínico Universitario de Valladolid, Valladolid, Castilla y León, Spain
| | - Manuel Cobo
- Department of Oncology, Hospital Regional Universitario de Málaga, Málaga, Andalucía, Spain
| | - Mireia Martínez
- Department of Oncology, Hospital Universitario de Álava - Txagorritxu, Vitoria, País Vasco, Spain
| | - José Carlos Villa
- Department of Oncology, Hospital General Universitario de Ciudad Real, Ciudad Real, Castilla-La Mancha, Spain
| | - Petra Rosado
- Department of Oncology, Hospital Universitario Puerto Real, Puerto Real, Andalucía, Spain
| | - Ana López
- Department of Oncology, Hospital Universitario Severo Ochoa, Leganés, Comunidad de Madrid, Spain
| | - María Guirado
- Department of Oncology, Hospital General Universitario de Elche, Elche, Conunidad Valenciana, Spain
| | - Santiago Viteri
- UOMI Cancer Center, Clínica Mis Tres Torres Barcelona, Cataluña, Spain
| | - Delvys Rodríguez
- Department of Oncology, Hospital Universitario Insular de Gran Canaria, Islas Canarias, Spain
| | - Florencia García
- Department of Oncology, Hospital Quirónsalud Barcelona, Barcelona, Cataluña, Spain
| | - Soraya Simón
- Department of Oncology, Hospital Virgen de la Luz, Cuenca, Castilla-La Mancha, Spain
| | - María Ángeles Moreno
- Department of Oncology, Hospital Universitario De Jerez, Jerez de la Frontera, Andalucía, Spain
| | | | | | - Clara Salas
- Pathology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Virginia Calvo
- Department of Oncology Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Atocha Romero
- Liquid Biopsy Laboratory, Medical Oncology Department. Instituto de Investigación Biomédica Puerta de Hierro - Segovia de Arana, Majadahonda, Comunidad de Madrid, Spain; Department of Oncology Hospital Universitario Puerta de Hierro, Majadahonda, Spain.
| | - Mariano Provencio
- Department of Oncology Hospital Universitario Puerta de Hierro, Majadahonda, Spain.
| |
Collapse
|
5
|
Belcaid L, Højgaard M, Tuxen I, Spanggaard I, Lassen U, Robinson L, Rossing M, Bagger F, Ahlborn LB, Schmidt AY, Hasselby JP, Santoni-Rugiu E, Nielsen FC, Yde CW, Rohrberg K. Copenhagen Prospective Personalized Oncology (CoPPO) - Impact of comprehensive genomic profiling in more than 2000 patients in a Phase I setting. Ann Oncol 2025:S0923-7534(25)00158-9. [PMID: 40246201 DOI: 10.1016/j.annonc.2025.04.004] [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: 11/15/2024] [Revised: 02/28/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Targeted therapy based on the molecular characterization of tumors has been among the most remarkable advances in cancer medicine. Here, we report the impact of almost 10 years of comprehensive genomic profiling in more than 2000 patients with advanced solid tumors at the Phase 1 unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark. MATERIALS AND METHODS A prospective, single-center, single-arm open-label study (NCT02290522) was conducted, enrolling patients with advanced cancer referred to a Phase I Unit. Fresh tumor tissue was obtained for whole genome or exome sequencing (germline and somatic), RNA sequencing and SNP array. In cases where fresh tumor tissue was unavailable, archived formalin-fixed paraffin-embedded tumor tissue or circulating tumor DNA extracted from plasma were obtained for targeted panels. Genomic reports were reviewed and discussed by a multidisciplinary tumor board and actionable alterations were classified according to the ESMO scale for the clinical actionability of molecular targets (ESCAT). When possible, patients were treated with regimen matched to the genomic profile. RESULTS A total of 2147 patients with advanced cancer and exhausted treatment options were enrolled from April 2013 to December 2021. Genomic profiles were obtained in 1866 patients (87%). At least one actionable target was identified in 1062 patients (57%) with a total of 1614 actionable alterations including high RNA CEACAM5 expression (N=559, 35%), homologous recombination deficiency (HRD) alteration (N=314, 20%) and tumor mutational burden-high (N=84, 5%). Overall, 256 patients (24% of the patients with an actionable target) were treated with targeted therapy and among these 14 patients were treated with more than one line of targeted therapy. In total, 274 targeted treatment regimens were initiated, and 259 treatments were evaluable with an overall response (OR) rate of 25% (CI95%: 0.20 - 0.30). ESCAT I/II was associated with improved OR, along with better progression-free survival (PFS) and overall survival (OS). CONCLUSION This large-scale study demonstrates that genomic profiling is efficient to identify actionable targets and to match patients to targeted therapies.
Collapse
Affiliation(s)
- L Belcaid
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark.
| | - M Højgaard
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - I Tuxen
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - I Spanggaard
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - U Lassen
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - L Robinson
- Center for Genomic Medicine, Rigshospitalet, Copenhagen, Denmark
| | - M Rossing
- Center for Genomic Medicine, Rigshospitalet, Copenhagen, Denmark
| | - F Bagger
- Center for Genomic Medicine, Rigshospitalet, Copenhagen, Denmark
| | - L B Ahlborn
- Center for Genomic Medicine, Rigshospitalet, Copenhagen, Denmark
| | - A Y Schmidt
- Center for Genomic Medicine, Rigshospitalet, Copenhagen, Denmark
| | - J P Hasselby
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - E Santoni-Rugiu
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - F C Nielsen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen, Denmark
| | - C W Yde
- Center for Genomic Medicine, Rigshospitalet, Copenhagen, Denmark
| | - K Rohrberg
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
6
|
Kotsifa E, Saffioti F, Mavroeidis VK. Cholangiocarcinoma: The era of liquid biopsy. World J Gastroenterol 2025; 31:104170. [PMID: 40124277 PMCID: PMC11924015 DOI: 10.3748/wjg.v31.i11.104170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 03/13/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive and heterogeneous malignancy arising from the epithelial cells of the biliary tract. The limitations of the current methods in the diagnosis of CCA highlight the urgent need for new, accurate tools for early cancer detection, better prognostication and patient monitoring. Liquid biopsy (LB) is a modern and non-invasive technique comprising a diverse group of methodologies aiming to detect tumour biomarkers from body fluids. These biomarkers include circulating tumour cells, cell-free DNA, circulating tumour DNA, RNA and extracellular vesicles. The aim of this review is to explore the current and potential future applications of LB in CCA management, with a focus on diagnosis, prognostication and monitoring. We examine both its significant potential and the inevitable limitations associated with this technology. We conclude that LB holds considerable promise, but further research is necessary to fully integrate it into precision oncology for CCA.
Collapse
Affiliation(s)
- Evgenia Kotsifa
- The Second Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, Athens 11527, Greece
| | - Francesca Saffioti
- Department of Gastroenterology and Hepatology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
- University College London Institute for Liver and Digestive Health and Sheila Sherlock Liver Unit, Royal Free Hospital and University College London, London NW3 2QG, United Kingdom
- Division of Clinical and Molecular Hepatology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina 98124, Italy
| | - Vasileios K Mavroeidis
- Department of Transplant Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of Gastrointestinal Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of HPB Surgery, Bristol Royal Infirmary, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS2 8HW, United Kingdom
| |
Collapse
|
7
|
Volders PJ, Aftimos P, Dedeurwaerdere F, Martens G, Canon JL, Beniuga G, Froyen G, Van Huysse J, De Pauw R, Prenen H, Lambin S, Decoster L, Vaeyens F, Rottey S, Van Dam PJ, Decoster L, Rutten A, Schreuer M, Loontiens S, Van der Meulen J, Mebis J, Cuppens K, Tejpar S, Vanden Bempt I, De Grève J, Schröder D, van Marcke C, Van Den Bulcke M, de Azambuja E, Punie K, Maes B. A nationwide comprehensive genomic profiling and molecular tumor board platform for patients with advanced cancer. NPJ Precis Oncol 2025; 9:66. [PMID: 40065106 PMCID: PMC11893761 DOI: 10.1038/s41698-025-00858-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
The Belgian Approach for Local Laboratory Extensive Tumor Testing (BALLETT) study assessed the feasibility of using comprehensive genomic profiling (CGP) in clinical decision-making for patients with advanced cancers. This multi-center study enrolled 872 patients from 12 Belgian hospitals. CGP was performed on tumor tissues using a standardized CGP panel (523 genes) across nine laboratories with success in 93% of patients and a median turnaround time of 29 days. Actionable genomic markers were identified in 81% of patients, substantially higher than the 21% using nationally reimbursed, small panels. A national molecular tumor board (nMTB) recommended treatments for 69% of patients, with 23% receiving matched therapies. Reasons for non-compliance were highly variable across clinical sites. Overall, BALLETT demonstrates the feasibility of implementing decentralized CGP and its potential to identify actionable targets in most patients with advanced cancers. BALLETT reinforces CGP's utility and emphasizes the importance of collaboration, standardization, and addressing implementation challenges.
Collapse
Affiliation(s)
- Pieter-Jan Volders
- Laboratory for Molecular Diagnostics, Jessa Hospital, Hasselt, Belgium
- Faculty of Medicine and Life Sciences, LCRC, University of Hasselt, Hasselt, Belgium
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
| | - Philippe Aftimos
- Clinical Trials Conduct Unit, Institut Jules Bordet-Université Libre de Bruxelles, Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | | | - Geert Martens
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Jean-Luc Canon
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi, Belgium
| | | | - Guy Froyen
- Laboratory for Molecular Diagnostics, Jessa Hospital, Hasselt, Belgium
- Faculty of Medicine and Life Sciences, LCRC, University of Hasselt, Hasselt, Belgium
| | | | - Rebecca De Pauw
- Department of Pulmonology, AZ Sint-Jan Brugge, Bruges, Belgium
| | - Hans Prenen
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium
- Center for Oncological research, University of Antwerp, Wilrijk, Belgium
| | - Suzan Lambin
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Lore Decoster
- Department of Medical Oncology, University Hospital Brussels, Brussels, Belgium
- Laboratory for Medical and Molecular Oncology, Vrije Universiteit, Brussels, Belgium
| | - Freya Vaeyens
- Centre for Medical Genetics, University Hospital Brussels, Brussels, Belgium
| | - Sylvie Rottey
- Department of Medical Oncology, Ghent University, Ghent, Belgium
| | | | - Lynn Decoster
- Department of Pulmonology, AZ Turnhout, Turnhout, Belgium
| | - Annemie Rutten
- Department of Medical Oncology, ZAS hospitals, Antwerp, Belgium
| | - Max Schreuer
- Department of Medical Oncology, ASZ Aalst, Aalst, Belgium
| | - Siebe Loontiens
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
- Molecular Diagnostics, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Joni Van der Meulen
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
- Molecular Diagnostics, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Jeroen Mebis
- Faculty of Medicine and Life Sciences, LCRC, University of Hasselt, Hasselt, Belgium
- Department of Medical Oncology, Jessa Hospital, Hasselt, Belgium
| | - Kristof Cuppens
- Faculty of Medicine and Life Sciences, LCRC, University of Hasselt, Hasselt, Belgium
- Department of Pulmonology and Thoracic Oncology, Jessa Hospital, Hasselt, Belgium
| | - Sabine Tejpar
- Digestive Oncology, University Hospitals KU Leuven, Leuven, Belgium
| | | | - Jacques De Grève
- Laboratory for Medical and Molecular Oncology, Vrije Universiteit, Brussels, Belgium
- Centre for Medical Genetics, University Hospital Brussels, Brussels, Belgium
| | - David Schröder
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Cédric van Marcke
- Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Medical Oncology, Institut Roi Albert II, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | - Evandro de Azambuja
- Institut Jules Bordet, Hôpital Universitaire de Bruxelles and l'Université Libre de Bruxelles, Brussels, Belgium
| | - Kevin Punie
- Department of Medical Oncology, ZAS hospitals, Antwerp, Belgium
| | - Brigitte Maes
- Laboratory for Molecular Diagnostics, Jessa Hospital, Hasselt, Belgium.
- Faculty of Medicine and Life Sciences, LCRC, University of Hasselt, Hasselt, Belgium.
| |
Collapse
|
8
|
Cierzniak A, Małodobra-Mazur M, Tokarski M. A new approach for the detection of genetic alterations utilizing modified loop-mediated isothermal amplification reaction (LAMP). Sci Rep 2025; 15:8071. [PMID: 40055506 PMCID: PMC11889157 DOI: 10.1038/s41598-025-93086-2] [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: 01/10/2025] [Accepted: 03/04/2025] [Indexed: 05/13/2025] Open
Abstract
The increasing use of genetic testing for personalised therapy, highlights the need for rapid, reliable diagnostics. Current methods are hindered by complex workflows, requiring advanced equipment, skilled personnel, and invasive tissue sampling. Loop-mediated isothermal amplification (LAMP) has emerged as a more efficient alternative to traditional PCR. LAMP eliminates thermal cycling, allowing faster, more cost-effective tests, and is less sensitive to inhibitors, enabling testing from minimally processed samples. Although LAMP is newer and has a longer assay development time than PCR, its potential in oncology, particularly for detecting genetic changes, is promising. We have developed a LAMP-based method for detecting genetic variations, optimized for point-of-care testing. This technique uses modified primers with alterations at the 3' end of either F2 or B2 primers, ensuring specificity for altered sequences. The assay only produces a positive signal when the genetic variant is present, distinguishing it from wild-type DNA. Our findings demonstrate that this method has high specificity and sensitivity, even in samples with both wild-type and mutated material. Paired with a portable device, this LAMP-based diagnostic method could revolutionize genetic alteration detection, offering quicker results and improving treatment outcomes, particularly for targeted therapies.
Collapse
Affiliation(s)
| | - Małgorzata Małodobra-Mazur
- Genomtec SA, Ul. Bierutowska 57-59, 51-317, Wrocław, Poland
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, Skłodowskiej-Curie 52, 50-369, Wrocław, Poland
| | - Miron Tokarski
- Genomtec SA, Ul. Bierutowska 57-59, 51-317, Wrocław, Poland.
| |
Collapse
|
9
|
Rimassa L, Lamarca A, O'Kane GM, Edeline J, McNamara MG, Vogel A, Fassan M, Forner A, Kendall T, Adeva J, Casadei-Gardini A, Fornaro L, Hollebecque A, Lowery MA, Macarulla T, Malka D, Mariamidze E, Niger M, Ustav A, Bridgewater J, Macias RI, Braconi C. New systemic treatment paradigms in advanced biliary tract cancer and variations in patient access across Europe. THE LANCET REGIONAL HEALTH. EUROPE 2025; 50:101170. [PMID: 40093395 PMCID: PMC11910789 DOI: 10.1016/j.lanepe.2024.101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 03/19/2025]
Abstract
In recent years, treatment options for patients with advanced biliary tract cancer (BTC) have increased significantly due to the positive results from phase 2/3 clinical trials of immune checkpoint inhibitors, combined with chemotherapy, and molecularly targeted agents. These advances have led to the need for molecular testing to identify actionable alterations and patients amenable to targeted therapies. However, these improvements have brought with them many questions and challenges, including the identification of resistance mechanisms and therapeutic sequences. In this Series paper we aim to provide an overview of the current systemic treatment options for patients with BTC, highlighting disparities in access to innovative treatments and molecular testing across European countries, which lead to inequalities in the possibilities of treating patients with advanced BTC. We also discuss how ongoing European collaborative projects, such as the COST Action Precision-BTC-Network CA22125, supported by COST (European Cooperation in Science and Technology), linked to the European Network for the Study of Cholangiocarcinoma (ENSCCA), can help overcome these disparities and improve the current scenario.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20072, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Angela Lamarca
- Department of Medical Oncology, Oncohealth Institute, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Fundación Jimenez Diaz University Hospital, Avda Reyes Católicos 2, Madrid, 28040, Spain
| | - Grainne M. O'Kane
- University College Dublin, Belfield, Dublin 4, Ireland
- Department of Medical Oncology, St. Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Julien Edeline
- INSERM, Department of Medical Oncology, University Rennes, CLCC Eugène Marquis, COSS [(Chemistry Oncogenesis Stress Signaling)] – UMR_S 1242, Rennes, F-35000, France
| | - Mairéad G. McNamara
- Division of Cancer Sciences, University of Manchester & Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Arndt Vogel
- Toronto General Hospital, UHN, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON, M5G 2M9, Canada
- Hannover Medical School, Carl-Neuberg Str. 1, Hannover, 30659, Germany
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Via Gabelli 61, Padua, 35121, Italy
- Veneto Institute of Oncology (IOV-IRCCS), Via Gattamelata 64, Padua, 35128, Italy
| | - Alejandro Forner
- Liver Unit, Barcelona Clinic Liver Cancer (BCLC) Group, ICMDM, Hospital Clinic IDIBAPS, University of Barcelona, Villarroel 170, Barcelona, 08036, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
| | - Timothy Kendall
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
- Edinburgh Pathology, University of Edinburgh, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
- CRUK Scotland Cancer Centre, Switchback Rd, Glasgow, G61 1BD, UK
| | - Jorge Adeva
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Av. de Córdoba, s/n, Usera, Madrid, 28041, Spain
| | - Andrea Casadei-Gardini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Via Olgettina 60, Milan, 20132, Italy
| | - Lorenzo Fornaro
- Medical Oncology 2 Unit, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, Pisa, 56126, Italy
| | - Antoine Hollebecque
- Département de Médecine Oncologique, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif, F-94805, France
| | - Maeve A. Lowery
- Trinity St James Cancer Institute, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Teresa Macarulla
- Vall d'Hebrón Institute of Oncology (VHIO), Vall d'Hebrón University Hospital, Centre Cellex, Carrer de Natzaret, 115-117, Barcelona, 08035, Spain
| | - David Malka
- Department of Medical Oncology, Institut Mutualiste Montsouris, 42 Boulevard Jourdan, Paris, 75014, France
| | - Elene Mariamidze
- Department of Oncology and Hematology, Todua Clinic, Tevdore Mgvdeli #13, Tbilisi, 0112, Georgia
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, Milan, 20133, Italy
| | - Anu Ustav
- Clinic of Oncology, North-Estonian Medical Centre, Sytiste Rd 19, Tallinn, 13419, Estonia
| | | | - Rocio I.R. Macias
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM) Group, University of Salamanca, IBSAL, CIBERehd, Campus M. Unamuno s/n, Salamanca, 37007, Spain
| | - Chiara Braconi
- CRUK Scotland Cancer Centre, Switchback Rd, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Switchback Rd, Glasgow, G61 1QH, UK
- Beatson West of Scotland Cancer Centre, 1053 Great Western Rd, Glasgow, G12 0YN, UK
| |
Collapse
|
10
|
Ligero M, El Nahhas OSM, Aldea M, Kather JN. Artificial intelligence-based biomarkers for treatment decisions in oncology. Trends Cancer 2025; 11:232-244. [PMID: 39814650 DOI: 10.1016/j.trecan.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
The development of new therapeutic strategies such as immune checkpoint inhibitors (ICIs) and targeted therapies has increased the complexity of the treatment landscape for solid tumors. At the current rate of annual FDA approvals, the potential treatment options could increase by tenfold over the next 5 years. The cost of personalized medicine technologies limits its accessibility, thus increasing socioeconomic disparities in the treated population. In this review we describe artificial intelligence (AI)-based solutions - including deep learning (DL) methods for routine medical imaging and large language models (LLMs) for electronic health records (EHRs) - to support cancer treatment decisions with cost-effective biomarkers. We address the current limitations of these technologies and propose the next steps towards their adoption in routine clinical practice.
Collapse
Affiliation(s)
- Marta Ligero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Mihaela Aldea
- Department of Cancer Medicine, Institut Gustave Roussy, Université Paris-Saclay, F-94805, Villejuif, France; Thoracic Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany; Department of Medicine I, University Hospital Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
11
|
Rimassa L, Khan S, Groot Koerkamp B, Roessler S, Andersen JB, Raggi C, Lleo A, Nault JC, Calderaro J, Gabbi C, Kather JN, Banales JM, Bargellini I, Morement H, Krawczyk M, Farazi PA, Carpino G, Avila MA, Saborowski A, Cardinale V, Braconi C, Macias RI. Mapping the landscape of biliary tract cancer in Europe: challenges and controversies. THE LANCET REGIONAL HEALTH. EUROPE 2025; 50:101171. [PMID: 40093398 PMCID: PMC11910794 DOI: 10.1016/j.lanepe.2024.101171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 03/19/2025]
Abstract
Biliary tract cancer (BTC) is becoming more common worldwide, with geographic differences in incidence and risk factors. In Europe, BTC may be associated with primary sclerosing cholangitis, lithiasis, and liver cirrhosis, but is more frequently observed as a sporadic disease. BTC increasingly affects patients under 60 years, resulting in a significant social and economic burden. Early diagnosis remains challenging due to vague symptoms in 50% of patients with BTC, and lack of specific biomarkers, resulting in late presentation and poor prognosis. The identification of patients at increased risk and reliable biomarkers require collaborative efforts to make faster progress. This Series paper highlights the disparities in access to diagnostic tools and multidisciplinary care in Europe, particularly in economically disadvantaged regions, while identifying priority areas for improvement. Addressing these inequities requires harmonised guidelines, accelerated pathways to curative treatments, and improved awareness among healthcare professionals and the public. Multidisciplinary teams (MDTs) are crucial for the diagnosis of BTC and for improving patient outcomes, yet inconsistencies exist in their implementation not only between different countries, but also between different centres within a country. Collaboration and standardisation of diagnostic and treatment protocols across Europe are essential to effectively address the management of patients with BTC.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via A. Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Shahid Khan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Liver Unit, St Mary's Hospital Campus, South Wharf Road, W2 1NY, London, UK
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Stephanie Roessler
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Jesper B. Andersen
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen N, DK-2200, Denmark
| | - Chiara Raggi
- Department of Experimental and Clinical Medicine, University of Florence, Cubo Centro Polivalente 2, Viale Pieraccini 6, 50139, Florence, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Internal Medicine and Hepatology Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Jean-Charles Nault
- Cordeliers Research Center, Sorbonne University, Inserm, Paris Cité University, “Functional Genomics of Solid Tumors” Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, 16 rue de l'École de Médecine, 75006, Paris, France
- Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, 125 Avenue de Stalingrad, 93000, Bobigny, France
| | - Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, 61 Av. du Général de Gaulle, 94000, Créteil, France
- Department of Pathology, Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, 1 Rue Gustave Eiffel, 94010, Créteil, France
- MINT-Hep, Mondor Integrative Hepatology, 1 Rue Gustave Eiffel, 94010, Créteil, France
| | - Chiara Gabbi
- Humanitas Medical Care, Via Domodossola 9/a, 20145, Milan, Italy
| | - Jakob N. Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307, Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute – Donostia University Hospital, CIBERehd, Paseo Dr. Begiristain, s/n, 20014, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Euskadi Pl., 5, Abando, 48009, Bilbao, Spain
- Department of Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena, s/n, 48940, Leioa, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Calle Irunlarrea 1, 31008, Pamplona, Spain
| | - Irene Bargellini
- Department of Surgical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
- Division of Diagnostic and Interventional Radiology, Candiolo Cancer Institute FPO-IRCCS, Strada Provinciale 142, 10060, Candiolo (TO), Italy
| | - Helen Morement
- AMMF – The Cholangiocarcinoma Charity, Enterprise House, Bassingbourn Road, Stansted, CM24 1QW, Essex, UK
| | - Marcin Krawczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
- Laboratory of Metabolic Liver Diseases, Medical University of Warsaw, Banacha Street 1B, 02-097, Warsaw, Poland
| | - Paraskevi A. Farazi
- School of Medicine, European University Cyprus, 6 Diogenes Street, 2404, Engomi, Nicosia, Cyprus
| | - Guido Carpino
- Department of Anatomical, Histological, Legal Medicine and Orthopedic Sciences, Sapienza University of Rome, Via Alfonso Borelli 50, 00161, Rome, Italy
| | - Matias A. Avila
- Hepatology Laboratory, Solid Tumors Program, CIMA, IdiSNA, CIBERehd, University of Navarra, Calle Irunlarrea 1, 31008, Pamplona, Spain
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Carl Neuberg Str. 1, 30625, Hannover, Germany
| | - Vincenzo Cardinale
- Department of Translational and Precision Medicine, Sapienza University of Rome, Via Alfonso Borelli 50, 00161, Rome, Italy
| | - Chiara Braconi
- School of Cancer Sciences, University of Glasgow, Switchback rd, G61 1QH, Glasgow, UK
- Beatson West of Scotland Cancer Centre, 1053 Great Western rd, G12 0YN, Glasgow, UK
- CRUK Scotland Cancer Centre, G61 1BD, Glasgow, UK
| | - Rocio I.R. Macias
- Experimental Hepatology and Drug Targeting (HEVEPHARM) Group, University of Salamanca, IBSAL, CIBERehd, Campus M. Unamuno s/n, 37007, Salamanca, Spain
| |
Collapse
|
12
|
Cherny NI, Trapani D, Galotti M, Saar M, Bricalli G, Roitberg F, Gyawali B, Curigliano G, Blay JY, Meier K, Latino NJ, de Vries EGE. ESMO Global Consortium Study on the availability, out-of-pocket costs, and accessibility of cancer medicines: 2023 update. Ann Oncol 2025; 36:247-262. [PMID: 39818519 DOI: 10.1016/j.annonc.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND The availability and affordability of safe, effective cancer therapies are core requirements for effective cancer control. Global disparities exist in access, however, yielding unequal cancer outcomes. The goal of this study was to provide updated data regarding the formulary availability, out-of-pocket costs, and accessibility of cancer medicines in countries across the full spectrum of economic development areas. METHODS This study was conducted through an online survey based on a previously validated methodology. It evaluated the formulary availability, out of pocket costs, and actual accessibility of essential generic cancer medication commonly used for a wide range of cancers, including those on the 22nd WHO Model List of Essential Medicines (EML), and cancer medications used in eight high-incidence cancers. A total of 853 field reporters from 170 countries were invited to participate. The collected data were collated and peer-reviewed on the ESMO website, with final adjustments made accordingly. RESULTS Data were collected by 317 reporters and 231 peer reviewers across 126 countries. The study revealed that patients in most high-income countries can access cancer medications without significant out-of-pocket expenditure, including novel treatments with high ESMO-Magnitude of Clinical Benefit Scale (ESMO-MCBS) scores. Conversely, in lower-middle and low-income countries, 40% of traditional chemotherapy agents deemed essential in the WHO EML are only available at full cost to patients. CONCLUSIONS This dataset provides a new and updated 'Global Reference' to enhance accountability for inequalities in access and availability of cancer medicines and to identify challenges and shortcomings to drive public health policies and positively impact national cancer control planning.
Collapse
Affiliation(s)
- N I Cherny
- Department of Medical Oncology, Cancer Pain and Palliative Medicine Service, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - D Trapani
- European Institute of Oncology, IRCCS, Milan, Italy; University of Milano, Department of Oncology and Haemato-Oncology, Milan, Italy
| | - M Galotti
- ESMO Head Office, European Society for Medical Oncology, Lugano, Switzerland
| | - M Saar
- Pharmacy Department, Tartu University Hospital, Tartu, Estonia; European Society of Oncology Pharmacy (ESOP), Luxemburg, Luxemburg
| | - G Bricalli
- ESMO Head Office, European Society for Medical Oncology, Lugano, Switzerland
| | - F Roitberg
- Hospital Sírio-Libanês, São Paulo, Brazil; Brazilian Hospital Company Services Network (Rede EBSERH), Brasilia, Brazil
| | - B Gyawali
- Department of Oncology, Queen's University, Kingston, Canada
| | - G Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy; University of Milano, Department of Oncology and Haemato-Oncology, Milan, Italy
| | - J-Y Blay
- Center Léon Bérard and University Claude Bernard Lyon I, Lyon, France
| | - K Meier
- European Society of Oncology Pharmacy (ESOP), Luxemburg, Luxemburg; Institute for Applied Healthcare Sciences (IFAHS), Hamburg, Germany
| | - N J Latino
- ESMO Head Office, European Society for Medical Oncology, Lugano, Switzerland
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Hofman P, Christopoulos P, D'Haene N, Gosney J, Normanno N, Schuuring E, Tsao MS, Quinn C, Russell J, Keating KE, López-Ríos F. Proposal of real-world solutions for the implementation of predictive biomarker testing in patients with operable non-small cell lung cancer. Lung Cancer 2025; 201:108107. [PMID: 39904223 DOI: 10.1016/j.lungcan.2025.108107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
The implementation of biomarker testing for targeted therapies and immune checkpoint inhibitors is a cornerstone in the management of metastatic and locally advanced non-small cell lung cancer (NSCLC), playing a pivotal role in guiding treatment decisions and patient care. The emergence of precision medicine in the realm of operable NSCLC has been marked by the recent approvals of osimertinib, atezolizumab, nivolumab, pembrolizumab and alectinib for early-stage disease, signifying a shift towards more tailored therapeutic strategies. Concurrently, the landscape of this disease is rapidly evolving, with several further pending approvals and numerous clinical trials in progress. To harness the benefits of these innovative neo-adjuvant and adjuvant therapies, the integration of predictive biomarker testing into standard clinical protocols is imperative for patients with operable NSCLC. A multidisciplinary international consortium has identified three primary obstacles impeding the effective testing of patients with operable NSCLC. These challenges encompass the limited number of test requests by physicians, the inadequacy of tissue samples for comprehensive testing, and the prevalence of cost-reduction measures leading to suboptimal testing practices. This review delineates the aforementioned challenges and proposed solutions, and strategic recommendations aimed at enhancing the testing process. By addressing these issues, we strive to optimize patient outcomes in operable NSCLC, ensuring that individuals receive the most appropriate and effective care based on their unique disease profile.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology, IHU RespirERA, FHU OncoAge, IRCAN, Biobank 0033-00025, Pasteur Hospital, University Côte d'Azur, Nice, France.
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumour Diseases at Heidelberg University Hospital, Heidelberg, Germany
| | - Nicky D'Haene
- Department of Pathology, Hopital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles, Brussels, Belgium
| | - John Gosney
- Cellular Pathology, Royal Liverpool and Broadgreen Hospitals NHS Trust, Liverpool, UK
| | - Nicola Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ming-Sound Tsao
- Princess Margaret Cancer Center, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Christine Quinn
- Diaceutics PLC, Dataworks at Kings Hall Health and Wellbeing Park, Belfast, UK
| | - Jayne Russell
- Diaceutics PLC, Dataworks at Kings Hall Health and Wellbeing Park, Belfast, UK
| | - Katherine E Keating
- Diaceutics PLC, Dataworks at Kings Hall Health and Wellbeing Park, Belfast, UK
| | - Fernando López-Ríos
- Department of Pathology, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Research Institute Hospital Universitario 12 de Octubre (i+12). CIBERONC, Madrid, Spain
| |
Collapse
|
14
|
Goffinet S, Bontoux C, Heeke S, Pezzuto F, Ilié M, Long-Mira E, Lassalle S, Bordone O, Lespinet V, Allégra M, Tanga V, Bonnetaud C, Garnier G, Benzaquen J, Cohen C, Ferrari V, Marquette C, Berthet JP, Calabrese F, Hofman P, Hofman V. EGFR status assessment using reflex testing targeted next-generation sequencing for resected non-squamous non-small cell lung cancer. Virchows Arch 2025; 486:531-539. [PMID: 39741211 DOI: 10.1007/s00428-024-04010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025]
Abstract
EGFR status assessment is mandatory for adjuvant decision-making of resected stage IB-IIIA non-squamous non-small cell lung cancer (NS-NSCLC). It is questionable whether single-gene RT-PCR versus next-generation sequencing (NGS) should be used for this evaluation. Moreover, co-occurring mutations have an impact on tumor behavior and may influence future therapeutic decision-making. We aimed to describe the clinico-pathological and molecular features, as well as the prognostic factors of resected EGFR-mutant NS-NSCLC evaluated with reflex NGS and RT-PCR, so as to compare the results of the two methods. We retrospectively included and collected data from patients with resected EGFR-mutant NS-NSCLC diagnosed in our institution between 2005 and 2024. Additional cases from another center were included. Tumors were analyzed using targeted NGS and RT-PCR. A total of 153 patients were selected. The median follow-up after surgery was 22 months. The positive percent agreement of RT-PCR compared to NGS for the detection of an EGFR mutation was 88%. Common single EGFR mutations (L858R/del19) were observed in 117/153 (77%) cases; 22/153 (14%) and 14/153 (9%) cases had uncommon single and compound EGFR mutations, respectively. 63/153 (41%) patients had a co-occurring mutation, including a TP53 mutation in 43/63 (68%) co-mutated patients. EGFR/TP53-mutant tumors were associated with positive PD-L1 expression compared to EGFR-mutant/TP53-wild-type tumors (62% vs 39%; p = 0.006). Shorter median event-free survival (EFS) in patients with an EGFR exon 18 mutation and those with TP53 exon 7 co-mutation was recorded. The EGFR status should be systematically evaluated using targeted NGS reflex testing for resected NS-NSCLC since future therapeutic decision-making could soon consider integrating the presence of co-occurring mutations.
Collapse
Affiliation(s)
- Samantha Goffinet
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Simon Heeke
- Department of Thoracic/Head & Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Federica Pezzuto
- Department of Cardiac, Thoracic,Vascular Science and Public Health, University of Padova, Padua, Italy
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Olivier Bordone
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Virginie Lespinet
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Maryline Allégra
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Virginie Tanga
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Christelle Bonnetaud
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Georges Garnier
- Department of Oncology, Princess Grace Hospital, Monaco, Monaco
| | - Jonathan Benzaquen
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
- Department of Pneumology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU respirERA, Nice, France
| | - Charlotte Cohen
- Department of Thoracic Surgery, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Victoria Ferrari
- Université Côte d'Azur, Centre de Lutte Contre Le Cancer Antoine Lacassagne, Nice, France
| | - Charles Marquette
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
- Department of Pneumology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU respirERA, Nice, France
| | - Jean Philippe Berthet
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
- Department of Thoracic Surgery, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic,Vascular Science and Public Health, University of Padova, Padua, Italy
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France.
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France.
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France.
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, CHU Nice, FHU OncoAge, IHU RespirERA, Nice, France
- Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| |
Collapse
|
15
|
Liu L, Mu BR, Zhou Y, Wu QL, Li B, Wang DM, Lu MH. Research Trends and Development Dynamics of qPCR-based Biomarkers: A Comprehensive Bibliometric Analysis. Mol Biotechnol 2025:10.1007/s12033-024-01356-7. [PMID: 39843617 DOI: 10.1007/s12033-024-01356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025]
Abstract
Quantitative polymerase chain reaction (qPCR) is a vital molecular technique for biomarker detection; however, its clinical application is impeded by the scarcity of robust biomarkers and the inherent limitations of the technology. This study conducted a bibliometric analysis of 4063 qPCR-based biomarker studies sourced from the Web of Science (WOS) database, employing VOSviewer and CiteSpace to generate multi-dimensional structural insights into this field. The results reveal a growing trend in research within this domain, with gene expression analysis playing a central role in the identification of potential biomarkers. Among these, cancer-related biomarkers are the most prominent, while research on biomarkers for other diseases remains limited. Liquid biopsy biomarkers, including microRNA (miRNA), circulating free DNA (cfDNA), and circulating tumor DNA (ctDNA), are increasingly being explored. The integration of bioinformatics, omics analysis, and high-throughput technologies with qPCR is accelerating biomarker discovery. Furthermore, large-scale parallel sequencing is emerging as a potential alternative to relative quantification and microarray techniques. Nevertheless, qPCR remains essential for validating specific biomarkers, and further standardization of its protocols is necessary to enhance reliability. This study provides a systematic analysis of qPCR-based biomarker research and underscores the need for future technological integration and standardization to facilitate broader clinical applications.
Collapse
Affiliation(s)
- Li Liu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Ben-Rong Mu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Ya Zhou
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Qing-Lin Wu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Bin Li
- Department of Respiratory Medicine, Guangyuan Hospital of Traditional Chinese Medicine, No.133 Jianshe Road, Lizhou District, Guangyuan, 628099, Sichuan, China
| | - Dong-Mei Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China.
| | - Mei-Hong Lu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
16
|
Cai XJ, Peng CR, Cui YY, Li L, Huang MW, Zhang HY, Zhang JY, Li TJ. Identification of genomic alteration and prognosis using pathomics-based artificial intelligence in oral leukoplakia and head and neck squamous cell carcinoma: a multicenter experimental study. Int J Surg 2025; 111:426-438. [PMID: 39248300 PMCID: PMC11745750 DOI: 10.1097/js9.0000000000002077] [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: 06/02/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Loss of chromosome 9p is an important biomarker in the malignant transformation of oral leukoplakia (OLK) to head and neck squamous cell carcinoma (HNSCC), and is associated with the prognosis of HNSCC patients. However, various challenges have prevented 9p loss from being assessed in clinical practice. The objective of this study was to develop a pathomics-based artificial intelligence (AI) model for the rapid and cost-effective prediction of 9p loss (9PLP). MATERIALS AND METHODS Three hundred thirty-three OLK cases were retrospectively collected with hematoxylin and eosin (H&E)-stained whole slide images and genomic alteration data from multicenter cohorts to develop the genomic alteration prediction AI model. They were divided into a training dataset ( n =217), a validation dataset ( n =93), and an external testing dataset ( n =23). The latest Transformer method and XGBoost algorithm were combined to develop the 9PLP model. The AI model was further applied and validated in two multicenter HNSCC datasets ( n =42 and n =365, respectively). Moreover, the combination of 9PLP with clinicopathological parameters was used to develop a nomogram model for assessing HNSCC patient prognosis. RESULTS 9PLP could predict chromosome 9p loss rapidly and effectively using both OLK and HNSCC images, with the area under the curve achieving 0.890 and 0.825, respectively. Furthermore, the predictive model showed high accuracy in HNSCC patient prognosis assessment (the area under the curve was 0.739 for 1-year prediction, 0.705 for 3-year prediction, and 0.691 for 5-year prediction). CONCLUSION To the best of our knowledge, this study developed the first genomic alteration prediction deep learning model in OLK and HNSCC. This novel AI model could predict 9p loss and assess patient prognosis by identifying pathomics features in H&E-stained images with good performance. In the future, the 9PLP model may potentially contribute to better clinical management of OLK and HNSCC.
Collapse
Affiliation(s)
- Xin-Jia Cai
- Central Laboratory, Peking University School and Hospital of Stomatology
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
| | - Chao-Ran Peng
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
| | - Ying-Ying Cui
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
| | - Long Li
- Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital, Central South University, Changsha, People’s Republic of China
| | - Ming-Wei Huang
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing
| | - He-Yu Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
| | - Jian-Yun Zhang
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
| | - Tie-Jun Li
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034)
| |
Collapse
|
17
|
Costa J, Membrino A, Zanchetta C, Rizzato S, Cortiula F, Rossetto C, Pelizzari G, Aprile G, Macerelli M. The Role of ctDNA in the Management of Non-Small-Cell Lung Cancer in the AI and NGS Era. Int J Mol Sci 2024; 25:13669. [PMID: 39769431 PMCID: PMC11727717 DOI: 10.3390/ijms252413669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Liquid biopsy (LB) involves the analysis of circulating tumour-derived DNA (ctDNA), providing a minimally invasive method for gathering both quantitative and qualitative information. Genomic analysis of ctDNA through next-generation sequencing (NGS) enables comprehensive genetic profiling of tumours, including non-driver alterations that offer prognostic insights. LB can be applied in both early-stage disease settings, for the diagnosis and monitoring of minimal residual disease (MRD), and advanced disease settings, for monitoring treatment response and understanding the mechanisms behind disease progression and tumour heterogeneity. Currently, LB has limited use in clinical practice, primarily due to its significant costs, limited diagnostic yield, and uncertain prognostic role. The application of artificial intelligence (AI) in the medical field is a promising approach to processing extensive information and applying it to individual cases to enhance therapeutic decision-making and refine risk assessment.
Collapse
Affiliation(s)
- Jacopo Costa
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.M.); (C.Z.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Alexandro Membrino
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.M.); (C.Z.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Carol Zanchetta
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (A.M.); (C.Z.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Simona Rizzato
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Francesco Cortiula
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Reproduction, 6229 ER Maastricht, The Netherlands
| | - Ciro Rossetto
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Giacomo Pelizzari
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Giuseppe Aprile
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| | - Marianna Macerelli
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (S.R.); (F.C.); (C.R.); (G.P.); (G.A.); (M.M.)
| |
Collapse
|
18
|
Ehman M, Punian J, Weymann D, Regier DA. Next-generation sequencing in oncology: challenges in economic evaluations. Expert Rev Pharmacoecon Outcomes Res 2024; 24:1115-1132. [PMID: 39096135 DOI: 10.1080/14737167.2024.2388814] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Next-generation sequencing (NGS) identifies genetic variants to inform personalized treatment plans. Insufficient evidence of cost-effectiveness impedes the integration of NGS into routine cancer care. The complexity of personalized treatment challenges conventional economic evaluation. Clearly delineating challenges informs future cost-effectiveness analyses to better value and contextualize health, preference-, and equity-based outcomes. AREAS COVERED We conducted a scoping review to characterize the applied methods and outcomes of economic evaluations of NGS in oncology and identify existing challenges. We included 27 articles published since 2016 from a search of PubMed, Embase, and Web of Science. Identified challenges included defining the evaluative scope, managing evidentiary limitations including lack of causal evidence, incorporating preference-based utility, and assessing distributional and equity-based impacts. These challenges reflect the difficulty of generating high-quality clinical effectiveness and real-world evidence (RWE) for NGS-guided interventions. EXPERT OPINION Adapting methodological approaches and developing life-cycle health technology assessment (HTA) guidance using RWE is crucial for implementing NGS in oncology. Healthcare systems, decision-makers, and HTA organizations are facing a pivotal opportunity to adapt to an evolving clinical paradigm and create innovative regulatory and reimbursement processes that will enable more sustainable, equitable, and patient-oriented healthcare.
Collapse
Affiliation(s)
- Morgan Ehman
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Jesman Punian
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Deirdre Weymann
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Dean A Regier
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
van de Haar J, Roepman P, Andre F, Balmaña J, Castro E, Chakravarty D, Curigliano G, Czarnecka AM, Dienstmann R, Horak P, Italiano A, Marchiò C, Monkhorst K, Pritchard CC, Reardon B, Russnes HEG, Sirohi B, Sosinsky A, Spanic T, Turnbull C, Van Allen E, Westphalen CB, Tamborero D, Mateo J. ESMO Recommendations on clinical reporting of genomic test results for solid cancers. Ann Oncol 2024; 35:954-967. [PMID: 39112111 DOI: 10.1016/j.annonc.2024.06.018] [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: 04/22/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Genomic tumour profiling has a crucial role in the management of patients with solid cancers, as it helps selecting and prioritising therapeutic interventions based on prognostic and predictive biomarkers, as well as identifying markers of hereditary cancers. Harmonised approaches to interpret the results of genomic testing are needed to support physicians in their decision making, prevent inequalities in precision medicine and maximise patient benefit from available cancer management options. METHODS The European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group assembled a group of international experts to propose recommendations for preparing clinical genomic reports for solid cancers. These recommendations aim to foster best practices in integrating genomic testing within clinical settings. After review of available evidence, several rounds of surveys and focused discussions were conducted to reach consensus on the recommendation statements. Only consensus recommendations were reported. Recommendation statements were graded in two tiers based on their clinical importance: level A (required to maintain common standards in reporting) and level B (optional but necessary to achieve ideal practice). RESULTS Genomics reports should present key information in a front page(s) followed by supplementary information in one or more appendices. Reports should be structured into sections: (i) patient and sample details; (ii) assay and data analysis characteristics; (iii) sample-specific assay performance and quality control; (iv) genomic alterations and their functional annotation; (v) clinical actionability assessment and matching to potential therapy indications; and (vi) summary of the main findings. Specific recommendations to prepare each of these sections are made. CONCLUSIONS We present a set of recommendations aimed at structuring genomics reports to enhance physician comprehension of genomic profiling results for solid cancers. Communication between ordering physicians and professionals reporting genomic data is key to minimise uncertainties and to optimise the impact of genomic tests in patient care.
Collapse
Affiliation(s)
- J van de Haar
- Department of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam
| | - P Roepman
- Medical Department, Hartwig Medical Foundation, Amsterdam, The Netherlands
| | - F Andre
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - J Balmaña
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - E Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
| | - D Chakravarty
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Curigliano
- Early Drug Development for Innovative Therapies Division, Istituto Europeo di Oncologia, IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - A M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw; Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - R Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona; University of Vic-Central University of Catalonia, Vic, Spain; Oncoclínicas, São Paulo, Brazil
| | - P Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Italiano
- Early Phase Trials and Sarcoma Units, Institut Bergonie, Bordeaux; DITEP, Gustave Roussy, Villejuif; Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - C Marchiò
- Department of Medical Sciences, University of Turin, Turin; Division of Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - K Monkhorst
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C C Pritchard
- Department of Laboratory Medicine and Pathology, Brotman Baty Institute for Precision Medicine, University of Washington, Seattle
| | - B Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Division of Population Sciences, Dana-Farber Cancer Institute, Boston, USA
| | - H E G Russnes
- Department of Pathology, Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - B Sirohi
- Department of Medical Oncology, Vedanta Medical Research Foundation (BALCO Medical Centre), Raipur, India
| | | | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - C Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London; The Royal Marsden NHS Foundation Trust, London, UK
| | - E Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston; Division of Population Sciences, Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Harvard University, Boston, USA
| | - C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich; German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D Tamborero
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona.
| |
Collapse
|
20
|
Budczies J, Kazdal D, Menzel M, Beck S, Kluck K, Altbürger C, Schwab C, Allgäuer M, Ahadova A, Kloor M, Schirmacher P, Peters S, Krämer A, Christopoulos P, Stenzinger A. Tumour mutational burden: clinical utility, challenges and emerging improvements. Nat Rev Clin Oncol 2024; 21:725-742. [PMID: 39192001 DOI: 10.1038/s41571-024-00932-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Tumour mutational burden (TMB), defined as the total number of somatic non-synonymous mutations present within the cancer genome, varies across and within cancer types. A first wave of retrospective and prospective research identified TMB as a predictive biomarker of response to immune-checkpoint inhibitors and culminated in the disease-agnostic approval of pembrolizumab for patients with TMB-high tumours based on data from the Keynote-158 trial. Although the applicability of outcomes from this trial to all cancer types and the optimal thresholds for TMB are yet to be ascertained, research into TMB is advancing along three principal avenues: enhancement of TMB assessments through rigorous quality control measures within the laboratory process, including the mitigation of confounding factors such as limited panel scope and low tumour purity; refinement of the traditional TMB framework through the incorporation of innovative concepts such as clonal, persistent or HLA-corrected TMB, tumour neoantigen load and mutational signatures; and integration of TMB with established and emerging biomarkers such as PD-L1 expression, microsatellite instability, immune gene expression profiles and the tumour immune contexture. Given its pivotal functions in both the pathogenesis of cancer and the ability of the immune system to recognize tumours, a profound comprehension of the foundational principles and the continued evolution of TMB are of paramount relevance for the field of oncology.
Collapse
Affiliation(s)
- Jan Budczies
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany.
- Center for Personalized Medicine (ZPM), Heidelberg, Germany.
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Michael Menzel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Susanne Beck
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Klaus Kluck
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Christian Altbürger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Michael Allgäuer
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Aysel Ahadova
- Department of Applied Tumour Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumour Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumour Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Solange Peters
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne University, Lausanne, Switzerland
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Petros Christopoulos
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumour Diseases at Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
- Translational Lung Research Center (TLRC) Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany.
- Center for Personalized Medicine (ZPM), Heidelberg, Germany.
| |
Collapse
|
21
|
Rassy E, Mosele MF, Di Meglio A, Pistilli B, Andre F. Precision oncology in patients with breast cancer: towards a 'screen and characterize' approach. ESMO Open 2024; 9:103716. [PMID: 39303452 PMCID: PMC11439525 DOI: 10.1016/j.esmoop.2024.103716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- E Rassy
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Oncostat U1018, Inserm, Université Paris-Saclay, Equipe labellisée Ligue Contre le Cancer, Villejuif
| | - M F Mosele
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif
| | - A Di Meglio
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif
| | - B Pistilli
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; INSERM U1279, Gustave Roussy, Villejuif, France
| | - F Andre
- Gustave Roussy, Département de Médecine Oncologique, Villjuif; Université Paris-Saclay, Gustave Roussy, Inserm U981, Villejuif.
| |
Collapse
|
22
|
Gristina V, Russo TDB, Barraco N, Gottardo A, Pepe F, Russo G, Fulfaro F, Incorvaia L, Badalamenti G, Troncone G, Malapelle U, Russo A, Bazan V, Galvano A. Clinical utility of ctDNA by amplicon based next generation sequencing in first line non small cell lung cancer patients. Sci Rep 2024; 14:22141. [PMID: 39333636 PMCID: PMC11436775 DOI: 10.1038/s41598-024-73046-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
The assessment of ctDNA has emerged as a minimally invasive avenue for molecular diagnosis and real-time tracking of tumor progression in NSCLC. However, the evaluation of ctDNA by amplicon-based NGS has been not endorsed by all the healthcare systems and remains to be fully integrated into clinical routine practice. To compare tissue single-gene with plasma multiplexed testing, we retrospectively evaluated 120 plasma samples from 12 consecutive patients with advanced non-squamous NSCLC who were part of a prospective study enrolling treatment-naïve patients and in which tissue samples were evaluated using a single-gene testing approach. While the plasma ctDNA detection of EGFR and BRAF mutations had an acceptable level of concordance with the archival tissue (85%), discordance was seen in all the patients in whom ALK alterations were only detected in tissue samples. Among six responders and six non-responders, early ctDNA mutant allelic frequency (MAF) reduction seemed to predict radiologic responses and longer survival, whereas increasing MAF values with the emergence of co-mutations like BRAFV600E, KRASG12V or TP53M237I seemed to be an early indicator of molecular and radiologic progression. This report using an amplicon-based NGS assay on ctDNA underscores the real-life need for plasma and tissue genotyping as complementary tools in the diagnostic and therapeutic decision-making process.
Collapse
Affiliation(s)
- Valerio Gristina
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Andrea Gottardo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fabio Fulfaro
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy.
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy.
| | - Viviana Bazan
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| |
Collapse
|
23
|
Vu M, Degeling K, Westerman D, IJzerman MJ. Scenario analysis and multi-criteria decision analysis to explore alternative reimbursement pathways for whole genome sequencing for blood cancer patients. J Cancer Policy 2024; 41:100501. [PMID: 39142605 DOI: 10.1016/j.jcpo.2024.100501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Whole genome sequencing (WGS) has transformative potential for blood cancer management, but reimbursement is hindered by uncertain benefits relative to added costs. This study employed scenario planning and multi-criteria decision analysis (MCDA) to evaluate stakeholders' preferences for alternative reimbursement pathways, informing future health technology assessment (HTA) submission of WGS in blood cancer. METHODS Key factors influencing WGS reimbursement in blood cancers were identified through a literature search. Hypothetical scenarios describing various evidential characteristics of WGS for HTA were developed using the morphological approach. An online survey, incorporating MCDA weights, was designed to gather stakeholder preferences (consumers/patients, clinicians/health professionals, industry representatives, health economists, and HTA committee members) for these scenarios. The survey assessed participants' approval of WGS reimbursement for each scenario, and scenario preferences were determined using the geometric mean method, applying an algorithm to improve reliability and precision by addressing inconsistent responses. RESULTS Nineteen participants provided complete survey responses, primarily clinicians or health professionals (n = 6; 32 %), consumers/patients and industry representatives (both at n = 5; 26 %). "Clinical impact of WGS results on patient care" was the most critical criterion (criteria weight of 0.25), followed by "diagnostic accuracy of WGS" (0.21), "cost-effectiveness of WGS" (0.19), "availability of reimbursed treatment after WGS" (0.16), and "eligibility criteria for reimbursed treatment based on actionable WGS results" and "cost comparison of WGS" (both at 0.09). Participants preferred a scenario with substantial clinical evidence, high access to reimbursed targeted treatment, cost-effectiveness below $50,000 per quality-adjusted life year (QALY) gained, and affordability relative to standard molecular tests. Reimbursement was initially opposed until criteria such as equal cost to standard tests and better treatment accessibility were met. CONCLUSION Payers commonly emphasize acceptable cost-effectiveness, but strong clinical evidence for many variants and comparable costs to standard tests are likely to drive positive reimbursement decisions for WGS.
Collapse
Affiliation(s)
- Martin Vu
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Koen Degeling
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - David Westerman
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia
| | - Maarten J IJzerman
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia; Erasmus School of Health Policy and Management, Rotterdam, the Netherlands.
| |
Collapse
|
24
|
Lawler M, Keeling P, Kholmanskikh O, Minnaard W, Moehlig-Zuttermeister H, Normanno N, Philip R, Popp C, Salgado R, Santiago-Walker AE, Trullas A, van Doorn-Khosrovani SBVW, Vart R, Vermeulen J, Vitaloni M, Verweij J. Empowering effective biomarker-driven precision oncology: A call to action. Eur J Cancer 2024; 209:114225. [PMID: 39053288 DOI: 10.1016/j.ejca.2024.114225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024]
Abstract
Precision oncology has a significant role to play in delivering optimal patient care. Biomarkers are critical enablers for precision oncology across the continuum of cancer diagnosis, in defining patient prognosis, and in predicting the response to treatments and their potential toxicities, as well as delineating the risk of hereditary cancer syndromes. Biomarkers also potentiate cancer drug development, accelerating patient access to safe and effective therapies. However, despite an accurate and timely diagnosis being critical to patient survival, advances in genomic testing are not being fully exploited in daily clinical practice, leading to missed opportunities to deliver the most effective treatments for patients. Biomarker testing availability and implementation often lag behind approvals of respective biomarker-informed therapies, limiting prompt patient access to these life-saving drugs. Multiple factors currently impede the routine adoption of biomarker testing including, but not limited to, cost, lack of test reimbursement, limited access, regulatory hurdles, lack of knowledge, insufficient cooperation on assay development, and the urgent need to harmonize and validate testing assays, all leading to inefficient diagnostic pathways. Clinical guidelines increasingly include genomic profiling, and recent evidence suggests that precision oncology can be delivered in a cost-effective way for financially-challenged health systems. Therefore, precision genomic testing for cancer biomarkers must be embedded into the clinical practice of oncology care delivery going forward. We articulate a series of recommendations and a call to action to underpin the mainstreaming of a biomarker-informed precision oncology approach to enhance patient outcomes and deliver cost effective 21st century cancer care.
Collapse
Affiliation(s)
- Mark Lawler
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, BT9 7AE, UK; Cancer Drug Development Forum, Clos Chapelle-aux-Champs 30, 1200 Brussels, Belgium.
| | - Peter Keeling
- Diaceutics PLC, Health and Wellbeing Park, Kings Hall, First Floor Building Two, Dataworks at, 490 Lisburn Rd, BT9 6GU Belfast, UK.
| | - Olga Kholmanskikh
- Federal Agency for Medicines and Health Products, Galileelaan 5/03, 1210 Brussels, Belgium.
| | - Warnyta Minnaard
- Missie Tumor Onbekend, Fannius Scholtenstraat 69H 1051 EV, Amsterdam, Netherlands.
| | | | - Nicola Normanno
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Via Piero Maroncelli, 40, 47014 Meldola, Italy
| | - Reena Philip
- Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Claudia Popp
- F. Hoffmann-La Roche Ltd, Building 2, CH-4070 Basel, Switzerland.
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Lindendreef 1, 2020 Antwerp, Belgium; Division of Research, Peter Mac Callum Cancer Centre, 305 Grattan Street, Melbourne VIC 3052, Australia.
| | - Ademi E Santiago-Walker
- Janssen Research & Development LLC, Johnson & Johnson Innovative Medicine, 1400 McKean Road, Spring House, PA 19477, USA.
| | - Ana Trullas
- European Medicines Agency, Domenico Scarlattilaan 6, 1083 HS Amsterdam, the Netherlands.
| | - Sahar B van Waalwijk van Doorn-Khosrovani
- Department of Medical Oncology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; CZ Health Insurance, Ringbaan West 236, 5038 KE Tilburg, the Netherlands.
| | - Richard Vart
- Eli Lilly and Company Ltd, 8 Arlington Square West, Downshire Way, Bracknell, RG12 1PU, UK.
| | - Jessica Vermeulen
- Janssen Biologics B.V., a Johnson & Johnson company, Emmy Noetherweg 6, 2333 BK Leiden, the Netherlands.
| | - Marianna Vitaloni
- Digestive Cancers Europe, Rue de la Loi 235/27, 1040 Brussels, Belgium.
| | - Jaap Verweij
- Cancer Drug Development Forum, Clos Chapelle-aux-Champs 30, 1200 Brussels, Belgium.
| |
Collapse
|
25
|
Smids J, Bomhof C, IJzerman M, Bunnik E. Whole genome sequencing as a ticket to cancer treatment in the Netherlands: Are inequalities in access to molecular diagnostics unfair? J Cancer Policy 2024; 41:100492. [PMID: 38908820 DOI: 10.1016/j.jcpo.2024.100492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/20/2024] [Accepted: 06/08/2024] [Indexed: 06/24/2024]
Abstract
Whole genome sequencing (WGS) of a tumour may sometimes reveal additional potential targets for medical treatment. Practice variation in the use of WGS is therefore a source of unequal access to targeted therapies and, as a consequence, of disparities in health outcomes. Moreover, this may even be more significant if patients seek access to WGS by paying a relatively limited amount of money out of pocket, and sometimes effectively buy themselves a ticket to (very) expensive publicly funded treatments. Should resulting unequal access to WGS be considered unfair? Drawing from current practice in the Dutch healthcare system, known as egalitarian, we argue that differences in employment of WGS between hospitals are the consequence of the fact that medical innovation and its subsequent uptake inevitably takes time. Consequently, temporal inequalities in access can be deemed acceptable, or at least tolerated, because and insofar as, ultimately, all patients benefit. However, we argue against allowing a practice of out-of-pocket payments for WGS in publicly funded healthcare systems, for four reasons: because allowing private spending favours patients with higher socio-economic status significantly more than practice variation between hospitals does, may lead to displacement of publicly funded health care, does not help to ultimately benefit all, and may undermine the solidaristic ethos essential for egalitarian healthcare systems.
Collapse
Affiliation(s)
- Jilles Smids
- Erasmus Medical Center, Rotterdam, the Netherlands.
| | | | - Maarten IJzerman
- Erasmus School of Health Policy & Management, Erasmus University, Rotterdam, the Netherlands; Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Eline Bunnik
- Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
26
|
Horgan D, Pesapane F, Van der Buckle M, de Maria R, Dube F, Singh J, Ługowska I, Bayle A, Hofman P, Malapelle U, Hills T, Capoluongo ED, Subbiah V. From theory to practice: Implementing next-generation sequencing and public health genomics in healthcare systems. Crit Rev Oncol Hematol 2024; 201:104433. [PMID: 38955310 DOI: 10.1016/j.critrevonc.2024.104433] [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: 04/19/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
If Europe's health systems make a conscious decision to increase their utilization of technology and techniques that can enhance prevention and expedite early-stage diagnosis, they can effectively address the growing challenges of disease. By embracing these advancements, these health systems can significantly improve their response to emerging health issues.However, at present the effective integration and exploitation of these opportunities remains hesitant and suboptimal, and health and health services underperform accordingly, with patients suffering from the continuing variations in diagnosis and access to innovation. This paper presents a comprehensive study that examines the current state of various influential disciplines and factors in European countries. It specifically focuses on the adoption of Next Generation Screening technologies and the development stage of Public Health Genomics. The assessment of these areas is presented in the context of a rapidly changing policy environment, which provides an opportunity for a fundamental reconsideration of how and where new tools can be integrated into healthcare systems and routine practices. Top of Form.
Collapse
Affiliation(s)
- Denis Horgan
- European Alliance for Personalised Medicine, Brussels 1040, Belgium; Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Faculty of Engineering and Technology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India.
| | - Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | | | - Ruggero de Maria
- Institute of General Pathology, Catholic University of the Sacred Heart, Rome 20123, Italy
| | - France Dube
- Astra Zeneca, Concord Pike, Wilmington, DE 19803, USA
| | - Jaya Singh
- European Alliance for Personalised Medicine, Brussels 1040, Belgium
| | - Iwona Ługowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Roentgena 5, Warsaw 02-781, Poland
| | - Arnaud Bayle
- Drug Development Department (DITEP), Gustave Roussy - Cancer Campus, Université Paris Sacla, Villejuif, France
| | - Paul Hofman
- European Liquid Biopsy Society, Hamburg, Germany
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples 80138, Italy
| | - Tanya Hills
- Boehringer Ingelheim International GmbH, Binger Str. 173, Ingelheim am Rhein 55218, Germany
| | - Ettore D Capoluongo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples 80138, Italy; Department of Clinical Pathology, Azienda Ospedaliera San Giovanni Addolorata, Via Amba Aradam 8, Rome 00184, Italy
| | - Vivek Subbiah
- Sarah Cannon Research Institute, Nashville, TN 37203, USA.
| |
Collapse
|
27
|
Mirza M, Goerke L, Anderson A, Wilsdon T. Assessing the Cost-Effectiveness of Next-Generation Sequencing as a Biomarker Testing Approach in Oncology and Policy Implications: A Literature Review. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:1300-1309. [PMID: 38729563 DOI: 10.1016/j.jval.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/21/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
OBJECTIVE A key hurdle in broader next-generation sequencing (NGS) biomarker testing access in oncology is the ongoing debate on NGS's cost-effectiveness. We conducted a systematic review of existing evidence of the costs of NGS as a biomarker testing strategy in oncology and developed policy suggestions. METHODS We searched multiple databases for studies reporting cost comparisons and cost-effectiveness of NGS across oncology indications and geographies between 2017 and 2022, inclusive. Inclusion criteria were established based on indication and type of cost-effectiveness analysis provided. We validated analyses and policy recommendations with 5 payer/policy maker interviews in the United States, Europe, and United Kingdom. RESULTS Of the 634 identified studies, 29 met inclusion criteria, spanning 12 countries and 6 indications. Cost comparisons of NGS were evaluated using 3 methodologies: (1) comparison of direct testing costs, (2) comparison of holistic testing costs, and (3) comparison of long-term patient outcomes and costs. Targeted panel testing (2-52 genes) was considered cost-effective when 4+ genes were assessed, and larger panels (hundreds of genes) were generally not cost-effective. Holistic analysis demonstrated that NGS reduces turnaround time, healthcare staff requirements, number of hospital visits, and hospital costs. Finally, studies evaluating NGS testing including the cost of targeted therapies generally found the incremental cost-effectiveness ratio to be above common thresholds but highlighted valuable patient benefits. CONCLUSIONS Current literature supports NGS's cost-effectiveness as an oncology biomarker testing strategy under specific conditions. These findings underscore the need to develop policies to support holistic assessment of NGS to ensure appropriate reimbursement and access.
Collapse
Affiliation(s)
- Myriam Mirza
- Charles River Associates, Palais Leopold, Munich, Germany.
| | - Lutz Goerke
- Charles River Associates, Palais Leopold, Munich, Germany
| | | | - Tim Wilsdon
- Charles River Associates, London, England, UK
| |
Collapse
|
28
|
Veney DJ, Wei LY, Toland AE, Presley CJ, Hampel HL, Padamsee TJ, Lee CN, Irvin WJ, Bishop MJ, Kim JJ, Hovick SR, Senter LA, Stover DG. A video intervention to improve patient understanding of tumor genomic testing in patients with cancer. Cancer Med 2024; 13:e70095. [PMID: 39258462 PMCID: PMC11387988 DOI: 10.1002/cam4.70095] [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: 02/02/2024] [Revised: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Tumor genomic testing (TGT) is standard-of-care for most patients with advanced/metastatic cancer. Despite established guidelines, patient education prior to TGT is frequently omitted. The purpose of this study was to evaluate the impact of a concise 4 min video for patient education prior to TGT. METHODS Based on a quality improvement cycle, an animated video was created to be applicable to any cancer type, incorporating culturally diverse images, available in English and Spanish. Patients undergoing standard-of-care TGT were enrolled at a tertiary academic institution and completed survey instruments prior to video viewing (T1) and immediately post-viewing (T2). Instruments included: (1) 10-question objective genomic knowledge; (2) 10-question video message-specific knowledge; (3) 11-question Trust in Provider; (4) attitudes regarding TGT. RESULTS A total of 150 participants were enrolled. For the primary objective, there was a significant increase in video message-specific knowledge (median 10 point increase; p < 0.0001) with no significant change in genomic knowledge/understanding (p = 0.89) or trust in physician/provider (p = 0.59). Results for five questions significantly improved, including the likelihood of TGT impact on treatment decision, incidental germline findings, and cost of testing. Improvement in video message-specific knowledge was consistent across demographic groups, including age, income, and education. CONCLUSIONS A concise, 3-4 min, broadly applicable video incorporating culturally diverse images administered prior to TGT significantly improved video message-specific knowledge across all demographic groups. This resource is publicly available at http://www.tumor-testing.com, with a goal to efficiently educate and empower patients regarding TGT while addressing guidelines within the flow of clinical practice.
Collapse
Affiliation(s)
- Deloris J. Veney
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Lai Y. Wei
- Department of Biomedical InformaticsOhio State UniversityColumbusOhioUSA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
- Department of Cancer Biology and GeneticsThe Ohio State UniversityColumbusOhioUSA
| | - Carolyn J. Presley
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Heather L. Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics ResearchCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Tasleem J. Padamsee
- Division of Health Services Management and PolicyCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
| | - Clara N. Lee
- Division of Health Services Management and PolicyCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
- Present address:
Division of Plastic and Reconstructive SurgeryUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | | | | | - James J. Kim
- Bon Secours‐Mercy Health St. ElizabethYoungstownOhioUSA
| | | | - Leigha A. Senter
- Division of Human Genetics, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Daniel G. Stover
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
- Department of Biomedical InformaticsOhio State UniversityColumbusOhioUSA
- Pelotonia Institute for Immuno‐Oncology, Ohio State University Comprehensive CancerColumbusOhioUSA
| |
Collapse
|
29
|
Ignatiadis M, Poulakaki F, Spanic T, Brain E, Lacombe D, Sonke GS, Vincent-Salomon A, Van Duijnhoven F, Meattini I, Kaidar-Person O, Aftimos P, Lecouvet F, Cardoso F, Retèl VP, Cameron D. EBCC-14 manifesto: Addressing disparities in access to innovation for patients with metastatic breast cancer across Europe. Eur J Cancer 2024; 207:114156. [PMID: 38861756 DOI: 10.1016/j.ejca.2024.114156] [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: 04/11/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
The European Breast Cancer Council (EBCC) traditionally identifies controversies or major deficiencies in the management of patients with breast cancer and selects a multidisciplinary expert team to collaborate in setting crucial principles and recommendations to improve breast cancer care. The 2024 EBCC manifesto focuses on disparities in the care of patients with metastatic breast cancer. There are several reasons for existing disparities both between and within countries. Our recommendations aim to address the stigma of metastatic disease, which has led to significant disparities in access to innovative care regardless of the gross national income of a country. These recommendations are for different stakeholders to promote the care of patients with metastatic breast cancer across Europe and worldwide.
Collapse
Affiliation(s)
- Michail Ignatiadis
- Department of Medical Oncology, Institut Bordet, Hôpital Universitaire de Bruxelles, Brussels, Belgium.
| | - Fiorita Poulakaki
- Breast Surgery Department, Athens Medical Center, Athens, Greece; Europa Donna - The European Breast Cancer Coalition, Milan, Italy
| | - Tanja Spanic
- Europa Donna - The European Breast Cancer Coalition, Milan, Italy; Europa Donna Slovenia, Ljubljana, Slovenia
| | - Etienne Brain
- Department of Medical Oncology, Institut Curie, Saint Cloud, France
| | - Denis Lacombe
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Gabe S Sonke
- University of Amsterdam, Amsterdam, the Netherlands
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, Paris, France
| | - Frederieke Van Duijnhoven
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy; Radiation Oncology & Breast Unit, Oncology Department, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Orit Kaidar-Person
- Department of Radiation Oncology, Sheba Medical Center, Ramat Gan, Israel; Tel Aviv School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Philippe Aftimos
- Department of Medical Oncology, Institut Bordet, Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | - Frederic Lecouvet
- Institut du Cancer Roi Albert II (IRA2), Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium; Department of Medical Imaging, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Erasmus School of Health Policy and Management, Erasmus University Rotterdam (ESHPM), Rotterdam, the Netherlands
| | - David Cameron
- Edinburgh University Cancer Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
30
|
Vu M, Degeling K, Ryland GL, Hofmann O, Ng AP, Westerman D, IJzerman MJ. Economic Impact of Whole Genome Sequencing and Whole Transcriptome Sequencing Versus Routine Diagnostic Molecular Testing to Stratify Patients with B-Cell Acute Lymphoblastic Leukemia. J Mol Diagn 2024; 26:673-684. [PMID: 39059881 DOI: 10.1016/j.jmoldx.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 07/28/2024] Open
Abstract
Whole genome and whole transcriptome sequencing (WGTS) can accurately distinguish B-cell acute lymphoblastic leukemia (B-ALL) genomic subtypes. However, whether this is economically viable remains unclear. This study compared the direct costs and molecular subtype classification yield using different testing strategies for WGTS in adolescent and young adult/adult patients with B-ALL. These approaches were: (1) combined BCR::ABL1 by fluorescence in situ hybridization (FISH) + WGTS for all patients; and (2) sequential BCR::ABL1 FISH + WGTS contingent on initial BCR::ABL1 FISH test outcome. The cost of routine diagnostic testing was estimated using Medicare or hospital fees, and the additional cost of WGTS was evaluated from the health care provider perspective using time-driven activity-based costing with resource identification elicited from experts. Molecular subtype classification yield data were derived from literature sources. Parameter uncertainty was assessed through deterministic sensitivity analysis; additional scenario analyses were performed. The total per patient cost of WGTS was $4319 (all costs reported in US dollars); consumables accounted for 74% of the overall cost, primarily driven by sequencing-related consumables. The incremental cost per additional patient categorized into molecular subtype was $8498 for combined BCR::ABL1 FISH + WGTS for all patients and $5656 for initial BCR::ABL1 FISH + WGTS for select patients compared with routine diagnostic testing. A reduction in the consumable costs of WGTS or an increase in the yield of molecular subtype classification is favorable.
Collapse
Affiliation(s)
- Martin Vu
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Koen Degeling
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Georgina L Ryland
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Oliver Hofmann
- Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ashley P Ng
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia; Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Victoria, Australia; WEHI (Walter and Eliza Hall Institute of Medical Research), Melbourne, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia; Department of Health Services Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - David Westerman
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia; Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Maarten J IJzerman
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia; Erasmus School of Health Policy and Management, Rotterdam, The Netherlands.
| |
Collapse
|
31
|
Boileve A, Smolenschi C, Lambert A, Boige V, Tarabay A, Valery M, Fuerea A, Pudlarz T, Conroy T, Hollebecque A, Ducreux M. Role of molecular biology in the management of pancreatic cancer. World J Gastrointest Oncol 2024; 16:2902-2914. [PMID: 39072173 PMCID: PMC11271790 DOI: 10.4251/wjgo.v16.i7.2902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/04/2024] [Accepted: 05/21/2024] [Indexed: 07/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant challenges in patient management due to a dismal prognosis, increasing incidence, and limited treatment options. In this regard, precision medicine, which personalizes treatments based on tumour molecular characteristics, has gained great interest. However, its widespread implementation is not fully endorsed in current recommendations. This review explores key molecular alterations in PDAC, while emphasizing differences between KRAS-mutated and KRAS-wild-type tumours. It assesses the practical application of precision medicine in clinical settings and outlines potential future directions with respect to PDAC. Actionable molecular targets are examined with the aim of enhancing our understanding of PDAC molecular biology. Insights from this analysis may contribute to a more refined and personalized approach to pancreatic cancer treatment, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Alice Boileve
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | | | - Aurélien Lambert
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | - Valérie Boige
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Anthony Tarabay
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Marine Valery
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Alina Fuerea
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thomas Pudlarz
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| | - Thierry Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Nancy 54519, France
| | | | - Michel Ducreux
- Department of Medical, Gustave Roussy, Villejuif 94800, France
| |
Collapse
|
32
|
Klümper N, Tran NK, Zschäbitz S, Hahn O, Büttner T, Roghmann F, Bolenz C, Zengerling F, Schwab C, Nagy D, Toma M, Kristiansen G, Heers H, Ivanyi P, Niegisch G, Grunewald CM, Darr C, Farid A, Schlack K, Abbas M, Aydogdu C, Casuscelli J, Mokry T, Mayr M, Niedersüß-Beke D, Rausch S, Dietrich D, Saal J, Ellinger J, Ritter M, Alajati A, Kuppe C, Meeks J, Vera Badillo FE, Nakauma-González JA, Boormans J, Junker K, Hartmann A, Grünwald V, Hölzel M, Eckstein M. NECTIN4 Amplification Is Frequent in Solid Tumors and Predicts Enfortumab Vedotin Response in Metastatic Urothelial Cancer. J Clin Oncol 2024; 42:2446-2455. [PMID: 38657187 PMCID: PMC11227306 DOI: 10.1200/jco.23.01983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 04/26/2024] Open
Abstract
PURPOSE The anti-NECTIN4 antibody-drug conjugate enfortumab vedotin (EV) is approved for patients with metastatic urothelial cancer (mUC). However, durable benefit is only achieved in a small, yet uncharacterized patient subset. NECTIN4 is located on chromosome 1q23.3, and 1q23.3 gains represent frequent copy number variations (CNVs) in urothelial cancer. Here, we aimed to evaluate NECTIN4 amplifications as a genomic biomarker to predict EV response in patients with mUC. MATERIALS AND METHODS We established a NECTIN4-specific fluorescence in situ hybridization (FISH) assay to assess the predictive value of NECTIN4 CNVs in a multicenter EV-treated mUC patient cohort (mUC-EV, n = 108). CNVs were correlated with membranous NECTIN4 protein expression, EV treatment responses, and outcomes. We also assessed the prognostic value of NECTIN4 CNVs measured in metastatic biopsies of non-EV-treated mUC (mUC-non-EV, n = 103). Furthermore, we queried The Cancer Genome Atlas (TCGA) data sets (10,712 patients across 32 cancer types) for NECTIN4 CNVs. RESULTS NECTIN4 amplifications are frequent genomic events in muscle-invasive bladder cancer (TCGA bladder cancer data set: approximately 17%) and mUC (approximately 26% in our mUC cohorts). In mUC-EV, NECTIN4 amplification represents a stable genomic alteration during metastatic progression and associates with enhanced membranous NECTIN4 protein expression. Ninety-six percent (27 of 28) of patients with NECTIN4 amplifications demonstrated objective responses to EV compared with 32% (24 of 74) in the nonamplified subgroup (P < .001). In multivariable Cox analysis adjusted for age, sex, and Bellmunt risk factors, NECTIN4 amplifications led to a 92% risk reduction for death (hazard ratio, 0.08 [95% CI, 0.02 to 0.34]; P < .001). In the mUC-non-EV, NECTIN4 amplifications were not associated with outcomes. TCGA Pan-Cancer analysis demonstrated that NECTIN4 amplifications occur frequently in other cancers, for example, in 5%-10% of breast and lung cancers. CONCLUSION NECTIN4 amplifications are genomic predictors of EV responses and long-term survival in patients with mUC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
| | - Ngoc Khanh Tran
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, National Center for Tumor Disease (NCT), University Hospital, Heidelberg, Germany
| | - Oliver Hahn
- Department of Urology and Pediatric Urology, Julius Maximilians University Medical Center of Würzburg, Würzburg, Germany
| | - Thomas Büttner
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Florian Roghmann
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology, Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Christian Bolenz
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology and Pediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Friedemann Zengerling
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology and Pediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Constantin Schwab
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Dora Nagy
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Marieta Toma
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Glen Kristiansen
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Hendrik Heers
- Department of Urology, University Hospital Marburg, Marburg, Germany
| | - Philipp Ivanyi
- Department of Hemostaseology, Oncology and Stem Cell Transplantation, Medical University Hannover, Hannover, Germany
| | - Günter Niegisch
- Department of Urology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Christopher Darr
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Arian Farid
- Department of Urology, University Medical Center Göttingen, Göttingen, Germany
| | - Katrin Schlack
- Department of Urology, University Hospital Münster, Münster, Germany
| | - Mahmoud Abbas
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Can Aydogdu
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jozefina Casuscelli
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Theresa Mokry
- Department of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Mayr
- Clinic Ottakring, Institute of Pathology and Microbiology, Wien, Austria
| | | | - Steffen Rausch
- Department of Urology, Eberhard Karls University, Tübingen, Germany
| | - Dimo Dietrich
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Jonas Saal
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Jörg Ellinger
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Manuel Ritter
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
| | - Abdullah Alajati
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology and Division of Nephrology, RWTH Aachen University, Aachen, Germany
| | - Joshua Meeks
- Department of Urology, Feinberg School of Medicine, Chicago, IL
| | | | - J. Alberto Nakauma-González
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Joost Boormans
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, Homburg, Germany
| | - Arndt Hartmann
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Center for Cancer Research (Bayerisches Zentrum für Krebsforschung, BZKF), Erlangen, Germany
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumor Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Markus Eckstein
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Center for Cancer Research (Bayerisches Zentrum für Krebsforschung, BZKF), Erlangen, Germany
| |
Collapse
|
33
|
Mosele MF, Westphalen CB, Stenzinger A, Barlesi F, Bayle A, Bièche I, Bonastre J, Castro E, Dienstmann R, Krämer A, Czarnecka AM, Meric-Bernstam F, Michiels S, Miller R, Normanno N, Reis-Filho J, Remon J, Robson M, Rouleau E, Scarpa A, Serrano C, Mateo J, André F. Recommendations for the use of next-generation sequencing (NGS) for patients with advanced cancer in 2024: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2024; 35:588-606. [PMID: 38834388 DOI: 10.1016/j.annonc.2024.04.005] [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: 02/20/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Advancements in the field of precision medicine have prompted the European Society for Medical Oncology (ESMO) Precision Medicine Working Group to update the recommendations for the use of tumour next-generation sequencing (NGS) for patients with advanced cancers in routine practice. METHODS The group discussed the clinical impact of tumour NGS in guiding treatment decision using the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT) considering cost-effectiveness and accessibility. RESULTS As for 2020 recommendations, ESMO recommends running tumour NGS in advanced non-squamous non-small-cell lung cancer, prostate cancer, colorectal cancer, cholangiocarcinoma, and ovarian cancer. Moreover, it is recommended to carry out tumour NGS in clinical research centres and under specific circumstances discussed with patients. In this updated report, the consensus within the group has led to an expansion of the recommendations to encompass patients with advanced breast cancer and rare tumours such as gastrointestinal stromal tumours, sarcoma, thyroid cancer, and cancer of unknown primary. Finally, ESMO recommends carrying out tumour NGS to detect tumour-agnostic alterations in patients with metastatic cancers where access to matched therapies is available. CONCLUSION Tumour NGS is increasingly expanding its scope and application within oncology with the aim of enhancing the efficacy of precision medicine for patients with cancer.
Collapse
Affiliation(s)
- M F Mosele
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - C B Westphalen
- Comprehensive Cancer Center Munich & Department of Medicine III, University Hospital, LMU Munich, Munich
| | - A Stenzinger
- Institute of Pathology, University Hospital Heidelberg and Center for Personalized Medicine (ZPM), Heidelberg, Germany
| | - F Barlesi
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre
| | - A Bayle
- Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre; Drug Development Department (DITEP), Gustave Roussy, Villejuif; Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - I Bièche
- Department of Genetics, Institut Curie, INSERM U1016, Université Paris Cité, Paris, France
| | - J Bonastre
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - E Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid
| | - R Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona; University of Vic-Central University of Catalonia, Vic, Spain; Oncoclínicas, São Paulo, Brazil
| | - A Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg; Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - A M Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw; Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - F Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Michiels
- Oncostat U1018, Inserm, Université Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif; Service de Biostatistique et Epidémiologie, Gustave Roussy, Villejuif
| | - R Miller
- Department of Medical Oncology, University College London, London; Department of Medical Oncology, St Bartholomew's Hospital, London, UK
| | - N Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - J Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - J Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - M Robson
- Breast Medicine and Clinical Genetics Services, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - E Rouleau
- Tumor Genetics Service, Medical Biology and Pathology Department, Gustave Roussy, Villejuif, France
| | - A Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona-School of Medicine, Verona, Italy
| | - C Serrano
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - J Mateo
- Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona
| | - F André
- INSERM U981, Gustave Roussy, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Faculty of Medicine, Université Paris-Saclay, Kremlin Bicêtre.
| |
Collapse
|
34
|
García-Foncillas J, Arnold D, Avouac B, Lassen U, Laurent-Puig P, Taieb J, Helland Å, Stenzinger A. Overcoming barriers in biomarker testing. Letter to the Editor regarding: 'ESMO study on the availability and accessibility of biomolecular technologies in oncology in Europe', by A. Bayle et al. Ann Oncol 2024; 35:677-678. [PMID: 38723866 DOI: 10.1016/j.annonc.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 06/06/2024] Open
Affiliation(s)
- J García-Foncillas
- University Cancer Institute & The Department of Oncology, University Hospital Fundacion Jimenez Diaz, Autonomous University, Madrid, Spain.
| | - D Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| | | | - U Lassen
- Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - P Laurent-Puig
- Institut du Cancer Paris CARPEM, APHP, Université Paris Cité, Paris, France
| | - J Taieb
- Georges Pompidou European Hospital SIRIC-CARPEM, Université Paris Cité, Paris, France
| | - Å Helland
- Division of Cancer Medicine, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - A Stenzinger
- Institute of Pathology and Center for Personalized Medicine (ZPM), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
35
|
Corbaux P, Bayle A, Besle S, Vinceneux A, Vanacker H, Ouali K, Hanvic B, Baldini C, Cassier PA, Terret C, Verlingue L. Patients' selection and trial matching in early-phase oncology clinical trials. Crit Rev Oncol Hematol 2024; 196:104307. [PMID: 38401694 DOI: 10.1016/j.critrevonc.2024.104307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Early-phase clinical trials (EPCT) represent an important part of innovations in medical oncology and a valuable therapeutic option for patients with metastatic cancers, particularly in the era of precision medicine. Nevertheless, adult patients' participation in oncology clinical trials is low, ranging from 2% to 8% worldwide, with unequal access, and up to 40% risk of early discontinuation in EPCT, mostly due to cancer-related complications. DESIGN We review the tools and initiatives to increase patients' orientation and access to early phase cancer clinical trials, and to limit early discontinuation. RESULTS New approaches to optimize the early-phase clinical trial referring process in oncology include automatic trial matching, tools to facilitate the estimation of patients' prognostic and/or to better predict patients' eligibility to clinical trials. Classical and innovative approaches should be associated to double patient recruitment, improve clinical trial enrollment experience and reduce early discontinuation rates. CONCLUSIONS Whereas EPCT are essential for patients to access the latest medical innovations in oncology, offering the appropriate trial when it is relevant for patients should increase by organizational and technological innovations. The oncologic community will need to closely monitor their performance, portability and simplicity for implementation in daily clinical practice.
Collapse
Affiliation(s)
- P Corbaux
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Medical Oncology, Institut de Cancérologie et d'Hématologie Universitaire de Saint-Étienne (ICHUSE), Centre Hospitalier Universitaire de Saint-Etienne, France
| | - A Bayle
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - S Besle
- Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - A Vinceneux
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - H Vanacker
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - K Ouali
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - B Hanvic
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - C Baldini
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - P A Cassier
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - C Terret
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - L Verlingue
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France.
| |
Collapse
|
36
|
Passaro A, Al Bakir M, Hamilton EG, Diehn M, André F, Roy-Chowdhuri S, Mountzios G, Wistuba II, Swanton C, Peters S. Cancer biomarkers: Emerging trends and clinical implications for personalized treatment. Cell 2024; 187:1617-1635. [PMID: 38552610 PMCID: PMC7616034 DOI: 10.1016/j.cell.2024.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 04/02/2024]
Abstract
The integration of cancer biomarkers into oncology has revolutionized cancer treatment, yielding remarkable advancements in cancer therapeutics and the prognosis of cancer patients. The development of personalized medicine represents a turning point and a new paradigm in cancer management, as biomarkers enable oncologists to tailor treatments based on the unique molecular profile of each patient's tumor. In this review, we discuss the scientific milestones of cancer biomarkers and explore future possibilities to improve the management of patients with solid tumors. This progress is primarily attributed to the biological characterization of cancers, advancements in testing methodologies, elucidation of the immune microenvironment, and the ability to profile circulating tumor fractions. Integrating these insights promises to continually advance the precision oncology field, fostering better patient outcomes.
Collapse
Affiliation(s)
- Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Emily G Hamilton
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Fabrice André
- Gustave-Roussy Cancer Center, Paris Saclay University, Villejuif, France
| | - Sinchita Roy-Chowdhuri
- Department of Anatomic Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Department of Oncology, University College London Hospitals, London, UK
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
37
|
Puiu A, Gómez Tapia C, Weiss MER, Singh V, Kamen A, Siebert M. Prediction uncertainty estimates elucidate the limitation of current NSCLC subtype classification in representing mutational heterogeneity. Sci Rep 2024; 14:6779. [PMID: 38514696 PMCID: PMC10958018 DOI: 10.1038/s41598-024-57057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
The heterogeneous pathogenesis and treatment response of non-small cell lung cancer (NSCLC) has led clinical treatment decisions to be guided by NSCLC subtypes, with lung adenocarcinoma and lung squamous cell carcinoma being the most common subtypes. While histology-based subtyping remains challenging, NSCLC subtypes were found to be distinct at the transcriptomic level. However, unlike genomic alterations, gene expression is generally not assessed in clinical routine. Since subtyping of NSCLC has remained elusive using mutational data, we aimed at developing a neural network model that simultaneously learns from adenocarcinoma and squamous cell carcinoma samples of other tissue types and is regularized using a neural network model trained from gene expression data. While substructures of the expression-based manifold were captured in the mutation-based manifold, NSCLC classification accuracy did not significantly improve. However, performance was increased when rejecting inconclusive samples using an ensemble-based approach capturing prediction uncertainty. Importantly, SHAP analysis of misclassified samples identified co-occurring mutations indicative of both NSCLC subtypes, questioning the current NSCLC subtype classification to adequately represent inherent mutational heterogeneity. Since our model captures mutational patterns linked to clinical heterogeneity, we anticipate it to be suited as foundational model of genomic data for clinically relevant prognostic or predictive downstream tasks.
Collapse
Affiliation(s)
- Andrei Puiu
- Advanta, Siemens SRL, Brasov, 500007, Romania
- Automation and Information Technology, Transilvania University of Brasov, Brasov, 500174, Romania
| | - Carlos Gómez Tapia
- Digital Technology and Innovation, Siemens Healthineers, Erlangen, 91052, Germany
| | - Maximilian E R Weiss
- Digital Technology and Innovation, Siemens Healthineers, Erlangen, 91052, Germany
| | - Vivek Singh
- Digital Technology and Innovation, Siemens Healthineers, Princeton, 08540, USA
| | - Ali Kamen
- Digital Technology and Innovation, Siemens Healthineers, Princeton, 08540, USA
| | - Matthias Siebert
- Digital Technology and Innovation, Siemens Healthineers, Erlangen, 91052, Germany.
| |
Collapse
|
38
|
Houda I, Dickhoff C, Uyl-de Groot CA, Damhuis RA, Reguart N, Provencio M, Levy A, Dziadziuszko R, Pompili C, Di Maio M, Thomas M, Brunelli A, Popat S, Senan S, Bahce I. Challenges and controversies in resectable non-small cell lung cancer: a clinician's perspective. THE LANCET REGIONAL HEALTH. EUROPE 2024; 38:100841. [PMID: 38476749 PMCID: PMC10928275 DOI: 10.1016/j.lanepe.2024.100841] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 03/14/2024]
Abstract
The treatment landscape of resectable early-stage non-small cell lung cancer (NSCLC) is transforming due to the approval of novel adjuvant and neoadjuvant systemic treatments. The European Medicines Agency (EMA) recently approved adjuvant osimertinib, adjuvant atezolizumab, adjuvant pembrolizumab, and neoadjuvant nivolumab combined with chemotherapy, and the approval of other agents or new indications may follow soon. Despite encouraging results, many unaddressed questions remain. Moreover, the transformed treatment paradigm in resectable NSCLC can pose major challenges to healthcare systems and magnify existing disparities in care as differences in reimbursement may vary across different European countries. This Viewpoint discusses the challenges and controversies in resectable early-stage NSCLC and how existing inequalities in access to these treatments could be addressed.
Collapse
Affiliation(s)
- Ilias Houda
- Department of Pulmonary Medicine, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV Amsterdam, the Netherlands
| | - Chris Dickhoff
- Department of Cardiothoracic Surgery, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV Amsterdam, the Netherlands
| | - Carin A. Uyl-de Groot
- Erasmus School of Health Policy & Management/Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR Rotterdam, the Netherlands
| | - Ronald A.M. Damhuis
- Department of Research, Netherlands Comprehensive Cancer Organisation, Godebaldkwartier 419, 3511 DT Utrecht, the Netherlands
| | - Noemi Reguart
- Department of Medical Oncology, Hospital Clínic de Barcelona, C. de Villarroel, 170, 08036 Barcelona, Spain
| | - Mariano Provencio
- Department of Medical Oncology, Hospital Universitario Puerta De Hierro, C. Joaquín Rodrigo, 1, Majadahonda, 28222 Madrid, Spain
| | - Antonin Levy
- Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Université Paris Saclay, Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif, France
| | - Rafal Dziadziuszko
- Faculty of Medicine, Department of Oncology and Radiotherapy, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Cecilia Pompili
- Department of Thoracic Surgery, University and Hospital Trust – Ospedale Borgo Trento, P.Le A. Stefani, 1, 37126 Verona, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Medical Oncology 1U, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and Heidelberg University Hospital, Im Neuenheimer Feld 672, 69120 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research, Heidelberg, Germany
| | - Alessandro Brunelli
- Department of Thoracic Surgery, St. James’s University Hospital, Beckett Street, LS9 7TF Leeds, UK
| | - Sanjay Popat
- Lung Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, SW3 6JJ London, UK
| | - Suresh Senan
- Department of Radiation Oncology, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV Amsterdam, the Netherlands
| | - Idris Bahce
- Department of Pulmonary Medicine, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV Amsterdam, the Netherlands
| |
Collapse
|
39
|
Klümper N, Eckstein M. Biomarkers of Response to Anti-NECTIN4 Antibody-Drug Conjugate Enfortumab Vedotin in Urothelial Cancer. Eur Urol Focus 2024; 10:224-226. [PMID: 38631991 DOI: 10.1016/j.euf.2024.04.001] [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: 02/07/2024] [Revised: 03/20/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024]
Abstract
Initial studies indicated that NECTIN4 expression is widespread in metastatic urothelial cancer (mUC), which led to approval of the anti-NECTIN4 antibody-drug conjugate (ADC) enfortumab vedotin (EV) for unselected patients with mUC. However, the recent literature suggests that there has been overestimation of membranous NECTIN4 expression in UC, which is a prerequisite for EV binding. It is well established from the development of Her2-targeting ADCs that treatment response is strongly dependent on membranous expression level of the relevant target antigen. In this context, it has been demonstrated that membranous NECTIN4 expression correlates with EV responses and outcomes. Another promising biomarker could be NECTIN4 copy number alteration, a genomic alteration that occurs in approximately 25% of mUC cases, which is associated with strong membranous NECTIN4 expression. Patients with NECTIN4 amplification exhibit an objective response rate of >90% to EV monotherapy and long-term survival. Given the heterogeneous expression of NECTIN4 in UC, future biomarker research is essential for the development of biomarker-driven mUC treatment strategies to further improve outcomes for patients with mUC. PATIENT SUMMARY: We reviewed current evidence on biomarkers for predicting response to enfortumab vedotin (EV) treatment for metastatic urinary tract cancer (mUC). Studies to date have shown that patients with high levels of the protein NECTIN4 on their cancer cells respond well to EV. This information has the potential to guide future treatment strategies for mUC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology, University Hospital Bonn, Bonn, Germany; Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf, Bonn, Germany; Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany.
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Bavarian Center for Cancer Research
| |
Collapse
|
40
|
de Jager VD, Timens W, Bayle A, Botling J, Brcic L, Büttner R, Fernandes MGO, Havel L, Hochmair MJ, Hofman P, Janssens A, Johansson M, van Kempen L, Kern I, Lopez-Rios F, Lüchtenborg M, Machado JC, Mohorcic K, Paz-Ares L, Popat S, Ryška A, Taniere P, Wolf J, Schuuring E, van der Wekken AJ. Developments in predictive biomarker testing and targeted therapy in advanced stage non-small cell lung cancer and their application across European countries. THE LANCET REGIONAL HEALTH. EUROPE 2024; 38:100838. [PMID: 38476742 PMCID: PMC10928289 DOI: 10.1016/j.lanepe.2024.100838] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/16/2023] [Accepted: 01/08/2024] [Indexed: 03/14/2024]
Abstract
In the past two decades, the treatment of metastatic non-small cell lung cancer (NSCLC), has undergone significant changes due to the introduction of targeted therapies and immunotherapy. These advancements have led to the need for predictive molecular tests to identify patients eligible for targeted therapy. This review provides an overview of the development and current application of targeted therapies and predictive biomarker testing in European patients with advanced stage NSCLC. Using data from eleven European countries, we conclude that recommendations for predictive testing are incorporated in national guidelines across Europe, although there are differences in their comprehensiveness. Moreover, the availability of recently EMA-approved targeted therapies varies between European countries. Unfortunately, routine assessment of national/regional molecular testing rates is limited. As a result, it remains uncertain which proportion of patients with metastatic NSCLC in Europe receive adequate predictive biomarker testing. Lastly, Molecular Tumor Boards (MTBs) for discussion of molecular test results are widely implemented, but national guidelines for their composition and functioning are lacking. The establishment of MTB guidelines can provide a framework for interpreting rare or complex mutations, facilitating appropriate treatment decision-making, and ensuring quality control.
Collapse
Affiliation(s)
- Vincent D. de Jager
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arnaud Bayle
- Oncostat U1018, Inserm, Paris-Saclay University, Gustave Roussy, Villejuif, France
| | - Johan Botling
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy of University of Gothenburg, Gothenburg, Sweden
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Reinhard Büttner
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | | | - Libor Havel
- Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Maximilian J. Hochmair
- Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
- Department of Respiratory and Critical Care Medicine, Klinik Floridsdorf, Vienna Healthcare Group, Vienna, Austria
| | - Paul Hofman
- IHU RespirERA, FHU OncoAge, Nice University Hospital, Côte d’Azur University, Nice, France
| | - Annelies Janssens
- Department of Oncology, University Hospital Antwerp, University of Antwerp, Edegem, Belgium
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Léon van Kempen
- Department of Pathology, University Hospital Antwerp, University of Antwerp, Edegem, Belgium
| | - Izidor Kern
- Laboratory for Cytology and Pathology, University Clinic Golnik, Golnik, Slovenia
| | - Fernando Lopez-Rios
- Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i+12), Ciberonc, Madrid, Spain
| | - Margreet Lüchtenborg
- National Disease Registration Service, NHS England, London, United Kingdom
- Centre for Cancer, Society & Public Health, King’s College London, London, United Kingdom
| | - José Carlos Machado
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Faculty of Medicine of the University of Porto, Institute for Research and Innovation in Health (i3S), Porto, Portugal
| | - Katja Mohorcic
- University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | - Luis Paz-Ares
- Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, H12O-CNIO Lung Cancer Clinical Research Unit, Research Institute Hospital 12 de Octubre (i+12)/Spanish National Cancer Research Center (CNIO), Ciberonc, Madrid, Spain
| | - Sanjay Popat
- Lung Unit, Royal Marsden NHS Trust, London, United Kingdom
| | - Aleš Ryška
- The Fingerland Department of Pathology, Charles University Medical Faculty and University Hospital, Czech Republic
| | - Phillipe Taniere
- Department of Histopathology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Jürgen Wolf
- Lung Cancer Group Cologne, Department I for Internal Medicine and Center for Integrated Oncology Cologne/Bonn, University Hospital Cologne, Cologne, Germany
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anthonie J. van der Wekken
- Department of Pulmonary Diseases and Tuberculosis, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
41
|
de Jager VD, Timens W, Bayle A, Botling J, Brcic L, Büttner R, Fernandes MGO, Havel L, Hochmair M, Hofman P, Janssens A, van Kempen L, Kern I, Machado JC, Mohorčič K, Popat S, Ryška A, Wolf J, Schuuring E, van der Wekken AJ. Future perspective for the application of predictive biomarker testing in advanced stage non-small cell lung cancer. THE LANCET REGIONAL HEALTH. EUROPE 2024; 38:100839. [PMID: 38476751 PMCID: PMC10928270 DOI: 10.1016/j.lanepe.2024.100839] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/16/2023] [Accepted: 01/08/2024] [Indexed: 03/14/2024]
Abstract
For patients with advanced stage non-small cell lung cancer (NSCLC), treatment strategies have changed significantly due to the introduction of targeted therapies and immunotherapy. In the last few years, we have seen an explosive growth of newly introduced targeted therapies in oncology and this development is expected to continue in the future. Besides primary targetable aberrations, emerging diagnostic biomarkers also include relevant co-occurring mutations and resistance mechanisms involved in disease progression, that have impact on optimal treatment management. To accommodate testing of pending biomarkers, it is necessary to establish routine large-panel next-generation sequencing (NGS) for all patients with advanced stage NSCLC. For cost-effectiveness and accessibility, it is recommended to implement predictive molecular testing using large-panel NGS in a dedicated, centralized expert laboratory within a regional oncology network. The central molecular testing center should host a regional Molecular Tumor Board and function as a hub for interpretation of rare and complex testing results and clinical decision-making.
Collapse
Affiliation(s)
- Vincent D. de Jager
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arnaud Bayle
- Oncostat U1018, Inserm, Paris-Saclay University, Gustave Roussy, Villejuif, France
| | - Johan Botling
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy of University of Gothenburg, Gothenburg, Sweden
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Reinhard Büttner
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | | | - Libor Havel
- Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Maximilian Hochmair
- Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
- Department of Respiratory and Critical Care Medicine, Klinik Floridsdorf, Vienna Healthcare Group, Vienna, Austria
| | - Paul Hofman
- IHU RespirERA, FHU OncoAge, Nice University Hospital, Côte d’Azur University, Nice, France
| | - Annelies Janssens
- Department of Oncology, University Hospital Antwerp, University of Antwerp, Edegem, Belgium
| | - Léon van Kempen
- Department of Pathology, University Hospital Antwerp, University of Antwerp, Edegem, Belgium
| | - Izidor Kern
- Laboratory for Cytology and Pathology, University Clinic Golnik, Golnik, Slovenia
| | - José Carlos Machado
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Faculty of Medicine of the University of Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Porto, Portugal
| | - Katja Mohorčič
- University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | - Sanjay Popat
- Lung Unit, Royal Marsden NHS Trust, London, England, UK
| | - Aleš Ryška
- The Fingerland Department of Pathology, Charles University Medical Faculty and University Hospital, Czech Republic
| | - Jürgen Wolf
- Lung Cancer Group Cologne, Department I for Internal Medicine and Center for Integrated Oncology Cologne/Bonn, University Hospital Cologne, Cologne, Germany
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anthonie J. van der Wekken
- Department of Pulmonary Diseases and Tuberculosis, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
42
|
Esposito Abate R, Pasquale R, Sacco A, Simeon V, Maiello MR, Frezzetti D, Chiodini P, Normanno N. Harmonization of tumor mutation burden testing with comprehensive genomic profiling assays: an IQN Path initiative. J Immunother Cancer 2024; 12:e007800. [PMID: 38309725 PMCID: PMC10840060 DOI: 10.1136/jitc-2023-007800] [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] [Accepted: 12/29/2023] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Although conflicting results emerged from different studies, the tumor mutational burden (TMB) appears as one of most reliable biomarkers of sensitivity to immune checkpoint inhibitors. Several laboratories are reporting TMB values when performing comprehensive genomic profiling (CGP) without providing a clinical interpretation, due to the lack of validated cut-off values. The International Quality Network for Pathology launched an initiative to harmonize TMB testing with CGP assay and favor the clinical implementation of this biomarker. METHODS TMB evaluation was performed with three commercially available CGP panels, TruSight Oncology 500 (TSO500), Oncomine Comprehensive Plus Assay (OCA) and QIAseq Multimodal Panel (QIA), versus the reference assay FoundationOne CDx (F1CDx). Archived clinical samples derived from 60 patients with non-small cell lung cancer were used for TMB assessment. Adjusted cut-off values for each panel were calculated. RESULTS Testing was successful for 91.7%, 100%, 96.7% and 100% of cases using F1CDx, TSO500, OCA and QIA, respectively. The matrix comparison analysis, between the F1CDx and CGP assays, showed a linear correlation for all three panels, with a higher correlation between F1CDx and TSO500 (rho=0.88) than in the other two comparisons (rho=0.77 for QIA; 0.72 for OCA). The TSO500 showed the best area under the curve (AUC, value 0.96), with a statistically significant difference when compared with the AUC of OCA (0.83, p value=0.01) and QIA (0.88, p value=0.028). The Youden Index calculation allowed us to extrapolate TMB cut-offs of the different panels corresponding to the 10 mutations/megabase (muts/Mb) cut-off of F1CDx: 10.19, 10.4 and 12.37 muts/Mb for TSO500, OCA and QIA, respectively. Using these values, we calculated the relative accuracy measures for the three panels. TSO500 showed 86% specificity and 96% sensitivity, while OCA and QIA had lower yet similar values of specificity and sensitivity (73% and 88%, respectively). CONCLUSION This study estimated TMB cut-off values for commercially available CGP panels. The results showed a good performance of all panels on clinical samples and the calculated cut-offs support better accuracy measures for TSO500. The validated cut-off values can drive clinical interpretation of TMB testing in clinical research and clinical practice.
Collapse
Affiliation(s)
- Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G.Pascale, Napoli, Italy
| | | | - Alessandra Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G.Pascale, Napoli, Italy
| | - Vittorio Simeon
- Medical Statistics Unit, Department of Mental Health and Public Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Monica Rosaria Maiello
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G.Pascale, Napoli, Italy
| | - Daniela Frezzetti
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G.Pascale, Napoli, Italy
| | - Paolo Chiodini
- Medical Statistics Unit, Department of Mental Health and Public Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori IRCCS Fondazione G.Pascale, Napoli, Italy
| |
Collapse
|
43
|
Hofman P, Berezowska S, Kazdal D, Mograbi B, Ilié M, Stenzinger A, Hofman V. Current challenges and practical aspects of molecular pathology for non-small cell lung cancers. Virchows Arch 2024; 484:233-246. [PMID: 37801103 PMCID: PMC10948551 DOI: 10.1007/s00428-023-03651-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
The continuing evolution of treatment options in thoracic oncology requires the pathologist to regularly update diagnostic algorithms for management of tumor samples. It is essential to decide on the best way to use tissue biopsies, cytological samples, as well as liquid biopsies to identify the different mandatory predictive biomarkers of lung cancers in a short turnaround time. However, biological resources and laboratory member workforce are limited and may be not sufficient for the increased complexity of molecular pathological analyses and for complementary translational research development. In this context, the surgical pathologist is the only one who makes the decisions whether or not to send specimens to immunohistochemical and molecular pathology platforms. Moreover, the pathologist can rapidly contact the oncologist to obtain a new tissue biopsy and/or a liquid biopsy if he/she considers that the biological material is not sufficient in quantity or quality for assessment of predictive biomarkers. Inadequate control of algorithms and sampling workflow may lead to false negative, inconclusive, and incomplete findings, resulting in inappropriate choice of therapeutic strategy and potentially poor outcome for patients. International guidelines for lung cancer treatment are based on the results of the expression of different proteins and on genomic alterations. These guidelines have been established taking into consideration the best practices to be set up in clinical and molecular pathology laboratories. This review addresses the current predictive biomarkers and algorithms for use in thoracic oncology molecular pathology as well as the central role of the pathologist, notably in the molecular tumor board and her/his participation in the treatment decision-making. The perspectives in this setting will be discussed.
Collapse
Affiliation(s)
- Paul Hofman
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France.
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France.
| | - Sabina Berezowska
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniel Kazdal
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Baharia Mograbi
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| | - Marius Ilié
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Véronique Hofman
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| |
Collapse
|
44
|
Febbo PG, Allo M, Alme EB, Cuyun Carter G, Dumanois R, Essig A, Kiernan E, Kubler CB, Martin N, Popescu MC, Leiman LC. Recommendations for the Equitable and Widespread Implementation of Liquid Biopsy for Cancer Care. JCO Precis Oncol 2024; 8:e2300382. [PMID: 38166232 PMCID: PMC10803048 DOI: 10.1200/po.23.00382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/07/2023] [Accepted: 10/12/2023] [Indexed: 01/04/2024] Open
Abstract
Liquid biopsies-tests that detect circulating tumor cellular components in the bloodstream-have the potential to transform cancer by reducing health inequities in screening, diagnostics, and monitoring. Today, liquid biopsies are being used to guide treatment choices for patients and monitor for cancer recurrence, and promising work in multi-cancer early detection is ongoing. However, without awareness of the barriers to adoption of this new technology and a willingness to build mitigation efforts into the implementation of widespread liquid biopsy testing, the communities that could most benefit may be the last to access and use them. In this work, we review the challenges likely to affect the accessibility of liquid biopsies in both the general population and underserved populations, and recommend specific actions to facilitate equitable access for all patients.
Collapse
|
45
|
Remon J, Lopez A, Planchard D, Besse B. Are we ready to escalate or de-escalate immune treatment strategies in NSCLC based on liquid biopsy data? Eur J Cancer 2023; 195:113369. [PMID: 37913681 DOI: 10.1016/j.ejca.2023.113369] [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: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023]
Affiliation(s)
- Jordi Remon
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France.
| | - Alvaro Lopez
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - David Planchard
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
46
|
Lamarca A, Vogel A. Futibatinib: second EMA approval for FGFR inhibitor in cholangiocarcinoma. ESMO Open 2023; 8:102049. [PMID: 37922686 PMCID: PMC10651450 DOI: 10.1016/j.esmoop.2023.102049] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Affiliation(s)
- A Lamarca
- Department of Medical Oncology - OncoHealth Institute, Fundación Jiménez Díaz University Hospital, Madrid, Spain; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK.
| | - A Vogel
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, Canada; Medical Oncology, Margaret Cancer Centre, Toronto, Canada
| |
Collapse
|
47
|
Aldea M, Vasseur D, Italiano A, Nikolaev SI. WGS/WES-RNAseq compared to targeted NGS in oncology: is there something to unlock? Ann Oncol 2023; 34:1090-1093. [PMID: 37816462 DOI: 10.1016/j.annonc.2023.09.3118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023] Open
Affiliation(s)
- M Aldea
- Department of Medical Oncology, Gustave Roussy, Villejuif; Paris-Saclay University, Kremlin-Bicetre; Precision Medicine, Gustave Roussy, Villejuif
| | - D Vasseur
- Precision Medicine, Gustave Roussy, Villejuif; Department of Molecular Pathology, Gustave Roussy, Villejuif
| | - A Italiano
- Precision Medicine, Gustave Roussy, Villejuif; Drug Development Department, Gustave Roussy, Villejuif
| | | |
Collapse
|
48
|
Stenzinger A, Moltzen EK, Winkler E, Molnar-Gabor F, Malek N, Costescu A, Jensen BN, Nowak F, Pinto C, Ottersen OP, Schirmacher P, Nordborg J, Seufferlein T, Fröhling S, Edsjö A, Garcia-Foncillas J, Normanno N, Lundgren B, Friedman M, Bolanos N, Tatton-Brown K, Hill S, Rosenquist R. Implementation of precision medicine in healthcare-A European perspective. J Intern Med 2023; 294:437-454. [PMID: 37455247 DOI: 10.1111/joim.13698] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
The technical development of high-throughput sequencing technologies and the parallel development of targeted therapies in the last decade have enabled a transition from traditional medicine to personalized treatment and care. In this way, by using comprehensive genomic testing, more effective treatments with fewer side effects are provided to each patient-that is, precision or personalized medicine (PM). In several European countries-such as in England, France, Denmark, and Spain-the governments have adopted national strategies and taken "top-down" decisions to invest in national infrastructure for PM. In other countries-such as Sweden, Germany, and Italy with regionally organized healthcare systems-the profession has instead taken "bottom-up" initiatives to build competence networks and infrastructure to enable equal access to PM. In this review, we summarize key learnings at the European level on the implementation process to establish sustainable governance and organization for PM at the regional, national, and EU/international levels. We also discuss critical ethical and legal aspects of implementing PM, and the importance of access to real-world data and performing clinical trials for evidence generation, as well as the need for improved reimbursement models, increased cross-disciplinary education and patient involvement. In summary, PM represents a paradigm shift, and modernization of healthcare and all relevant stakeholders-that is, healthcare, academia, policymakers, industry, and patients-must be involved in this system transformation to create a sustainable, non-siloed ecosystem for precision healthcare that benefits our patients and society at large.
Collapse
Affiliation(s)
- Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Germany
| | - Ejner K Moltzen
- Innovation Fund Denmark, International Consortium for Personalised Medicine (IC PerMed), Aarhus, Denmark
| | - Eva Winkler
- Section of Translational Medical Ethics, National Center for Tumour Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Nisar Malek
- Centers for Personalized Medicine (ZPM), Germany
- Department for Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | | | | | | | - Carmine Pinto
- Medical Oncology, Comprehensive Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Germany
| | - Jenni Nordborg
- Lif - The Research-Based Pharmaceutical Industry, Stockholm, Sweden
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - 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), Sweden
| | - Jesus Garcia-Foncillas
- Department of Oncology and Cancer Institute, Fundacion Jimenez Diaz University Hospital, Autonomous University, Madrid, Spain
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | | | - Mikaela Friedman
- Genomic Medicine Sweden (GMS), Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Katrina Tatton-Brown
- National Genomics Education, NHS England, London, UK
- St George's University Hospitals NHS Foundation Trust, London, UK
- St George's University of London, London, UK
| | - Sue Hill
- Office of Chief Scientific Officer and the Genomics Unit, NHS England, London, UK
| | - Richard Rosenquist
- Genomic Medicine Sweden (GMS), Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
49
|
Normanno N, Machado JC, Pescarmona E, Buglioni S, Navarro L, Esposito Abate R, Ferro A, Mensink R, Lambiase M, Lespinet-Fabre V, Calgua B, Jermann PM, Ilié M, Hofman P. European Real-World Assessment of the Clinical Validity of a CE-IVD Panel for Ultra-Fast Next-Generation Sequencing in Solid Tumors. Int J Mol Sci 2023; 24:13788. [PMID: 37762091 PMCID: PMC10531166 DOI: 10.3390/ijms241813788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Molecular profiling of solid tumors facilitates personalized, targeted therapeutic interventions. The ability to perform next-generation sequencing (NGS), especially from small tissue samples, in a short turnaround time (TAT) is essential to providing results that enable rapid clinical decisions. This multicenter study evaluated the performance of a CE in vitro diagnostic (IVD) assay, the Oncomine Dx Express Test, on the Ion Torrent Genexus System for detecting DNA and RNA variants in solid tumors. Eighty-two archived formalin-fixed paraffin embedded (FFPE) tissue samples from lung, colorectal, central nervous system, melanoma, breast, gastric, thyroid, and soft tissue cancers were used to assess the presence of single nucleotide variants (SNVs), insertions and deletions (indels), copy number variations (CNVs), gene fusions, and splice variants. These clinical samples were previously characterized at the various academic centers using orthogonal methods. The Oncomine Dx Express Test showed high performance with 100% concordance with previous characterization for SNVs, indels, CNVs, gene fusions, and splice variants. SNVs and indels with allele frequencies as low as 5% were correctly identified. The test detected all the expected ALK, RET, NTRK1, and ROS1 fusion isoforms and MET exon 14-skipping splice variants. The average TAT from extracted nucleic acids to the final variant report was 18.3 h. The Oncomine Dx Express Test in combination with the Ion Torrent Genexus System is a CE-IVD-compliant, performant, and multicenter reproducible method for NGS detection of actionable biomarkers from a range of tumor samples, providing results in a short TAT that could support timely decision- making for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - José Carlos Machado
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Edoardo Pescarmona
- I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy; (E.P.); (S.B.)
| | - Simonetta Buglioni
- I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy; (E.P.); (S.B.)
| | - Lara Navarro
- Consorcio Hospital General de Valencia, 46014 Valencia, Spain;
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - Anabela Ferro
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Rob Mensink
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Matilde Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| | - Byron Calgua
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (B.C.); (P.M.J.)
| | - Philip M. Jermann
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (B.C.); (P.M.J.)
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| |
Collapse
|
50
|
Hofman P. Implementation of the clinical practice of liquid biopsies for thoracic oncology the experience of the RespirERA university hospital institute (Nice, France). THE JOURNAL OF LIQUID BIOPSY 2023; 1:100004. [PMID: 40027288 PMCID: PMC11863941 DOI: 10.1016/j.jlb.2023.100004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 03/05/2025]
Abstract
According to international guidelines, it is mandatory to evaluate predictive biomarkers of targeted therapies and the response to immune check point inhibitors for patients with non-squamous non-small cell lung cancer (NS-NSCLC). For this purpose, a tissue sample is nowadays the gold standard, but biofluids, particularly peripheral blood, can be a complementary and sometimes an alternative approach to assess the status of different druggable genomic alterations of advanced NS-NSCLC. A liquid biopsy (LB) is an attractive approah for better treatment decision-making by thoracic oncologists for NSCLC patients in daily practice at both initial diagnosis and tumor progression. We describe the experience of a clinical and molecular pathology laboratory (LPCE, Nice, France) developing the use of in-house LB in thoracic oncology. Moreover, we report the changes in clinical care, the advantages, but also the possible constraints associated with implantation of LB in routine clinical practice.
Collapse
Affiliation(s)
- Paul Hofman
- Côte d’Azur University, IHU RespirERA, FHU OncoAge, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, Nice, France
- Côte d’Azur University, IRCAN, Inserm U1081, CNRS 7284, France
| |
Collapse
|