1
|
Denisenko TV, Ivanova AE, Koval A, Silachev DN, Jia L, Sukhikh GT, Katanaev VL. Signalomics for molecular tumor boards and precision oncology of breast and gynecological cancers. Mol Syst Biol 2025:10.1038/s44320-025-00125-1. [PMID: 40490498 DOI: 10.1038/s44320-025-00125-1] [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: 03/16/2025] [Revised: 05/08/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
Precision oncology led to the establishment and widespread application of molecular tumor boards (MTBs)-multidisciplinary units combining molecular and clinical assessment of individual cancer cases for swift selection of personalized treatments. Whole-exome or gene panel sequencing, combined with transcriptomic, immunohistochemical, and other molecular analyses, often permits dissection of molecular drivers of a tumor and identification of its potential targetable vulnerabilities, instructing clinical oncologists on sometimes unconventional treatment options. However, cancer drivers are often unleashed mutation-independently, especially in breast and gynecological cancers, and deleterious mutations are not always pathogenic. To complement the MTB arsenal, we chart here the molecular toolset we call Signalomics that permits fast and robust assessment of a panel of oncogenic signaling pathways in fresh tumor samples. Using transcriptional reporters introduced in primary tumor cells, this approach identifies the pathways overactivated in a given tumor and validates their sensitivity to targeted therapies, providing actionable insights for personalized treatment strategies. Integration of Signalomics into MTB workflows bridges the gap between molecular profiling and functional pathway analysis, refining clinical treatment decisions and advancing precision oncology.
Collapse
Affiliation(s)
- Tatiana V Denisenko
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 4 Akademika Oparina Str., Moscow, 117997, Russia
| | - Anna E Ivanova
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 4 Akademika Oparina Str., Moscow, 117997, Russia
| | - Alexey Koval
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211, Geneva, Switzerland
| | - Denis N Silachev
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 4 Akademika Oparina Str., Moscow, 117997, Russia
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211, Geneva, Switzerland
- Department of Functional Biochemistry of Biopolymers, A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992, Moscow, Russia
| | - Lee Jia
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Gennadiy T Sukhikh
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 4 Akademika Oparina Str., Moscow, 117997, Russia
| | - Vladimir L Katanaev
- Translational Research Centre in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211, Geneva, Switzerland.
- Translational Oncology Research Center, Qatar Biomedical Research Institute (QBRI), College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 34110, Doha, Qatar.
| |
Collapse
|
2
|
Carr A, Jackson JB, Coldren C, Chandra P, Koohestani F, Shiller M, Auber R. Tumor diagnosis recharacterization enabled by comprehensive genomic profiling to guide precision medicine strategy. NPJ Precis Oncol 2025; 9:149. [PMID: 40399445 PMCID: PMC12095656 DOI: 10.1038/s41698-025-00942-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 05/11/2025] [Indexed: 05/23/2025] Open
Abstract
Comprehensive genomic profiling (CGP) via next-generation sequencing is standard clinical practice for advanced and metastatic cancers in the U.S. and can help identify clinically actionable alterations in patients who may benefit from targeted therapies. CGP can also complement clinicopathological findings and in certain cases, may lead to diagnostic recharacterization resulting in more precise therapeutic strategies. Here, we highlight examples where molecular findings resulted in tumor re-evaluation and subsequent recharacterization. Twenty-eight cases where CGP results were inconsistent with initial pathological diagnosis and clinical presentation were selected for secondary clinicopathological review to explore alternative diagnostic explanations more consistent with the genomic results. Genomic profiling identified clinically actionable and prognostic variants leading to more accurate therapeutic recommendations based on the updated diagnoses highlighting the value of CGP beyond biomarker detection for therapy selection and supporting its complementary use in diagnostic confirmation to unveil opportunities for precision medicine strategies.
Collapse
Affiliation(s)
- Ann Carr
- PathGroup, Nashville, TN, 37217, USA.
| | | | | | | | | | | | | |
Collapse
|
3
|
Herrero Colomina J, Johnston E, Duffus K, Zaïr ZM, Thistlethwaite F, Krebs M, Carter L, Graham D, Cook N. Real-world experience of Molecular Tumour Boards for clinical decision-making for cancer patients. NPJ Precis Oncol 2025; 9:87. [PMID: 40133697 PMCID: PMC11937402 DOI: 10.1038/s41698-025-00863-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Molecular Tumour Boards (MTBs) play a crucial role in interpreting genomic results and providing treatment recommendations. We investigated the real-world impact of MTBs on clinical decision-making by surveying health care professionals (HCPs) across the UK; 44 participants from 11 MTBs took part in the study. 97.7% of respondents felt that MTBs increased awareness of available clinical trials matched to genomic alterations, 84% reported more confidence in interpreting genomic data, and 95.4% valued MTBs as educational. Hurdles to the discussion at MTBs included frequency and capacity of MTBs (ctDNA), sample collection and laboratory turnaround time (Tissue samples). One-third of respondents encountered challenges attending MTBs regularly due to workload. The survey highlighted areas for optimisation, such as meeting efficiency, rapid molecular analysis turnaround time, reliable trial matching tools, and ensuring MTBs are included in HCP's job plans.
Collapse
Affiliation(s)
| | | | - Kate Duffus
- The Christie NHS Foundation Trust, Manchester, UK
| | | | - Fiona Thistlethwaite
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Matthew Krebs
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Louise Carter
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Donna Graham
- The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Natalie Cook
- The Christie NHS Foundation Trust, Manchester, UK.
- Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
4
|
Wahida A, Kurzrock R. The Molecular Tumor Board Turns 10: The Age of Complexity. Oncologist 2025; 30:oyae271. [PMID: 39419766 PMCID: PMC11954507 DOI: 10.1093/oncolo/oyae271] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Gene sequencing has brought a titanic of complex data into clinical precision oncology. Deciphering this complexity for practice requires new constructs. In 2014, the Molecular Tumor Board (MTB) was introduced into the literature by a publication in The Oncologist. Ten years later, MTBs have become globally established vehicles that integrate rapidly emerging "omic" information, helping to transform cancer management.
Collapse
Affiliation(s)
- Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Centre Munich, Neuherberg, Germany
| | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, United States
- WIN Consortium, Paris, France
| |
Collapse
|
5
|
Berman E, Sundberg Malek H, Bitzer M, Malek N, Eickhoff C. Retrieval Augmented Therapy Suggestion for Molecular Tumor Boards: Algorithmic Development and Validation Study. J Med Internet Res 2025; 27:e64364. [PMID: 40053768 PMCID: PMC11923455 DOI: 10.2196/64364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/21/2024] [Accepted: 01/02/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Molecular tumor boards (MTBs) require intensive manual investigation to generate optimal treatment recommendations for patients. Large language models (LLMs) can catalyze MTB recommendations, decrease human error, improve accessibility to care, and enhance the efficiency of precision oncology. OBJECTIVE In this study, we aimed to investigate the efficacy of LLM-generated treatments for MTB patients. We specifically investigate the LLMs' ability to generate evidence-based treatment recommendations using PubMed references. METHODS We built a retrieval augmented generation pipeline using PubMed data. We prompted the resulting LLM to generate treatment recommendations with PubMed references using a test set of patients from an MTB conference at a large comprehensive cancer center at a tertiary care institution. Members of the MTB manually assessed the relevancy and correctness of the generated responses. RESULTS A total of 75% of the referenced articles were properly cited from PubMed, while 17% of the referenced articles were hallucinations, and the remaining were not properly cited from PubMed. Clinician-generated LLM queries achieved higher accuracy through clinician evaluation than automated queries, with clinicians labeling 25% of LLM responses as equal to their recommendations and 37.5% as alternative plausible treatments. CONCLUSIONS This study demonstrates how retrieval augmented generation-enhanced LLMs can be a powerful tool in accelerating MTB conferences, as LLMs are sometimes capable of achieving clinician-equal treatment recommendations. However, further investigation is required to achieve stable results with zero hallucinations. LLMs signify a scalable solution to the time-intensive process of MTB investigations. However, LLM performance demonstrates that they must be used with heavy clinician supervision, and cannot yet fully automate the MTB pipeline.
Collapse
Affiliation(s)
- Eliza Berman
- Center for Digital Health, University Hospital Tuebingen, Tuebingen, Germany
| | - Holly Sundberg Malek
- Center for Personalized Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Michael Bitzer
- Department of Internal Medicine I, University Hospital Tuebingen, Tuebingen, Germany
| | - Nisar Malek
- Department of Internal Medicine I, University Hospital Tuebingen, Tuebingen, Germany
| | - Carsten Eickhoff
- Center for Digital Health, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
6
|
Yang J, Wang M, Dönitz J, Chapuy B, Beißbarth T. Advancing personalized cancer therapy: Onko_DrugCombScreen-a novel Shiny app for precision drug combination screening. NAR Genom Bioinform 2025; 7:lqaf004. [PMID: 39897104 PMCID: PMC11783568 DOI: 10.1093/nargab/lqaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Identifying and validating genotype-guided drug combinations for a specific molecular subtype in cancer therapy represents an unmet medical need and is important in enhancing efficacy and reducing toxicity. However, the exponential increase in combinatorial possibilities constrains the ability to identify and validate effective drug combinations. In this context, we have developed Onko_DrugCombScreen, an innovative tool aiming at advancing precision medicine based on identifying significant drug combination candidates in a target cancer cohort compared to a comparison cohort. Onko_DrugCombScreen, inspired by the molecular tumor board process, synergizes drug knowledgebase analysis with various statistical methodologies and data visualization techniques to pinpoint drug combination candidates. Validated through a TCGA-BRCA case study, Onko_DrugCombScreen has demonstrated its proficiency in discerning established drug combinations in a specific cancer type and in revealing potential novel drug combinations. By enhancing the capability of drug combination discovery through drug knowledgebases, Onko_DrugCombScreen represents a significant advancement in personalized cancer treatment by identifying promising drug combinations, setting the stage for the development of more precise and potent combination treatments in cancer care. The Onko_DrugCombScreen Shiny app is available at https://rshiny.gwdg.de/apps/onko_drugcombscreen/. The Git repository can be accessed at https://gitlab.gwdg.de/MedBioinf/mtb/onko_drugcombscreen.
Collapse
Affiliation(s)
- Jingyu Yang
- Medical Bioinformatics, University Medical Center Göttingen, Göttingen, 37077, Germany
| | - Meng Wang
- Department of Hematology, Oncology, and Cancer Immunology, Charité—University Medical Center Berlin, Campus Benjamin Franklin, Berlin, 12203, Germany
| | - Jürgen Dönitz
- Medical Bioinformatics, University Medical Center Göttingen, Göttingen, 37077, Germany
- Campus Institute Data Science (CIDAS), University of Göttingen, Göttingen, 37073, Germany
- Comprehensive Cancer Center Niedersachsen (CCCN), Göttingen, 37075, Germany
| | - Björn Chapuy
- Department of Hematology, Oncology, and Cancer Immunology, Charité—University Medical Center Berlin, Campus Benjamin Franklin, Berlin, 12203, Germany
- Department of Hematology and Medical Oncology, Georg-August University Göttingen, Göttingen, 37075, Germany
| | - Tim Beißbarth
- Medical Bioinformatics, University Medical Center Göttingen, Göttingen, 37077, Germany
- Campus Institute Data Science (CIDAS), University of Göttingen, Göttingen, 37073, Germany
- Comprehensive Cancer Center Niedersachsen (CCCN), Göttingen, 37075, Germany
| |
Collapse
|
7
|
Jansen L, Nachtsheim L, Mayer M, Arolt C, Quaas A, Klußmann JP, Wolber P. [Clinical and molecular epidemiology of malignant salivary gland tumors]. Laryngorhinootologie 2025; 104:87-93. [PMID: 39419038 PMCID: PMC11790319 DOI: 10.1055/a-2373-5741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Salivary gland carcinomas are a rare and heterogeneous group of malignant tumors, accounting for 3-6% of all malignant tumors in the head and neck region. The 1-, 3- and 5-year survival rates are 83%, 69% and 63% respectively. Due to new molecular pathological and genetic findings, new entities are constantly being defined as part of the recurring WHO classification of salivary gland carcinomas, so that the incidence rates of the entities are subject to constant change. The only certain risk factor for the development of salivary gland carcinomas is ionizing radiation. In addition, large tumors, cervical lymph node involvement and perineural sheath involvement significantly worsen the prognosis. Today, molecular pathology is coming to the fore, with which potential targets have been identified that can offer prognosis-improving treatment options, particularly in recurrent or distant metastatic stages. Entity-specific tyrosine kinase inhibitors such as axitinib in adenoid cystic carcinoma or larotrectinib in secretory carcinoma and cross-entity therapies such as HER2 inhibition and androgen deprivation can prolong median and progression-free survival with a favorable side effect profile.
Collapse
Affiliation(s)
- Louis Jansen
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Lisa Nachtsheim
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Marcel Mayer
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Christoph Arolt
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Alexander Quaas
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Jens Peter Klußmann
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| | - Philipp Wolber
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Kopf-Hals-Chirurgie, Universität zu Köln, Medizinische Fakultät, Köln, Germany
| |
Collapse
|
8
|
Beaudry FEG, Li Z, Borgida A, Zorigtbaatar A, Wang X, Hildebrand M, Hamza O, Jang GH, Bucur R, Dodd A, Wilson J, Auer RC, Saibil S, Tsang ES, Vogel A, O’Kane GM, Gallinger S, Knox JJ, Notta F, Sapisochin G, Grant RC. Molecular Tumor Board-Guided Targeted Treatments for Biliary Tract Cancers in a Publicly Funded Healthcare System. Curr Oncol 2025; 32:80. [PMID: 39996880 PMCID: PMC11854319 DOI: 10.3390/curroncol32020080] [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: 12/23/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Comprehensive molecular profiling can identify alterations in biliary tract cancer (BTC) potentially treatable with targeted therapies. However, the impact of whole-genome and transcriptome sequencing (WGTS) on therapeutic decision-making in a public healthcare system is unknown. Here, BTC patients prospectively received WGTS to inform clinical care at a large Canadian academic cancer center. We characterized the proportion of targetable alterations, the treatment recommendations generated by a molecular tumor board, targeted therapies received, patient outcomes, and the financing of these treatments. A total of 55 patients with BTC prospectively underwent WGTS to inform clinical care. Of those 55, 28 (51%, 95% CI 38-64%) harbored targetable alterations. Molecular tumor boards recommended consideration of targeted therapies for 43 (78% CI: 66-87%) of 55 cases. Among the 15 patients who progressed to second-line therapy and harbored targetable alterations, 8 received nine targeted therapies. No targeted therapies were funded through the public system, and most therapies were funded through compassionate access programs from companies. These results highlight the challenges and potential for inequities when implementing precision oncology in a publicly funded healthcare system.
Collapse
Affiliation(s)
- Felix E. G. Beaudry
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Zhihao Li
- Toronto General Hospital, Toronto, ON M5G 2C4, Canada
| | | | - Anudari Zorigtbaatar
- Toronto General Hospital, Toronto, ON M5G 2C4, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Xin Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Eliot Phillipson Clinician-Scientist Training Program, University of Toronto, Toronto, ON M5S 3H2, Canada
| | | | - Oumaima Hamza
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
| | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
| | - Roxana Bucur
- Toronto General Hospital, Toronto, ON M5G 2C4, Canada
| | - Anna Dodd
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Julie Wilson
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
| | - Rebecca C. Auer
- Ottawa Hospital Research Institute, Ottawa, ON K1Y 1J8, Canada
| | - Samuel Saibil
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Erica S. Tsang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Arndt Vogel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Toronto General Hospital, Toronto, ON M5G 2C4, Canada
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Grainne M. O’Kane
- St. Vincent’s University Hospital, School of Medicine, University College Dublin, Dublin D04 T6F4, Ireland
| | - Steven Gallinger
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
- Department of Surgery, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Jennifer J. Knox
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Gonzalo Sapisochin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Toronto General Hospital, Toronto, ON M5G 2C4, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Robert C. Grant
- Ontario Institute for Cancer Research, Toronto, ON M5G 1M1, Canada; (F.E.G.B.)
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5G 2C4, Canada
- Institute for Clinical Evaluative Sciences, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
9
|
Schmidl B, Hütten T, Pigorsch S, Stögbauer F, Hoch CC, Hussain T, Wollenberg B, Wirth M. Artificial intelligence for image recognition in diagnosing oral and oropharyngeal cancer and leukoplakia. Sci Rep 2025; 15:3625. [PMID: 39880876 PMCID: PMC11779835 DOI: 10.1038/s41598-025-85920-4] [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: 06/25/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Visual diagnosis is one of the key features of squamous cell carcinoma of the oral cavity (OSCC) and oropharynx (OPSCC), both subsets of head and neck squamous cell carcinoma (HNSCC) with a heterogeneous clinical appearance. Advancements in artificial intelligence led to Image recognition being introduced recently into large language models (LLMs) such as ChatGPT 4.0. This exploratory study, for the first time, evaluated the application of image recognition by ChatGPT to diagnose squamous cell carcinoma and leukoplakia based on clinical images, with images without any lesion as a control group. A total of 45 clinical images were analyzed, comprising 15 cases each of SCC, leukoplakia, and non-lesion images. ChatGPT 4.0 was tasked with providing the most likely diagnosis based on these images in scenario one. In scenario two the image and the clinical history were provided, whereas in scenario three only the clinical history was given. The results and the accuracy of the LLM were rated by two independent reviewers and the overall performance was evaluated using the modified Artificial Intelligence Performance Index (AIPI. In this study, ChatGPT 4.0 demonstrated the ability to correctly identify leukoplakia cases using image recognition alone, while the ability to diagnose SCC was insufficient, but improved by including the clinical history in the prompt. Providing only the clinical history resulted in a misclassification of most leukoplakia and some SCC cases. Oral cavity lesions were more likely to be diagnosed correctly. In this exploratory study of 45 images of oral lesions, ChatGPT 4.0 demonstrated a convincing performance for detecting SCC only when the clinical history was added, whereas Leukoplakia was detected solely by image recognition. ChatGPT is therefore currently insufficient for reliable OPSCC and OSCC diagnosis, but further technological advancements may pave the way for the use in the clinical setting.
Collapse
Affiliation(s)
- Benedikt Schmidl
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany.
| | - Tobias Hütten
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Steffi Pigorsch
- Department of RadioOncology, Technical University Munich, Munich, Germany
| | - Fabian Stögbauer
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Cosima C Hoch
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Timon Hussain
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| |
Collapse
|
10
|
Goudie C, Zawati MH, Knoppers BM, Laberge AM. Genomic sequencing in paediatric oncology: navigating conflicting roles and responsibilities. J Med Genet 2025; 62:138-146. [PMID: 39653388 DOI: 10.1136/jmg-2024-110410] [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: 09/27/2024] [Accepted: 11/20/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND This study explores the ethical and moral challenges faced by paediatric oncologists when they are informed of patient genomic results, particularly during molecular tumour boards (MTBs), highlighting the interplay between their clinic, research and expert roles. METHODS This was an explanatory sequential mixed-methods study using a survey distributed to paediatric oncologists in Quebec followed by optional semi-structured interviews. Oncologists' attitudes and comfort levels with six hypothetical germline DNA results identified in a patient from a clinical vignette were assessed using Likert scales. Hypothetical genetic results represented ethical challenges of extended paediatric genomic sequencing. Interviews were conducted with a subgroup of participants to gain insight and context on key survey results. RESULTS Eighty per cent (n=28) of oncologists in Quebec completed the survey; five participated in the interviews. Comfort levels of oncologists were influenced by the type of genetic result (expected, secondary, incidental finding), whether or not the oncologist was the patient's treating physician, and whether the information disclosed to the patient aligned with the information that they had received. Awareness of a genetic result was sufficient to trigger a feeling of responsibility and liability for that result. CONCLUSION Oncologists who take part in genomic sequencing initiatives and who attend MTBs have privileged access to genomic results, above what may be accessible to patients. This imbalance in knowledge contributes to moral discomfort experienced by oncologists who feel responsible for genomic information they are aware of. We propose recommendations applicable to consent processes, policies and pipelines for sharing genomic results.
Collapse
Affiliation(s)
- Catherine Goudie
- Department of Pediatrics, Division of Hematology-Oncology, Montreal Children's Hospital, McGill University Health Centre, Montreal, Qc, Canada
- Department of Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, Qc, Canada
| | - Ma'n H Zawati
- Centre of Genomics and Policy, McGill University, Montreal, Qc, Canada
| | | | - Anne-Marie Laberge
- Division of Medical Genetics, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Qc, Canada
- Department of Pediatrics, Faculty of Medicine; Department of Social and Preventive Medicine, School of Public Health, Université de Montreal, Montreal, Qc, Canada
| |
Collapse
|
11
|
Strantz C, Böhm D, Ganslandt T, Börries M, Metzger P, Pauli T, Blaumeiser A, Scheiter A, Jung IC, Christoph J, Manuilova I, Strauch K, Ustjanzew A, Reimer N, Busch H, Unberath P. Empowering personalized oncology: evolution of digital support and visualization tools for molecular tumor boards. BMC Med Inform Decis Mak 2025; 25:29. [PMID: 39819625 PMCID: PMC11736948 DOI: 10.1186/s12911-024-02821-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Molecular tumor boards (MTBs) play a pivotal role in personalized oncology, leveraging complex data sets to tailor therapy for cancer patients. The integration of digital support and visualization tools is essential in this rapidly evolving field facing fast-growing data and changing clinical processes. This study addresses the gap in understanding the evolution of software and visualization needs within MTBs and evaluates the current state of digital support. Alignment between user requirements and software development is crucial to avoid waste of resources and maintain trust. METHODS In two consecutive nationwide medical informatics projects in Germany, surveys and expert interviews were conducted as stage 1 (n = 14), stage 2 (n = 30), and stage 3 (n = 9). Surveys, via the SoSci Survey tool, covered participants' roles, working methods, and support needs. The second survey additionally addressed requirements for visualization solutions in molecular tumor boards. These aimed to understand diverse requirements for preparation, implementation, and documentation. Nine semi-structured expert interviews complemented quantitative findings through open discussion. RESULTS Using quantitative and qualitative analyses, we show that existing digital tools may improve therapy recommendations and streamline MTB case preparation, while continuous training and system improvements are needed. CONCLUSIONS Our study contributes to the field by highlighting the importance of developing user-centric, customizable software solutions that can adapt to the fast-paced environment of MTBs to advance personalized oncology. In doing so, it lays the foundation for further advances in personalized medicine in oncology and points to a shift towards more efficient, technology-driven clinical decision-making processes. This research not only enriches our understanding of the integration of digital tools into MTBs, but also signals a broader shift towards technological innovation in healthcare.
Collapse
Affiliation(s)
- Cosima Strantz
- Medical Informatics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Dominik Böhm
- Medical Center for Information and Communication Technology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Thomas Ganslandt
- Medical Informatics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Melanie Börries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Freiburg, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Pauli
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Blaumeiser
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center-University of Freiburg, Freiburg, Germany
| | - Alexander Scheiter
- Institute of Pathology, University of Regensburg, Regensburg, Germany
- Bavarian Center for Cancer Research / BZKF, Regensburg, Germany
| | - Ian-Christopher Jung
- Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Jan Christoph
- Medical Informatics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Junior Research Group (Bio-)medical Data Science, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Iryna Manuilova
- Junior Research Group (Bio-)medical Data Science, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Arsenij Ustjanzew
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Niklas Reimer
- Group for Medical Systems Biology, Lübeck Institute of Experimental Dermatology, Universität zu Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, Lübeck, Germany
- Medical Data Integration Center, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Hauke Busch
- Group for Medical Systems Biology, Lübeck Institute of Experimental Dermatology, Universität zu Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, Lübeck, Germany
| | - Philipp Unberath
- Medical Center for Information and Communication Technology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- SRH University of Applied Sciences, Fürth, Germany
| |
Collapse
|
12
|
Lebedeva A, Belova E, Kavun A, Taraskina A, Bartoletti M, Bièche I, Curigliano G, Dupain C, Rios-Hoyo A, Kamal M, Luchini C, Poyarkov S, Le Tourneau C, Veselovsky E, Mileyko V, Ivanov M. Multi-Institutional Evaluation of Interrater Agreement of Biomarker-Drug Pair Rankings Based on the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT) and Sources of Discordance. Mol Diagn Ther 2025; 29:91-101. [PMID: 39368036 DOI: 10.1007/s40291-024-00748-4] [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: 09/22/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND The widespread use of next-generation sequencing in clinical practice has contributed to the accumulation of a large number of genomic findings associated with targeted therapy; therefore, the problem of ranking the detected findings has become acute. The European Society for Medical Oncology Scale of Clinical Actionability of molecular Targets (ESCAT) system was designed by the European Society for Medical Oncology to rank biomarkers into levels of evidence that reflect their potency and clinical significance based on published clinical data. However, the ESCAT system remains imperfect, as it is based on a subjective assessment of the levels of evidence. OBJECTIVE The objective of this study was to determine whether the ranking of LOE for biomarker-drug pairs based on the ESCAT system is dependent on the human factor, and to uncover potential issues associated with the use of the framework. METHODS To evaluate the inter-rater agreement, we created a dataset of a total of 154 biomarker-drug pairs for 18 unique tumor types. We aimed to include biomarker-drug pairs that could be considered standard of care as well as less common and under investigated pairs. Fourteen precision oncology experts were invited to assign an ESCAT level of evidence for biomarker-drug pairs. Statistical analysis was carried out using Cohen's kappa and the Kolmogorov-Smirnov test. RESULTS The inter-rater agreement was low with some exceptions, and significant deviations from the consensus level of evidence were observed. For biomarker-drug associations, the deviations from the consensus were observed for more than 50% of the contributors' rankings. The most agreement between the contributors was observed for lung adenocarcinoma (p < 0.005), while the most disagreement was observed for esophageal cancer (p < 0.01) biomarker-drug pairs in our dataset. CONCLUSIONS This study demonstrates noteworthy discordances between the precision oncology experts and may provide the directions for future developments in modifying the ESCAT framework and the overall applicability of the results of genomic profiling into clinical practice.
Collapse
Affiliation(s)
- Alexandra Lebedeva
- OncoAtlas LLC, Leninsky Ave, 1/4A, Moscow, 119049, Russia.
- Laboratory of Applied Genomic Technologies, Institute of Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Ekaterina Belova
- OncoAtlas LLC, Leninsky Ave, 1/4A, Moscow, 119049, Russia
- Laboratory of Applied Genomic Technologies, Institute of Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | | | | | - Michele Bartoletti
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Ivan Bièche
- Department of Genetics, Institut Curie, Paris, France
- INSERM U1016, Faculty of Pharmaceutical and Biological Sciences, Université Paris Cité, Paris, France
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - Célia Dupain
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | | | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- Institut Hospitalo Universitaire PRISM, National PRecISion Medicine Center in Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | | | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- INSERM U900 Research unit, Institut Curie, Saint-Cloud, France
- Faculty of Medicine, Paris-Saclay University, Paris, France
| | | | - Vladislav Mileyko
- OncoAtlas LLC, Leninsky Ave, 1/4A, Moscow, 119049, Russia
- Laboratory of Applied Genomic Technologies, Institute of Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maxim Ivanov
- OncoAtlas LLC, Leninsky Ave, 1/4A, Moscow, 119049, Russia
- Laboratory of Applied Genomic Technologies, Institute of Personalized Oncology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
13
|
Lutz S, D'Angelo A, Hammerl S, Schmutz M, Claus R, Fischer NM, Kramer F, Hammoud Z. Unveiling the Digital Evolution of Molecular Tumor Boards. Target Oncol 2025; 20:27-43. [PMID: 39609355 PMCID: PMC11762447 DOI: 10.1007/s11523-024-01109-1] [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] [Accepted: 10/08/2024] [Indexed: 11/30/2024]
Abstract
Molecular tumor boards (MTB) are interdisciplinary conferences involving various experts discussing patients with advanced tumors, to derive individualized treatment suggestions based on molecular variants. These discussions involve using heterogeneous internal data, such as patient clinical data, but also external resources such as knowledge databases for annotations and search for relevant clinical studies. This imposes a certain level of complexity that requires huge effort to homogenize the data and use it in a speedy manner to reach the needed treatment. For this purpose, most institutions involving an MTB are heading toward automation and digitalization of the process, hence reducing manual work requiring human intervention and subsequently time in deriving personalized treatment suggestions. The tools are also used to better visualize the patient's data, which allows a refined overview for the board members. In this paper, we present the results of our thorough literature research about MTBs, their process, the most common knowledge bases, and tools used to support this decision-making process.
Collapse
Affiliation(s)
- Sebastian Lutz
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany.
| | - Alicia D'Angelo
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Sonja Hammerl
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Maximilian Schmutz
- Institute of Digital Medicine (IDM), Medical Faculty, University of Augsburg, Augsburg, Germany
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Rainer Claus
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nina M Fischer
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Frank Kramer
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Zaynab Hammoud
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| |
Collapse
|
14
|
Barducci MC, Tozzi VD, Pelizzari G, Aprile G, Grossi F, Pinto C, Fasola G. Precision oncology implementation in a regional-based health care system: A professional consensus to define the pathway. J Cancer Policy 2024; 42:100515. [PMID: 39532218 DOI: 10.1016/j.jcpo.2024.100515] [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: 07/03/2024] [Revised: 10/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Precision Oncology requires deep changes in organizational settings but little evidence has been identified about the best strategy to guarantee the delivery of this innovation to patients. In the Italian health care system, high heterogeneity could jeopardize equal access opportunity for patients. Following a consensus method, we aim to define shared solutions to address these issues in clinical practice. METHOD A Delphi RAND method was chosen to record the consensus among involved health care professionals in the Italian region of Friuli Venezia Giulia. The item generation phase was conducted following a bottom-up approach. RESULTS Ten statements were defined on the main topics that emerged from the direct observation of the current practice, focusing on Molecular Tumor Board organization, massive parallel sequencing technology application, laboratory report content and informed consensus submission. All the statements reached a strong consensus and have been shared with the health care government authorities of our region. CONCLUSIONS The direct observation of the current practice in different health care authorities allowed to define ten statements as solution proposals to solve the identified complexities. This methodological approach could be applied in different organizational models but aiming to achieve a homogeneous clinical outcome for patients.
Collapse
Affiliation(s)
- Maria Carla Barducci
- Santa Maria della Misericordia University Hospital, ASUFC, Piazzale Santa Maria della Misericordia 15, Udine 33100, Italy.
| | - Valeria Domenica Tozzi
- SDA Bocconi School of Management, Centre for Research on Health and Social Care Management (CERGAS), Bocconi University, Milan 20136, Italy
| | - Giacomo Pelizzari
- Santa Maria della Misericordia University Hospital, ASUFC, Piazzale Santa Maria della Misericordia 15, Udine 33100, Italy
| | - Giuseppe Aprile
- San Bortolo Hospital of Vicenza, Azienda ULSS8 Berica, Viale Ferdinando Rodolfi 37, Vicenza 36100, Italy
| | - Francesco Grossi
- University of Insubria, ASST dei Sette Laghi, Via Ravasi 2, Varese 21100, Italy
| | - Carmine Pinto
- Medical Oncology, Comprehensive Cancer Centre, AUSL-IRCCS di Reggio Emilia, Via Giovanni Amendola, 2, Reggio Emilia 42122, Italy
| | - Gianpiero Fasola
- Santa Maria della Misericordia University Hospital, ASUFC, Piazzale Santa Maria della Misericordia 15, Udine 33100, Italy
| |
Collapse
|
15
|
Niarakis A, Laubenbacher R, An G, Ilan Y, Fisher J, Flobak Å, Reiche K, Rodríguez Martínez M, Geris L, Ladeira L, Veschini L, Blinov ML, Messina F, Fonseca LL, Ferreira S, Montagud A, Noël V, Marku M, Tsirvouli E, Torres MM, Harris LA, Sego TJ, Cockrell C, Shick AE, Balci H, Salazar A, Rian K, Hemedan AA, Esteban-Medina M, Staumont B, Hernandez-Vargas E, Martis B S, Madrid-Valiente A, Karampelesis P, Sordo Vieira L, Harlapur P, Kulesza A, Nikaein N, Garira W, Malik Sheriff RS, Thakar J, Tran VDT, Carbonell-Caballero J, Safaei S, Valencia A, Zinovyev A, Glazier JA. Immune digital twins for complex human pathologies: applications, limitations, and challenges. NPJ Syst Biol Appl 2024; 10:141. [PMID: 39616158 PMCID: PMC11608242 DOI: 10.1038/s41540-024-00450-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/27/2024] [Indexed: 12/06/2024] Open
Abstract
Digital twins represent a key technology for precision health. Medical digital twins consist of computational models that represent the health state of individual patients over time, enabling optimal therapeutics and forecasting patient prognosis. Many health conditions involve the immune system, so it is crucial to include its key features when designing medical digital twins. The immune response is complex and varies across diseases and patients, and its modelling requires the collective expertise of the clinical, immunology, and computational modelling communities. This review outlines the initial progress on immune digital twins and the various initiatives to facilitate communication between interdisciplinary communities. We also outline the crucial aspects of an immune digital twin design and the prerequisites for its implementation in the clinic. We propose some initial use cases that could serve as "proof of concept" regarding the utility of immune digital technology, focusing on diseases with a very different immune response across spatial and temporal scales (minutes, days, months, years). Lastly, we discuss the use of digital twins in drug discovery and point out emerging challenges that the scientific community needs to collectively overcome to make immune digital twins a reality.
Collapse
Affiliation(s)
- Anna Niarakis
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, UPS, CNRS, Toulouse, France.
- Lifeware Group, Inria, Saclay-île de France, Palaiseau, France.
| | | | - Gary An
- Department of Surgery, University of Vermont Larner College of Medicine, Vermont, USA
| | - Yaron Ilan
- Faculty of Medicine Hebrew University, Hadassah Medical Center, Jerusalem, Israel
| | - Jasmin Fisher
- UCL Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6BT, UK
| | - Åsmund Flobak
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- The Cancer Clinic, St Olav's University Hospital, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Kristin Reiche
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Institute of Clinical Immunology, Medical Faculty, University Hospital, University of Leipzig, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden/Leipzig, Germany
| | - María Rodríguez Martínez
- Department of Biomedical Informatics & Data Science, Yale School of Medicine, New Haven, CT, USA
| | - Liesbet Geris
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Biomechanics Research Unit, GIGA Molecular and Computational Biology, University of Liège, Liège, Belgium
| | - Luiz Ladeira
- Biomechanics Research Unit, GIGA Molecular and Computational Biology, University of Liège, Liège, Belgium
| | - Lorenzo Veschini
- Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, London, UK
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, 47408, USA
| | - Michael L Blinov
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, 06030, USA
| | - Francesco Messina
- Department of Epidemiology, Preclinical Research and Advanced Diagnostic, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - I.R.C.C.S., Rome, Italy
| | - Luis L Fonseca
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Sandra Ferreira
- Mathematics Department and Center of Mathematics, University of Beira Interior, Covilhã, Portugal
| | - Arnau Montagud
- Barcelona Supercomputing Center (BSC), Barcelone, Spain
- Institute for Integrative Systems Biology (I2SysBio), CSIC-UV, Valencia, Spain
| | - Vincent Noël
- Institut Curie, Université PSL, F-75005, Paris, France
- INSERM, U900, F-75005, Paris, France
- Mines ParisTech, Université PSL, F-75005, Paris, France
| | - Malvina Marku
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Eirini Tsirvouli
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marcella M Torres
- Department of Mathematics and Statistics, University of Richmond, Richmond, VA, USA
| | - Leonard A Harris
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
- Interdisciplinary Graduate Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR, USA
- Cancer Biology Program, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - T J Sego
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Chase Cockrell
- Department of Surgery, University of Vermont Larner College of Medicine, Vermont, USA
| | - Amanda E Shick
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA
| | - Hasan Balci
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - Albin Salazar
- INRIA Paris/CNRS/École Normale Supérieure/PSL Research University, Paris, France
| | - Kinza Rian
- Andalusian Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
| | - Ahmed Abdelmonem Hemedan
- Bioinformatics Core Unit, Luxembourg Centre of Systems Biomedicine LCSB, Luxembourg University, Esch-sur-Alzette, Luxembourg
| | - Marina Esteban-Medina
- Andalusian Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
| | - Bernard Staumont
- Biomechanics Research Unit, GIGA Molecular and Computational Biology, University of Liège, Liège, Belgium
| | - Esteban Hernandez-Vargas
- Department of Mathematics and Statistical Science, University of Idaho, Moscow, ID, 83844-1103, USA
| | | | | | | | | | - Pradyumna Harlapur
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | | | - Niloofar Nikaein
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, SE-70182, Örebro, Sweden
- X-HiDE - Exploring Inflammation in Health and Disease Consortium, Örebro University, Örebro, Sweden
| | - Winston Garira
- Multiscale Mathematical Modelling of Living Systems program (M3-LSP), Kimberley, South Africa
- Department of Mathematical Sciences, Sol Plaatje University, Kimberley, South Africa
- Private Bag X5008, Kimberley, 8300, South Africa
| | - Rahuman S Malik Sheriff
- European Bioinformatics Institute, European Molecular Biology Laboratory (EMBL-EBI), Hinxton, Cambridge, UK
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Juilee Thakar
- Department of Microbiology & Immunology and Department of Biostatistics & Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Van Du T Tran
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Soroush Safaei
- Institute of Biomedical Engineering and Technology, Ghent University, Gent, Belgium
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Barcelone, Spain
- ICREA, 23 Passeig Lluís Companys, 08010, Barcelona, Spain
| | | | - James A Glazier
- Biocomplexity Institute and Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, 47408, USA
| |
Collapse
|
16
|
Schmidl B, Hütten T, Pigorsch S, Stögbauer F, Hoch CC, Hussain T, Wollenberg B, Wirth M. Assessing the use of the novel tool Claude 3 in comparison to ChatGPT 4.0 as an artificial intelligence tool in the diagnosis and therapy of primary head and neck cancer cases. Eur Arch Otorhinolaryngol 2024; 281:6099-6109. [PMID: 39112556 PMCID: PMC11512878 DOI: 10.1007/s00405-024-08828-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/03/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) is a complex malignancy that requires a multidisciplinary tumor board approach for individual treatment planning. In recent years, artificial intelligence tools have emerged to assist healthcare professionals in making informed treatment decisions. This study investigates the application of the newly published LLM Claude 3 Opus compared to the currently most advanced LLM ChatGPT 4.0 for the diagnosis and therapy planning of primary HNSCC. The results were compared to that of a conventional multidisciplinary tumor board; (2) Materials and Methods: We conducted a study in March 2024 on 50 consecutive primary head and neck cancer cases. The diagnostics and MDT recommendations were compared to the Claude 3 Opus and ChatGPT 4.0 recommendations for each patient and rated by two independent reviewers for the following parameters: clinical recommendation, explanation, and summarization in addition to the Artificial Intelligence Performance Instrument (AIPI); (3) Results: In this study, Claude 3 achieved better scores for the diagnostic workup of patients than ChatGPT 4.0 and provided treatment recommendations involving surgery, chemotherapy, and radiation therapy. In terms of clinical recommendations, explanation and summarization Claude 3 scored similar to ChatGPT 4.0, listing treatment recommendations which were congruent with the MDT, but failed to cite the source of the information; (4) Conclusion: This study is the first analysis of Claude 3 for primary head and neck cancer cases and demonstrates a superior performance in the diagnosis of HNSCC than ChatGPT 4.0 and similar results for therapy recommendations. This marks the advent of a newly launched advanced AI model that may be superior to ChatGPT 4.0 for the assessment of primary head and neck cancer cases and may assist in the clinical diagnostic and MDT setting.
Collapse
Affiliation(s)
- Benedikt Schmidl
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany.
| | - Tobias Hütten
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Steffi Pigorsch
- Department of RadioOncology, Technical University Munich, Munich, Germany
| | - Fabian Stögbauer
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Cosima C Hoch
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Timon Hussain
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| |
Collapse
|
17
|
Vitale A, Mastrantoni L, Russo J, Giacomini F, Giannarelli D, Duranti S, Vita E, Nero C, D'Argento E, Pasciuto T, Giacò L, Di Salvatore M, Panfili A, Stefani A, Cancellieri A, Lococo F, De Paolis E, Livi V, Daniele G, Trisolini R, Minucci A, Margaritora S, Lorusso D, Normanno N, Scambia G, Tortora G, Bria E. Impact of Comprehensive Genome Profiling on the Management of Advanced Non-Small Cell Lung Cancer: Preliminary Results From the Lung Cancer Cohort of the FPG500 Program. JCO Precis Oncol 2024; 8:e2400297. [PMID: 39374480 DOI: 10.1200/po.24.00297] [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: 05/07/2024] [Revised: 07/02/2024] [Accepted: 08/16/2024] [Indexed: 10/09/2024] Open
Abstract
PURPOSE The clinical and research FPG500 program (ClinicalTrials.gov identifier: NCT06020625) is currently ongoing at the Fondazione Policlinico Universitario Agostino Gemelli IRCCS to tailor matched targeted therapies (MTTs) according to biomarkers predictive of response identified by comprehensive genome profiling (CGP). MATERIALS AND METHODS The non-small cell lung cancer (NSCLC) cohort results from the FPG500 program are outlined. CGP was performed by TruSight Oncology 500 High Throughput (TSO500HT) assay or Oncomine Focus Assay plus Archer's FusionPlex Lung Panel according to tumor cell content and DNA/RNA quantity. Relevant issues for Molecular Tumor Board (MTB) evaluation included uncommon genomic findings, evaluation for off-label therapies, uncertain result confirmation, and variants of suspect germline origin requiring genetic counseling. Progression-free survival (PFS) and overall survival (OS) for the enrolled patients were assessed using Kaplan-Meier analysis. RESULTS In 2022, 283 patients with NSCLC were considered for sequencing, with 93% meeting eligibility criteria. TSO500HT sequencing was conducted in 76% of patients. Follow-up data were obtained for 187 patients, among whom 81% received treatment. Potential driver alterations were identified in 59% of patients, with 41% receiving MTT: 25% were prescribed approved MTTs, whereas 16% gained access to experimental drugs post-MTB evaluation; of note, 18% did not receive any MTT because the regimen was not yet reimbursed in our country. Median PFS and OS varied among treatment groups, with standard chemotherapy/immunotherapy at 7.7 and 10.7 months, approved tyrosine kinase inhibitors at 18.8 and 23.9 months, and MTT post-MTB discussion at 14 and 23.4 months, respectively. CONCLUSION The early data of the FPG program (NSCLC cohort) support the implementation of CGP and MTB in clinical practice to grant access to patients harboring actionable molecular alterations to the most effective and individualized available treatment options, thus improving their survival outcomes.
Collapse
Affiliation(s)
- Antonio Vitale
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Mastrantoni
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jacopo Russo
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Flavia Giacomini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Diana Giannarelli
- Biostatistical Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Simona Duranti
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Emanuele Vita
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Camilla Nero
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Women, Children and Public Health Sciences Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ettore D'Argento
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Tina Pasciuto
- Data Collection Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luciano Giacò
- Bioinformatics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Mariantonietta Di Salvatore
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Arianna Panfili
- Scientific Directorate, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Alessio Stefani
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandra Cancellieri
- Pathology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Filippo Lococo
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Thoracic Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Elisa De Paolis
- Laboratory of Clinical Molecular and Personalized Diagnostics, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Vanina Livi
- Interventional Pulmonology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gennaro Daniele
- Phase 1 Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Rocco Trisolini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Interventional Pulmonology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angelo Minucci
- Laboratory of Clinical Molecular and Personalized Diagnostics, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Stefano Margaritora
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Thoracic Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Domenica Lorusso
- Gynecologic Oncology Unit, Humanitas San Pio X, Humanitas University, Milan, Italy
| | - Nicola Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori," Meldola, Italy
| | - Giovanni Scambia
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Women, Children and Public Health Sciences Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Ospedale Isola Tiberina-Gemelli Isola, Rome, Italy
| |
Collapse
|
18
|
Rapoport N, Pavelchek C, Michelson AP, Shew MA. Artificial Intelligence in Otology and Neurotology. Otolaryngol Clin North Am 2024; 57:791-802. [PMID: 38871535 DOI: 10.1016/j.otc.2024.04.009] [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] [Indexed: 06/15/2024]
Abstract
Clinical applications of artificial intelligence (AI) have grown exponentially with increasing computational power and Big Data. Data rich fields such as Otology and Neurotology are still in the infancy of harnessing the power of AI but are increasingly involved in training and developing ways to incorporate AI into patient care. Current studies involving AI are focused on accessible datasets; health care wearables, tabular data from electronic medical records, electrophysiologic measurements, imaging, and "omics" provide huge amounts of data to utilize. Health care wearables, such as hearing aids and cochlear implants, are a ripe environment for AI implementation.
Collapse
Affiliation(s)
- Nicholas Rapoport
- Washington University School of Medicine in St. Louis, 660 South Euclid Avenue, PO Box 8115, St Louis, MO 63110, USA
| | - Cole Pavelchek
- Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Andrew P Michelson
- Department of Pulmonary Critical Care, Washington University School of Medicine, 660 South Euclid Avenue, PO Box 8052-43-14, St Louis, MO 63110, USA; Institute for Informatics, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew A Shew
- Otology & Neurotology, Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine in St. Louis, 660 South Euclid Avenue, PO Box 8115, St Louis, MO 63110, USA.
| |
Collapse
|
19
|
Tommasi S, Maurmo L, Rizzo A, Carella C, Ranieri G, De Summa S, Mannavola F, Chiurì VE, Guida M, Nisi C, Montrone M, Giotta F, Patruno M, Lacalamita R, Pilato B, Zito FA, Fucci L, Coppola CA, Ditonno P, Nardulli P, Quaresmini D, Strippoli S. The molecular tumor board as a step in cancer patient management: a southern Italian experience. Front Med (Lausanne) 2024; 11:1432628. [PMID: 39323465 PMCID: PMC11422073 DOI: 10.3389/fmed.2024.1432628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction The management of cancer patients follows a Diagnostic Therapeutic and Care Pathway (PDTA) approach, aimed at achieving the optimal balance between care and quality of life. To support this process, precision medicine and innovative technologies [e.g., next-generation sequencing (NGS)] allow rapid identification of genetic-molecular alterations useful for the design of PDTA-approved therapies. If the standard approach proves inadequate, the Molecular Tumor Board (MTB), a group comprising specialists from diverse disciplines, can step in to evaluate a broader molecular profile, proposing potential therapies beyond evidence levels I-II or considering enrolment in clinical trials. Our aim is to analyze the role of the MTB in the entire management of patients in our institute and its impact on the strategy of personalized medicine, particularly when all approved treatments have failed. Materials and methods In alignment with European and national guidelines, a panel of clinicians and preclinical specialists from our institution was defined as the MTB core team. We designed and approved a procedure for the operation of this multidisciplinary group, which is the only one operating in the Puglia region. Results and discussion In 29 months (2021-2023), we discussed and analyzed 93 patients. A total of 44% presented pathogenic alterations, of which 40.4% were potentially actionable. Only 11 patients were proposed for enrollment in clinical trials, treatment with off-label drugs, or AIFA (the Italian pharmaceutical agency for drugs)-5% funding. Our process indicators, time to analysis, and number of patient cases discussed are in line with the median data of other European institutions. Such findings underscore both the importance and usefulness of the integration of an MTB process into the care of oncology patients.
Collapse
Affiliation(s)
- Stefania Tommasi
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Leonarda Maurmo
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Alessandro Rizzo
- Unità Operativa di Oncologia Medica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Claudia Carella
- Unità Operativa di Oncologia Medica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Girolamo Ranieri
- Unità di Oncologia Interventistica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Simona De Summa
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Francesco Mannavola
- Unità di Oncologia Medica, Azienda Ospedaliera Policlinico Consorziale di Bari, Bari, Italy
| | | | - Michele Guida
- Unità Operativa di Oncologia Medica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Claudia Nisi
- Reparto di Oncologia, Ospedale San Giuseppe Moscati Taranto, Taranto, Italy
| | - Michele Montrone
- Unità Operativa di Oncologia Medica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Francesco Giotta
- Unità Operativa di Oncologia Medica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Margherita Patruno
- Centro Studi Tumori eredo-familiari, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Rosanna Lacalamita
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Brunella Pilato
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Francesco Alfredo Zito
- Unità Operativa di Anatomia Patologica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Livia Fucci
- Unità Operativa di Anatomia Patologica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Claudio Antonio Coppola
- Unità di Diagnostica Molecolare e Farmacogenetica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Paolo Ditonno
- Unità Operativa di Ematologia, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Patrizia Nardulli
- Unità Operativa Farmacia e U.M.A.C.A., IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Davide Quaresmini
- Unità Operativa di Oncologia Medica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| | - Sabino Strippoli
- Unità Operativa di Oncologia Medica, IRCCS Istituto Tumori Giovanni Paolo II Bari, Bari, Italy
| |
Collapse
|
20
|
Schmidl B, Hütten T, Pigorsch S, Stögbauer F, Hoch CC, Hussain T, Wollenberg B, Wirth M. Assessing the role of advanced artificial intelligence as a tool in multidisciplinary tumor board decision-making for recurrent/metastatic head and neck cancer cases - the first study on ChatGPT 4o and a comparison to ChatGPT 4.0. Front Oncol 2024; 14:1455413. [PMID: 39301542 PMCID: PMC11410764 DOI: 10.3389/fonc.2024.1455413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Background Recurrent and metastatic head and neck squamous cell carcinoma (HNSCC) is characterized by a complex therapeutic management that needs to be discussed in multidisciplinary tumor boards (MDT). While artificial intelligence (AI) improved significantly to assist healthcare professionals in making informed treatment decisions for primary cases, an application in the even more complex recurrent/metastatic setting has not been evaluated yet. This study also represents the first evaluation of the recently published LLM ChatGPT 4o, compared to ChatGPT 4.0 for providing therapy recommendations. Methods The therapy recommendations for 100 HNSCC cases generated by each LLM, 50 cases of recurrence and 50 cases of distant metastasis were evaluated by two independent reviewers. The primary outcome measured was the quality of the therapy recommendations measured by the following parameters: clinical recommendation, explanation, and summarization. Results In this study, ChatGPT 4o and 4.0 provided mostly general answers for surgery, palliative care, or systemic therapy. ChatGPT 4o proved to be 48.5% faster than ChatGPT 4.0. For clinical recommendation, explanation, and summarization both LLMs obtained high scores in terms of performance of therapy recommendations, with no significant differences between both LLMs, but demonstrated to be mostly an assisting tool, requiring validation by an experienced clinician due to a lack of transparency and sometimes recommending treatment modalities that are not part of the current treatment guidelines. Conclusion This research demonstrates that ChatGPT 4o and 4.0 share a similar performance, while ChatGPT 4o is significantly faster. Since the current versions cannot tailor therapy recommendations, and sometimes recommend incorrect treatment options and lack information on the source material, advanced AI models at the moment can merely assist in the MDT setting for recurrent/metastatic HNSCC.
Collapse
Affiliation(s)
- Benedikt Schmidl
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Tobias Hütten
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Steffi Pigorsch
- Department of RadioOncology, Technical University Munich, Munich, Germany
| | - Fabian Stögbauer
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Cosima C Hoch
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Timon Hussain
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| |
Collapse
|
21
|
Fabi A, Cortesi L, Duranti S, Cordisco EL, Di Leone A, Terribile D, Paris I, de Belvis AG, Orlandi A, Marazzi F, Muratore M, Garganese G, Fuso P, Paoletti F, Dell'Aquila R, Minucci A, Scambia G, Franceschini G, Masetti R, Genuardi M. Multigenic panels in breast cancer: Clinical utility and management of patients with pathogenic variants other than BRCA1/2. Crit Rev Oncol Hematol 2024; 201:104431. [PMID: 38977141 DOI: 10.1016/j.critrevonc.2024.104431] [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: 03/26/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Multigene panels can analyze high and moderate/intermediate penetrance genes that predispose to breast cancer (BC), providing an opportunity to identify at-risk individuals within affected families. However, considering the complexity of different pathogenic variants and correlated clinical manifestations, a multidisciplinary team is needed to effectively manage BC. A classification of pathogenic variants included in multigene panels was presented in this narrative review to evaluate their clinical utility in BC. Clinical management was discussed for each category and focused on BC, including available evidence regarding the multidisciplinary and integrated management of patients with BC. The integration of both genetic testing and counseling is required for customized decisions in therapeutic strategies and preventative initiatives, as well as for a defined multidisciplinary approach, considering the continuous evolution of guidelines and research in the field.
Collapse
Affiliation(s)
- Alessandra Fabi
- Precision Medicine Unit in Senology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Laura Cortesi
- Department of Oncology and Haematology, Modena Hospital University, Modena Italy (Cortesi)
| | - Simona Duranti
- Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Emanuela Lucci Cordisco
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Genetics Unit, Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alba Di Leone
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Terribile
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ida Paris
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Giulio de Belvis
- Value Lab, Faculty of Economics, Università Cattolica del Sacro Cuore, Rome, Italy; Critical Pathways and Outcomes Evaluation Unit, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Armando Orlandi
- Unit of Oncology, Comprehensive Cancer Centre, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabio Marazzi
- UOC Oncological Radiotherapy, Department of Diagnostic Imaging, Radiation Oncology and Haematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Margherita Muratore
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori"
| | - Giorgia Garganese
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Section of Obstetrics and Gynecology, Department of Woman and Child Health and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paola Fuso
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Filippo Paoletti
- Critical Pathways and Outcomes Evaluation Unit, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Rossella Dell'Aquila
- Critical Pathways and Outcomes Evaluation Unit, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Angelo Minucci
- Genomics Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- Division of Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Gianluca Franceschini
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Riccardo Masetti
- Breast Unit, Department of Woman and Child's Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Maurizio Genuardi
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Medical Genetics Unit, Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
22
|
Bartels M, Chibaudel B, Dienstmann R, Lehtiö J, Piccolo A, Michielin O, O’Kane G, Pruneri G. Evolving and Improving the Sustainability of Molecular Tumor Boards: The Value and Challenges. Cancers (Basel) 2024; 16:2888. [PMID: 39199658 PMCID: PMC11353190 DOI: 10.3390/cancers16162888] [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: 07/09/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
The increasing volume of information for cancer care, and the evolution of molecularly guided therapies, have increased the need for molecular tumor boards (MTBs), which can integrate such data into personalized treatment plans to improve patient outcomes. However, recommendations for improving the sustainability of MTBs are lacking. A diverse committee of MTB experts was assembled (February-March 2023), with extensive experience in sustainability in healthcare ecosystems. The aim was to identify MTB-related hurdles throughout the patient journey and develop a general framework for MTBs to operate on larger scales locally, nationally, and internationally. The committee identified ten key pillars for sustainable and scalable MTBs, including technical solutions for data integration and visualization, interoperability, learning loops, clinical trial access, legal considerations, criteria for patient testing, decision standardization, making MTBs official bodies for treatment decisions, local leaders, and international networks. The need for scalable frameworks at academic and community levels was recognized, along with integrating MTBs into national health systems to enhance sustainability and ensure optimal treatment decisions. Irrespective of the health ecosystem, the sustainability and scalability of MTBs are essential. Our framework provides guidelines to address this and to help MTBs evolve towards integrated, essential components of the oncology healthcare system.
Collapse
Affiliation(s)
- Marius Bartels
- Department of Medical Oncology, Praxis für Onkologie Mönchengladbach, 41066 Mönchengladbach, Germany
| | - Benoist Chibaudel
- Department of Medical Oncology, Hôpital Franco-Britannique—Fondation Cognacq-Jay, Cancérologie ParisOuest, 92300 Levallois-Perret, France
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
- University of Vic-Central University of Catalonia, 08500 Vic, Spain
- Oncoclínicas, São Paulo 04543-906, Brazil
| | - Janne Lehtiö
- Science for Life Laboratory and Karolinska Comprehensive Cancer Center, Department of Oncology and Pathology, Karolinska Institutet, 17177 Solna, Sweden
| | - Alberta Piccolo
- Pathology Unit 2, Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Olivier Michielin
- Department of Oncology and the Precision Oncology Service, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Grainne O’Kane
- Trinity St. James’s Cancer Institute, St. James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Giancarlo Pruneri
- Pathology Unit 2, Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
23
|
Louie BH, Kato S, Lim JS, Kim KH, Lim HJ, Okamura R, Lee S, Kim L, Sicklick JK, Lippman SM, Kurzrock R. Molecular Tumor Board for Unicorns: Outcomes for rare and ultra-rare cancers using an N-of-One personalized treatment strategy. iScience 2024; 27:110465. [PMID: 39148716 PMCID: PMC11324991 DOI: 10.1016/j.isci.2024.110465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 05/06/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Treatment of rare/ultra-rare tumors is an unmet need due to a lack of standardized therapies and clinical trials. We developed the Molecular Tumor Board (MTB), a multidisciplinary team that integrates molecular profiling to generate personalized, N-of-One treatments for advanced cancers. This study evaluates 112 patients with rare/ultra-rare tumors who presented to the MTB and were evaluable for clinical therapeutic outcome. Overall, 46/112 patients (41%) received a treatment regimen with a high degree of matching between tumor molecular alterations and drugs given (reflected by a high Matching Score (≥50%)). Patients with a high versus low Matching Score experienced significantly longer progression-free survival (p = 0.005) and overall survival (p = 0.047), and higher rates of clinical benefit (stable disease ≥6 months, partial response, or complete response) (54% vs. 32% p = 0.027). The MTB facilitated personalized N-of-One matching of drugs to tumor molecular alterations, which was associated with improved clinical outcomes in patients with rare/ultra-rare cancers.
Collapse
Affiliation(s)
- Bryan H Louie
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jordan S Lim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Ki Hwan Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Hyo Jeong Lim
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Ryosuke Okamura
- Department of Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Lisa Kim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Division of Surgical Oncology, Department of Surgery, and Department of Pharmacology, UC San Diego Health Sciences, San Diego, CA, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, Paris, France
- Medical College of Wisconsin Cancer Center and Genomic Sciences and Precision Medicine Center, Milwaukee, WI, USA
- University of Nebraska, Omaha, NE, USA
| |
Collapse
|
24
|
Pallocca M, Betti M, Baldinelli S, Palombo R, Bucci G, Mazzarella L, Tonon G, Ciliberto G. Clinical bioinformatics desiderata for molecular tumor boards. Brief Bioinform 2024; 25:bbae447. [PMID: 39297878 PMCID: PMC11411775 DOI: 10.1093/bib/bbae447] [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: 12/18/2023] [Revised: 06/28/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Clinical Bioinformatics is a knowledge framework required to interpret data of medical interest via computational methods. This area became of dramatic importance in precision oncology, fueled by cancer genomic profiling: most definitions of Molecular Tumor Boards require the presence of bioinformaticians. However, all available literature remained rather vague on what are the specific needs in terms of digital tools and expertise to tackle and interpret genomics data to assign novel targeted or biomarker-driven targeted therapies to cancer patients. To fill this gap, in this article, we present a catalog of software families and human skills required for the tumor board bioinformatician, with specific examples of real-world applications associated with each element presented.
Collapse
Affiliation(s)
- Matteo Pallocca
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Martina Betti
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | - Sara Baldinelli
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | - Ramona Palombo
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Gabriele Bucci
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milan, Italy
| | - Luca Mazzarella
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IRCCS IEO - European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
- Department of Experimental Oncology, IRCCS IEO - European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, and Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milan, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| |
Collapse
|
25
|
Pastò B, Buzzatti G, Schettino C, Malapelle U, Bergamini A, De Angelis C, Musacchio L, Dieci MV, Kuhn E, Lambertini M, Passarelli A, Toss A, Farolfi A, Roncato R, Capoluongo E, Vida R, Pignata S, Callari M, Baldassarre G, Bartoletti M, Gerratana L, Puglisi F. Unlocking the potential of Molecular Tumor Boards: from cutting-edge data interpretation to innovative clinical pathways. Crit Rev Oncol Hematol 2024; 199:104379. [PMID: 38718940 DOI: 10.1016/j.critrevonc.2024.104379] [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: 01/30/2024] [Revised: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
The emerging era of precision medicine is characterized by an increasing availability of targeted anticancer therapies and by the parallel development of techniques to obtain more refined molecular data, whose interpretation may not always be straightforward. Molecular tumor boards gather various professional figures, in order to leverage the analysis of molecular data and provide prognostic and predictive insights for clinicians. In addition to healthcare development, they could also become a tool to promote knowledge and research spreading. A growing body of evidence on the application of molecular tumor boards to clinical practice is forming and positive signals are emerging, although a certain degree of heterogeneity exists. This work analyzes molecular tumor boards' potential workflows, figures involved, data sources, sample matrices and eligible patients, as well as available evidence and learning examples. The emerging concept of multi-institutional, disease-specific molecular tumor boards is also considered by presenting two ongoing nationwide experiences.
Collapse
Affiliation(s)
- Brenno Pastò
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Giulia Buzzatti
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy
| | - Clorinda Schettino
- Clinical Trials Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli 80131, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Napoli 80131, Italy
| | - Alice Bergamini
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milano 20132, Italy; Unit of Obstetrics and Gynaecology, IRCCS San Raffaele Scientific Institute, Milano 20132, Italy
| | - Carmine De Angelis
- Oncology Unit - Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli 80131, Italy
| | - Lucia Musacchio
- Department of Women and Child Health, Division of Gynaecologic Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Roma 00168, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova 35122, Italy; Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova 35128, Italy
| | - Elisabetta Kuhn
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milano 20122, Italy; Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova 16132, Italy
| | - Anna Passarelli
- Department of Urology and Gynaecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli 80131, Italy
| | - Angela Toss
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena 41124, Italy; Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena 41124, Italy
| | - Alberto Farolfi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola 47014, Italy
| | - Rossana Roncato
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Ettore Capoluongo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Napoli 80131, Italy; Clinical Pathology Unit, Azienda Ospedaliera San Giovanni Addolorata, Roma 00184, Italy
| | - Riccardo Vida
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Sandro Pignata
- Department of Urology and Gynaecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli 80131, Italy
| | | | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Michele Bartoletti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Lorenzo Gerratana
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy.
| | - Fabio Puglisi
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| |
Collapse
|
26
|
Mansour N, Heinrich K, Zhang D, Winkelmann M, Ingenerf M, Gold L, Klambauer K, Rudelius M, Klauschen F, von Bergwelt-Baildon M, Ricke J, Heinemann V, Westphalen CB, Kunz WG. Patient eligibility for trials with imaging response assessment at the time of molecular tumor board presentation. Cancer Imaging 2024; 24:70. [PMID: 38849902 PMCID: PMC11157753 DOI: 10.1186/s40644-024-00708-5] [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: 12/01/2023] [Accepted: 05/11/2024] [Indexed: 06/09/2024] Open
Abstract
PURPOSE To assess the eligibility of patients with advanced or recurrent solid malignancies presented to a molecular tumor board (MTB) at a large precision oncology center for inclusion in trials with the endpoints objective response rate (ORR) or duration of response (DOR) based on Response Evaluation Criteria in Solid Tumors (RECIST version 1.1). METHODS Prospective patients with available imaging at the time of presentation in the MTB were included. Imaging data was reviewed for objectifiable measurable disease (MD) according to RECIST v1.1. Additionally, we evaluated the patients with MD for representativeness of the identified measurable lesion(s) in relation to the overall tumor burden. RESULTS 262 patients with different solid malignancies were included. 177 patients (68%) had MD and 85 (32%) had non-measurable disease (NMD) at the time point of MTB presentation in accordance with RECIST v1.1. MD was not representative of the overall tumor burden in eleven patients (6%). The main reasons for NMD were lesions with longest diameter shorter than 10 mm (22%) and non-measurable peritoneal carcinomatosis (18%). Colorectal cancer and malignant melanoma displayed the highest rates of MD (> 75%). In contrast, gastric cancer, head and neck malignancies, and ovarian carcinoma had the lowest rates of MD (< 55%). In case of MD, the measurable lesions were representative of the overall tumor burden in the vast majority of cases (94%). CONCLUSION Approximately one third of cancer patients with advanced solid malignancies are not eligible for treatment response assessment in trials with endpoints ORR or DOR at the time of MTB presentation. The rate of patients eligible for trials with imaging endpoints differs significantly based on the underlying malignancy and should be taken under consideration during the planning of new precision oncology trials.
Collapse
Affiliation(s)
- Nabeel Mansour
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
| | - Danmei Zhang
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK partner site Munich), Heidelberg, Germany
| | - Michael Winkelmann
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Maria Ingenerf
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Lukas Gold
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Konstantin Klambauer
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK partner site Munich), Heidelberg, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Volker Heinemann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
| | - C Benedikt Westphalen
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Comprehensive Cancer Center München-LMU (CCCM LMU), LMU Munich, Munich, Germany.
| |
Collapse
|
27
|
Thiery J, Fahrner M. Integration of proteomics in the molecular tumor board. Proteomics 2024; 24:e2300002. [PMID: 38143279 DOI: 10.1002/pmic.202300002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
Cancer remains one of the most complex and challenging diseases in mankind. To address the need for a personalized treatment approach for particularly complex tumor cases, molecular tumor boards (MTBs) have been initiated. MTBs are interdisciplinary teams that perform in-depth molecular diagnostics to cooperatively and interdisciplinarily advise on the best therapeutic strategy. Current molecular diagnostics are routinely performed on the transcriptomic and genomic levels, aiming to identify tumor-driving mutations. However, these approaches can only partially capture the actual phenotype and the molecular key players of tumor growth and progression. Thus, direct investigation of the expressed proteins and activated signaling pathways provide valuable complementary information on the tumor-driving molecular characteristics of the tissue. Technological advancements in mass spectrometry-based proteomics enable the robust, rapid, and sensitive detection of thousands of proteins in minimal sample amounts, paving the way for clinical proteomics and the probing of oncogenic signaling activity. Therefore, proteomics is currently being integrated into molecular diagnostics within MTBs and holds promising potential in aiding tumor classification and identifying personalized treatment strategies. This review introduces MTBs and describes current clinical proteomics, its potential in precision oncology, and highlights the benefits of multi-omic data integration.
Collapse
Affiliation(s)
- Johanna Thiery
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Fahrner
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany
| |
Collapse
|
28
|
Hanssen F, Garcia MU, Folkersen L, Pedersen A, Lescai F, Jodoin S, Miller E, Seybold M, Wacker O, Smith N, Gabernet G, Nahnsen S. Scalable and efficient DNA sequencing analysis on different compute infrastructures aiding variant discovery. NAR Genom Bioinform 2024; 6:lqae031. [PMID: 38666213 PMCID: PMC11044436 DOI: 10.1093/nargab/lqae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
DNA variation analysis has become indispensable in many aspects of modern biomedicine, most prominently in the comparison of normal and tumor samples. Thousands of samples are collected in local sequencing efforts and public databases requiring highly scalable, portable, and automated workflows for streamlined processing. Here, we present nf-core/sarek 3, a well-established, comprehensive variant calling and annotation pipeline for germline and somatic samples. It is suitable for any genome with a known reference. We present a full rewrite of the original pipeline showing a significant reduction of storage requirements by using the CRAM format and runtime by increasing intra-sample parallelization. Both are leading to a 70% cost reduction in commercial clouds enabling users to do large-scale and cross-platform data analysis while keeping costs and CO2 emissions low. The code is available at https://nf-co.re/sarek.
Collapse
Affiliation(s)
- Friederike Hanssen
- Quantitative Biology Center, Eberhard-Karls University of Tübingen, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
- Department of Computer Science, Eberhard-Karls University of Tübingen, 72076 Baden-Württemberg, Germany
- M3 Research Center, University Hospital, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University of Tübingen, Tübingen 72076, Baden-Württemberg, Germany
| | - Maxime U Garcia
- Seqera Labs, Carrer de Marià Aguilò, 28, Barcelona 08005, Spain
- Barntumörbanken, Department of Oncology-Pathology, Karolinska Institutet, BioClinicum, Visionsgatan 4, Solna 17164, Sweden
- National Genomics Infrastructure, SciLifeLab, SciLifeLab, Tomtebodavägen 23, Solna 17165, Sweden
| | | | | | - Francesco Lescai
- Department of Biology and Biotechnology ”L. Spallanzani”, University of Pavia, via Ferrata, 9, Pavia, 27100 PV, Italy
| | - Susanne Jodoin
- Quantitative Biology Center, Eberhard-Karls University of Tübingen, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
- M3 Research Center, University Hospital, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
| | - Edmund Miller
- Department of Biological Sciences and Center for Systems Biology, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA
| | - Matthias Seybold
- Quantitative Biology Center, Eberhard-Karls University of Tübingen, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
| | - Oskar Wacker
- Quantitative Biology Center, Eberhard-Karls University of Tübingen, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
- M3 Research Center, University Hospital, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
| | - Nicholas Smith
- Department of Informatics, Technical University of Munich, Boltzmannstr. 3, Garching, 85748 Bavaria, Germany
| | - Gisela Gabernet
- Quantitative Biology Center, Eberhard-Karls University of Tübingen, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
- Department of Pathology, Yale School of Medicine, 300 George, New Haven, CT 06510, USA
| | - Sven Nahnsen
- Quantitative Biology Center, Eberhard-Karls University of Tübingen, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
- Department of Computer Science, Eberhard-Karls University of Tübingen, 72076 Baden-Württemberg, Germany
- M3 Research Center, University Hospital, Otfried-Müller Str. 37, Tübingen 72076, Baden-Württemberg, Germany
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard-Karls University of Tübingen, Tübingen 72076, Baden-Württemberg, Germany
- Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard-Karls University of Tübingen, Tübingen 72076, Baden-Württemberg, Germany
| |
Collapse
|
29
|
Schmidl B, Hütten T, Pigorsch S, Stögbauer F, Hoch CC, Hussain T, Wollenberg B, Wirth M. Assessing the role of advanced artificial intelligence as a tool in multidisciplinary tumor board decision-making for primary head and neck cancer cases. Front Oncol 2024; 14:1353031. [PMID: 38854718 PMCID: PMC11157509 DOI: 10.3389/fonc.2024.1353031] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 03/26/2024] [Indexed: 06/11/2024] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is a complex malignancy that requires a multidisciplinary approach in clinical practice, especially in tumor board discussions. In recent years, artificial intelligence has emerged as a tool to assist healthcare professionals in making informed decisions. This study investigates the application of ChatGPT 3.5 and ChatGPT 4.0, natural language processing models, in tumor board decision-making. Methods We conducted a pilot study in October 2023 on 20 consecutive head and neck cancer patients discussed in our multidisciplinary tumor board (MDT). Patients with a primary diagnosis of head and neck cancer were included. The MDT and ChatGPT 3.5 and ChatGPT 4.0 recommendations for each patient were compared by two independent reviewers and the number of therapy options, the clinical recommendation, the explanation and the summarization were graded. Results In this study, ChatGPT 3.5 provided mostly general answers for surgery, chemotherapy, and radiation therapy. For clinical recommendation, explanation and summarization ChatGPT 3.5 and 4.0 scored well, but demonstrated to be mostly an assisting tool, suggesting significantly more therapy options than our MDT, while some of the recommended treatment modalities like primary immunotherapy are not part of the current treatment guidelines. Conclusions This research demonstrates that advanced AI models at the moment can merely assist in the MDT setting, since the current versions list common therapy options, but sometimes recommend incorrect treatment options and in the case of ChatGPT 3.5 lack information on the source material.
Collapse
Affiliation(s)
- Benedikt Schmidl
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Tobias Hütten
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Steffi Pigorsch
- Department of RadioOncology, Technical University Munich, Munich, Germany
| | - Fabian Stögbauer
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Cosima C. Hoch
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Timon Hussain
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology Head and Neck Surgery, Technical University Munich, Munich, Germany
| |
Collapse
|
30
|
Kim SY, Kim JH, Kim TY, Park SR, Yoon S, Lee S, Lee SH, Kim TM, Han SW, Kim HR, Yun H, Lee S, Kim J, Choi YL, Choi KS, Chae H, Ryu H, Lee GW, Zang DY, Ahn JB. Pragmatic nationwide master observational trial based on genomic alterations in advanced solid tumors: KOrean Precision Medicine Networking Group Study of MOlecular profiling guided therapy based on genomic alterations in advanced Solid tumors (KOSMOS)-II study protocol KCSG AL-22-09. BMC Cancer 2024; 24:574. [PMID: 38724991 PMCID: PMC11080169 DOI: 10.1186/s12885-024-12338-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) has been introduced to many Korean institutions to support molecular diagnostics in cancer since 2017, when it became eligible for reimbursement by the National Health Insurance Service. However, the uptake of molecularly guided treatment (MGT) based on NGS results has been limited because of stringent regulations regarding prescriptions outside of approved indications, a lack of clinical trial opportunities, and limited access to molecular tumor boards (MTB) at most institutions. The KOSMOS-II study was designed to demonstrate the feasibility and effectiveness of MGT, informed by MTBs, using a nationwide precision medicine platform. METHODS The KOSMOS-II trial is a large-scale nationwide master observational study. It involves a framework for screening patients with metastatic solid tumors for actionable genetic alterations based on local NGS testing. It recommends MGT through a remote and centralized MTB meeting held biweekly. MGT can include one of the following options: Tier 1, the therapeutic use of investigational drugs targeting genetic alterations such as ALK, EGFR, ERBB2, BRAF, FH, ROS1, and RET, or those with high tumor mutational burden; Tier 2, comprising drugs with approved indications or those permitted for treatment outside of the indications approved by the Health Insurance Review and Assessment Service of Korea; Tier 3, involving clinical trials matching the genetic alterations recommended by the MTB. Given the anticipated proportion of patients receiving MGT in the range of 50% ± 3.25%, this study aims to enroll 1,000 patients. Patients must have progressed to one or more lines of therapy and undergone NGS before enrollment. DISCUSSION This pragmatic master protocol provides a mass-screening platform for rare genetic alterations and high-quality real-world data. Collateral clinical trials, translational studies, and clinico-genomic databases will contribute to generating evidence for drug repositioning and the development of new biomarkers. TRIAL REGISTRATION NCT05525858.
Collapse
Grants
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- HA22C0052 Ministry of Health and Welfare, Republic of Korea
- Roche, Basel, Switzerland
- Lunit, Seoul, Republic of Korea
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sook Ryun Park
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Shinkyo Yoon
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Soohyeon Lee
- Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul, South Korea
| | - Se-Hoon Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sae-Won Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul, South Korea
| | - Hongseok Yun
- Center for Genomic Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sejoon Lee
- Center for Precision Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jihun Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kui Son Choi
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, South Korea
| | - Heejung Chae
- Department of Internal Medicine, National Cancer Center, Goyang, South Korea
| | - Hyewon Ryu
- Division of Hematology and Oncology, Department of Internal Medicine, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Gyeong-Won Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Dae Young Zang
- Department of Internal Medicine, Hallym University College of Medicine, Hallym University Sacred Heart Hospital, Anyang, South Korea
| | - Joong Bae Ahn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Cancer Center, Seoul, South Korea
| |
Collapse
|
31
|
Gkountakos A, Singhi AD, Westphalen CB, Scarpa A, Luchini C. Fusion genes in pancreatic tumors. Trends Cancer 2024; 10:430-443. [PMID: 38378317 DOI: 10.1016/j.trecan.2024.01.009] [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: 10/18/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024]
Abstract
Gene fusions and rearrangements play a crucial role in tumor biology. They are rare events typically detected in KRAS wild-type (WT) pancreatic tumors. Their identification can inform clinical management by enabling precision oncology, as fusions involving BRAF, FGFR2, RET, NTRK, NRG1, and ALK represent actionable targets in KRAS-WT cancers, and serve diagnostic purposes since fusions involving PRKACA/B represent the diagnostic hallmark of intraductal oncocytic papillary neoplasms (IOPNs). Although they are rare, the therapeutic and diagnostic importance of these genomic events should not be underestimated, highlighting the need for quality-ensured molecular diagnostics in the management of cancer. Herein we review the existing literature on the role of fusion genes in pancreatic tumors and their clinical potential as effective biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | - Aatur D Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - C Benedikt Westphalen
- Department of Medicine III and Comprehensive Cancer Centre (CCC), LMU University Hospital Munich and German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Aldo Scarpa
- ARC-Net Research Center, University of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy.
| | - Claudio Luchini
- ARC-Net Research Center, University of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy.
| |
Collapse
|
32
|
Michaelis J, Himmelsbach R, Metzger P, Lassmann S, Börries M, Werner M, Miething C, Höfflin R, Illert AL, Duyster J, Becker H, Sigle A, Gratzke C, Grabbert M. Primary Results of Patients with Genitourinary Malignancies Presented at a Molecular Tumor Board. Urol Int 2024; 108:383-391. [PMID: 38626735 DOI: 10.1159/000538908] [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: 03/03/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
INTRODUCTION Personalized medicine poses great opportunities and challenges. While the therapeutic landscape markedly expands, descriptions about status, clinical implementation and real-world benefits of precision oncology and molecular tumor boards (MTB) remain sparse, particularly in the field of genitourinary (GU) cancer. Hence, this study characterized urological MTB cases to better understand the potential role of MTB in uro-oncology. METHODS We analyzed patients with complete data sets being reviewed at an MTB from January 2019 to October 2022, focusing on results of molecular analysis and treatment recommendations. RESULTS We evaluated 102 patients with GU cancer with a mean patient age of 61.7 years. Prostate cancer (PCa) was the most frequent entity with 52.9% (54/102), followed by bladder cancer (18.6%, 19/102) and renal cell carcinoma (14.7%, 15/102). On average, case presentation at MTB took place 54.9 months after initial diagnosis and after 2.7 previous lines of therapy. During the study period, 49.0% (50/102) of patients deceased. Additional MTB-based treatment recommendations were achieved in a majority of 68.6% (70/102) of patients, with a recommendation for targeted therapy in 64.3% (45/70) of these patients. Only 6.7% (3/45) of patients - due to different reasons - received the recommended MTB-based therapy though, with 33% (1/3) of patients reaching disease control. Throughout the MTB study period, GU cancer case presentations and treatment recommendations increased, while the time interval between initial presentation and final therapy recommendation were decreasing over time. CONCLUSION Presentation of uro-oncological patients at the MTB is a highly valuable measure for clinical decision-making. Prospectively, earlier presentation of patients at the MTB and changing legislative issues regarding comprehensive molecular testing and targeted treatment approval might further improve patients' benefits from comprehensive molecular diagnostics.
Collapse
Affiliation(s)
- Jakob Michaelis
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ruth Himmelsbach
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Metzger
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Silke Lassmann
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Börries
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Werner
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius Miething
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rouven Höfflin
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna L Illert
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Justus Duyster
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Becker
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - August Sigle
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus Grabbert
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
33
|
Boehm D, Strantz C, Christoph J, Busch H, Ganslandt T, Unberath P. Data Visualization Support for Tumor Boards and Clinical Oncology: Protocol for a Scoping Review. JMIR Res Protoc 2024; 13:e53627. [PMID: 38441925 PMCID: PMC10951826 DOI: 10.2196/53627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Complex and expanding data sets in clinical oncology applications require flexible and interactive visualization of patient data to provide the maximum amount of information to physicians and other medical practitioners. Interdisciplinary tumor conferences in particular profit from customized tools to integrate, link, and visualize relevant data from all professions involved. OBJECTIVE The scoping review proposed in this protocol aims to identify and present currently available data visualization tools for tumor boards and related areas. The objective of the review will be to provide not only an overview of digital tools currently used in tumor board settings, but also the data included, the respective visualization solutions, and their integration into hospital processes. METHODS The planned scoping review process is based on the Arksey and O'Malley scoping study framework. The following electronic databases will be searched for articles published in English: PubMed, Web of Knowledge, and SCOPUS. Eligible articles will first undergo a deduplication step, followed by the screening of titles and abstracts. Second, a full-text screening will be used to reach the final decision about article selection. At least 2 reviewers will independently screen titles, abstracts, and full-text reports. Conflicting inclusion decisions will be resolved by a third reviewer. The remaining literature will be analyzed using a data extraction template proposed in this protocol. The template includes a variety of meta information as well as specific questions aiming to answer the research question: "What are the key features of data visualization solutions used in molecular and organ tumor boards, and how are these elements integrated and used within the clinical setting?" The findings will be compiled, charted, and presented as specified in the scoping study framework. Data for included tools may be supplemented with additional manual literature searches. The entire review process will be documented in alignment with the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews) flowchart. RESULTS The results of this scoping review will be reported per the expanded PRISMA-ScR guidelines. A preliminary search using PubMed, Web of Knowledge, and Scopus resulted in 1320 articles after deduplication that will be included in the further review process. We expect the results to be published during the second quarter of 2024. CONCLUSIONS Visualization is a key process in leveraging a data set's potentially available information and enabling its use in an interdisciplinary setting. The scoping review described in this protocol aims to present the status quo of visualization solutions for tumor board and clinical oncology applications and their integration into hospital processes. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/53627.
Collapse
Affiliation(s)
- Dominik Boehm
- Medical Center for Information and Communication Technology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Cancer Research Center (Bayerisches Zentrum für Krebsforschung), Erlangen, Germany
| | - Cosima Strantz
- Chair of Medical Informatics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Christoph
- Chair of Medical Informatics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Junior Research Group (Bio-)medical Data Science, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Hauke Busch
- Group for Medical Systems Biology, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Thomas Ganslandt
- Chair of Medical Informatics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Unberath
- Medical Center for Information and Communication Technology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- SRH Fürth University of Applied Sciences, Fürth, Germany
| |
Collapse
|
34
|
Pestana RC, Lopes David BB, Pires de Camargo V, Munhoz RR, Lopes de Mello CA, González Donna ML, Haro Varas JC, Zapata ML, Cunha Martins CL, Chacon M, Schmerling R, Jesus-Garcia R. Challenges and opportunities for sarcoma care and research in Latin America: a position paper from the LACOG sarcoma group. LANCET REGIONAL HEALTH. AMERICAS 2024; 30:100671. [PMID: 38259252 PMCID: PMC10801304 DOI: 10.1016/j.lana.2023.100671] [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: 05/28/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024]
Abstract
As a developing region, Latin America faces unique cancer control and prevention challenges, which are intensified when considering rare cancers, including sarcomas. Sarcomas are a group of malignancies that arise in the connective tissues of the body-such as muscle, fat, nerves, blood vessels, and bones-accounting for a diverse range of tumours that, although rare, require specialized attention. Sarcoma care and research in Latin America require a comprehensive approach that includes deeper epidemiologic knowledge, diagnostic capacity building, access to innovative treatments, increased patient advocacy, and strengthening of clinical research capacity. This article will review current challenges and opportunities for treating patients with sarcoma in Latin America and outline a pathway toward improvement for regional collaborative groups.
Collapse
Affiliation(s)
- Roberto Carmagnani Pestana
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Bruna Bianca Lopes David
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Oncoclínicas, Rio de Janeiro, Brazil
- Instituto Nacional do Câncer, Rio de Janeiro, Brazil
| | - Veridiana Pires de Camargo
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Hospital Beneficência Portuguesa, São Paulo, Brazil
| | - Rodrigo Ramella Munhoz
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Hospital Sírio Libanês, São Paulo, Brazil
| | - Celso Abdon Lopes de Mello
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- AC Camargo Cancer Center, São Paulo, Brazil
| | - María Lucila González Donna
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Instituto Nacional del Cancer Paraguay, Capiata, Paraguay
| | - Juan Carlos Haro Varas
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Instituto Nacional de Enfermedades Neoplásicas, Lima, Perú
| | - Maycos L. Zapata
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Instituto de Cancerologia las Americas AUNA, Medellin, Colombia
| | - Cicero Luiz Cunha Martins
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Instituto Nacional do Câncer, Rio de Janeiro, Brazil
- Americas Oncologia, Rio de Janeiro, Brazil
| | - Matias Chacon
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Fleming Institute, Buenos Aires, Argentina
| | - Rafael Schmerling
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Dasa Oncologia, São Paulo, Brazil
| | - Reynaldo Jesus-Garcia
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
35
|
Riedl JM, Moik F, Esterl T, Kostmann SM, Gerger A, Jost PJ. Molecular diagnostics tailoring personalized cancer therapy-an oncologist's view. Virchows Arch 2024; 484:169-179. [PMID: 37982847 PMCID: PMC10948510 DOI: 10.1007/s00428-023-03702-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/27/2023] [Accepted: 11/04/2023] [Indexed: 11/21/2023]
Abstract
Medical oncology is rapidly evolving with the implementation of personalized, targeted therapies. Advances in molecular diagnostics and the biologic understanding of cancer pathophysiology led to the identification of specific genetic alterations as drivers of cancer progression. Further, improvements in drug development enable the direct interference with these pathways, which allow tailoring personalized treatments based on a distinct molecular characterization of tumors. Thereby, we are currently experiencing a paradigm-shift in the treatment of cancers towards cancer-type agnostic, molecularly targeted, personalized therapies. However, this concept has several important hurdles and limitations to overcome to ultimately increase the proportion of patients benefitting from the precision oncology approach. These include the assessment of clinical relevancy of identified alterations, capturing and interpreting levels of heterogeneity based on intra-tumoral or time-dependent molecular evolution, and challenges in the practical implementation of precision oncology in routine clinical care. In the present review, we summarize the current state of cancer-agnostic precision oncology, discuss the concept of molecular tumor boards, and consider current limitations of personalized cancer therapy. Further, we provide an outlook towards potential future developments including the implementation of functionality assessments of identified genetic alterations and the broader use of liquid biopsies in order to obtain more comprehensive and longitudinal genetic information that might guide personalized cancer therapy in the future.
Collapse
Affiliation(s)
- Jakob M Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Moik
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Tamara Esterl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Sarah M Kostmann
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp J Jost
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
- Medical Department III for Haematology and Oncology, School of Medicine, Technical University of Munich, Munich, Germany.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
36
|
Hönikl LS, Lange S, Butenschoen VM, Delbridge C, Meyer B, Combs SE, Illert AL, Schmidt-Graf F. The role of molecular tumor boards in neuro-oncology: a nationwide survey. BMC Cancer 2024; 24:108. [PMID: 38243190 PMCID: PMC10797778 DOI: 10.1186/s12885-024-11858-x] [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: 08/07/2023] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND In neuro-oncology, the inclusion of tumor patients in the molecular tumor board has only become increasingly widespread in recent years, but so far there are no standards for indication, procedure, evaluation, therapy recommendations and therapy implementation of neuro-oncological patients. The present work examines the current handling of neuro-oncological patients included in molecular tumor boards in Germany. METHODS We created an online based survey with questions covering the handling of neuro-oncologic patient inclusion, annotation of genetic analyses, management of target therapies and the general role of molecular tumor boards in neuro-oncology in Germany. We contacted all members of the Neuro-Oncology working group (NOA) of the German Cancer Society (DKG) by e-mail. RESULTS 38 responses were collected. The majority of those who responded were specialists in neurosurgery or neurology with more than 10 years of professional experience working at a university hospital. Molecular tumor boards (MTB) regularly take place once a week and all treatment disciplines of neuro-oncology patients take part. The inclusions to the MTB are according to distinct tumors and predominantly in case of tumor recurrence. An independently MTB member mostly create the recommendations, which are regularly implemented in the tumor treatment. Recommendations are given for alteration classes 4 and 5. Problems exist mostly within the cost takeover of experimental therapies. The experimental therapies are mostly given in the department of medical oncology. CONCLUSIONS Molecular tumor boards for neuro-oncological patients, by now, are not standardized in Germany. Similarities exists for patient inclusion and interpretation of molecular alterations; the time point of inclusion and implementation during the patient treatment differ between the various hospitals. Further studies for standardization and harmonisation are needed. In summary, most of the interviewees envision great opportunities and possibilities for molecular-based neuro-oncological therapy in the future.
Collapse
Affiliation(s)
- Lisa S Hönikl
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, 81675, Munich, Germany.
- Center for Personalized Medicine (ZPM), Klinikum rechts der Isar, Technical Universitiy of Munich (TUM), Munich, Germany.
| | - Sebastian Lange
- Center for Personalized Medicine (ZPM), Klinikum rechts der Isar, Technical Universitiy of Munich (TUM), Munich, Germany
- Department of Medicine II, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Vicki M Butenschoen
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, 81675, Munich, Germany
| | - Claire Delbridge
- Center for Personalized Medicine (ZPM), Klinikum rechts der Isar, Technical Universitiy of Munich (TUM), Munich, Germany
- Department of Neuropathology, Institute of Pathology, Technical University of Munich (TUM), Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, 81675, Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Anna Lena Illert
- Center for Personalized Medicine (ZPM), Klinikum rechts der Isar, Technical Universitiy of Munich (TUM), Munich, Germany
- Department of Medicine III, Faculty of Medicine, Klinikum Rechts der Isar, Technical University Munich (TUM), Munich, Germany
| | - Friederike Schmidt-Graf
- Center for Personalized Medicine (ZPM), Klinikum rechts der Isar, Technical Universitiy of Munich (TUM), Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
37
|
Sotelo-Rodríguez C, Vallejo-Ardila D, Ruiz-Patiño A, Chamorro DF, Rodríguez J, Moreno-Pérez DA, Carranza H, Otero J, Vargas C, Archila P, Rojas L, Zuluaga J, Rubio C, Ordóñez-Reyes C, Garcia-Robledo JE, Mejía S, Jaller E, Arrieta O, Cardona AF. Molecular Tumor Board Improves Outcomes for Hispanic Patients With Advanced Solid Tumors. JCO Glob Oncol 2024; 10:e2300011. [PMID: 38237094 PMCID: PMC10805441 DOI: 10.1200/go.23.00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/10/2023] [Accepted: 10/18/2023] [Indexed: 01/23/2024] Open
Abstract
PURPOSE Multidisciplinary molecular tumor boards (MTBs) decode complex genomic data into clinical recommendations. Although MTBs are well-established in the oncology practice in developed countries, this strategy needs to be better explored in developing countries. Herein, we describe the possible benefits and limitations of the first MTB established in Colombia. METHODS Demographic, clinical, and genomic information was collected between August 2020 and November 2021. By mid-2020, an MTB strategy was created to discuss clinical cases with one or more genomic alterations identified by next-generation sequencing using an open-access virtual platform. We characterized the patient population as benefiting from the recommended treatment option. We assessed the benefits and access to available targeted therapies that have the potential to change clinical management by making recommendations to treating oncologists on the basis of genomic profiling. However, we did not assess the treatment oncologists' compliance with MTB recommendations because they were not intended to replace clinical judgment/standard of care. RESULTS A total of 146 patients were included in the discussions of the MTB. The median age was 59 years, and 59.6% were women. Genomic results prompting a change in therapeutic decisions were obtained in 53.1% of patients (95% CI, 44.9 to 61.3). The most prevalent malignancy was non-small-cell lung cancer (51%). Other malignancies represented 60%, 50%, and 30% of patients with soft-tissue sarcomas, brain tumors, and breast cancer, respectively. CONCLUSION Using an open-access virtual platform, MTBs were feasible in low- and middle-income countries on the basis of the capability to provide the benefits and access to available targeted therapies that are not standard of care. Furthermore, MTB recommendations were made available to the treating oncologist in different locations across Colombia, providing the option to modify clinical management in most of these patients.
Collapse
Affiliation(s)
- Carolina Sotelo-Rodríguez
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Dora Vallejo-Ardila
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Alejandro Ruiz-Patiño
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Diego F. Chamorro
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - July Rodríguez
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Darwin A. Moreno-Pérez
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Hernán Carranza
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Jorge Otero
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Carlos Vargas
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Pilar Archila
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Leonardo Rojas
- Thoracic Oncology Unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá, Colombia
| | - Jairo Zuluaga
- Thoracic Oncology Unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá, Colombia
| | - Cladelis Rubio
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
| | - Camila Ordóñez-Reyes
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | | | - Sergio Mejía
- Thoracic Oncology Unit, Clínica Las Américas, Medellín, Colombia
| | - Elvira Jaller
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit, National Cancer Institute (INCan), México City, México
| | - Andrés F. Cardona
- Thoracic Oncology Unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá, Colombia
- Direction of Research, Science and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá, Colombia
| |
Collapse
|
38
|
Grauman Å, Ancillotti M, Veldwijk J, Mascalzoni D. Precision cancer medicine and the doctor-patient relationship: a systematic review and narrative synthesis. BMC Med Inform Decis Mak 2023; 23:286. [PMID: 38098034 PMCID: PMC10722840 DOI: 10.1186/s12911-023-02395-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The implementation of precision medicine is likely to have a huge impact on clinical cancer care, while the doctor-patient relationship is a crucial aspect of cancer care that needs to be preserved. This systematic review aimed to map out perceptions and concerns regarding how the implementation of precision medicine will impact the doctor-patient relationship in cancer care so that threats against the doctor-patient relationship can be addressed. METHODS Electronic databases (Pubmed, Scopus, Web of Science, Social Science Premium Collection) were searched for articles published from January 2010 to December 2021, including qualitative, quantitative, and theoretical methods. Two reviewers completed title and abstract screening, full-text screening, and data extraction. Findings were summarized and explained using narrative synthesis. RESULTS Four themes were generated from the included articles (n = 35). Providing information addresses issues of information transmission and needs, and of complex concepts such as genetics and uncertainty. Making decisions in a trustful relationship addresses opacity issues, the role of trust, and and physicians' attitude towards the role of precision medicine tools in decision-making. Managing negative reactions of non-eligible patients addresses patients' unmet expectations of precision medicine. Conflicting roles in the blurry line between clinic and research addresses issues stemming from physicians' double role as doctors and researchers. CONCLUSIONS Many findings have previously been addressed in doctor-patient communication and clinical genetics. However, precision medicine adds complexity to these fields and further emphasizes the importance of clear communication on specific themes like the distinction between genomic and gene expression and patients' expectations about access, eligibility, effectiveness, and side effects of targeted therapies.
Collapse
Affiliation(s)
- Å Grauman
- Centre for Research Ethics and Bioethics, Uppsala University, Box 564, Uppsala, SE-751 22, Sweden.
| | - M Ancillotti
- Centre for Research Ethics and Bioethics, Uppsala University, Box 564, Uppsala, SE-751 22, Sweden
| | - J Veldwijk
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, the Netherlands
- Erasmus Choice Modelling Centre, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - D Mascalzoni
- Centre for Research Ethics and Bioethics, Uppsala University, Box 564, Uppsala, SE-751 22, Sweden
- Erasmus Choice Modelling Centre, Erasmus University Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
39
|
Buchholz SM, Nause N, König U, Reinecke J, Steuber B, Ammer-Herrmenau C, Reuter-Jessen K, Bohnenberger H, Biggemann L, Braulke F, Neesse A, Ellenrieder V, Ströbel P, Adler M, König A. Time to Deliver on Promises: The Role of ERBB2 Alterations as Treatment Options for Colorectal Cancer Patients in the Era of Precision Oncology. J Pers Med 2023; 13:1701. [PMID: 38138928 PMCID: PMC10745079 DOI: 10.3390/jpm13121701] [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: 10/10/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Receptor tyrosine kinase erythroblastic oncogene B2 (ERBB2), also known as human epidermal growth factor receptor 2 (HER2), represents an oncogenic driver and has been effectively targeted in breast and gastric cancer. Recently, next-generation sequencing (NGS) discovered ERBB2 as a promising therapeutic target in metastatic colorectal cancer (mCRC), where it is altered in 3-5% of patients, but no therapies are currently approved for this use. Herein, we present the experience of a single center in diagnosing actionable genetic ERBB2 alterations using NGS and utilizing the latest therapeutic options. Between October 2019 and December 2022, a total of 107 patients with advanced CRC underwent molecular analysis, revealing actionable ERBB2 mutations in two patients and ERBB2 amplifications in two other patients. These findings correlated with immunohistochemical (IHC) staining. Of these four patients, two were treated with trastuzumab-deruxtecan (T-DXd). We present two exemplary cases of patients with actionable ERBB2 alterations to demonstrate the effectiveness of T-DXd in heavily pretreated ERBB2-positive mCRC patients and the need for early molecular profiling. To fully exploit the potential of this promising treatment, earlier molecular profiling and the initiation of targeted therapies are essential.
Collapse
Affiliation(s)
- Soeren M. Buchholz
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Nelia Nause
- Göttingen Comprehensive Cancer Center (G-CCC), University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ute König
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Johanna Reinecke
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Benjamin Steuber
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Christoph Ammer-Herrmenau
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Kirsten Reuter-Jessen
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Lorenz Biggemann
- Institute of Diagnostic and Interventional Radiology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Friederike Braulke
- Göttingen Comprehensive Cancer Center (G-CCC), University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Marius Adler
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
- Department of Gastroenterology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Alexander König
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
40
|
Werner T, Fahrner M, Schilling O. Using proteomics for stratification and risk prediction in patients with solid tumors. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:176-182. [PMID: 37999758 DOI: 10.1007/s00292-023-01261-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/25/2023]
Abstract
Proteomics, the study of proteins and their functions, has greatly evolved due to advances in analytical chemistry and computational biology. Unlike genomics or transcriptomics, proteomics captures the dynamic and diverse nature of proteins, which play crucial roles in cellular processes. This is exemplified in cancer, where genomic and transcriptomic information often falls short in reflecting actual protein expression and interactions. Liquid chromatography-mass spectrometry (LC-MS) is pivotal in proteomic data generation, enabling high-throughput analysis of protein samples. The MS-based workflow involves protein digestion, chromatographic separation, ionization, and fragmentation, leading to peptide identification and quantification. Computational biostatistics, particularly using tools in R (R Foundation for Statistical Computing, Vienna, Austria; www.R-project.org ), aid in data analysis, revealing protein expression patterns and correlations with clinical variables. Proteomic studies can be explorative, aiming to characterize entire proteomes, or targeted, focusing on specific proteins of interest. The integration of proteomics with genomics addresses database limitations and enhances peptide identification. Case studies in intrahepatic cholangiocarcinoma, glioblastoma multiforme, and pancreatic ductal adenocarcinoma highlight proteomics' clinical applications, from subtyping cancers to identifying diagnostic markers. Moreover, proteomic data augment molecular tumor boards by providing deeper insights into pathway activities and genomic mutations, supporting personalized treatment decisions. Overall, proteomics contributes significantly to advancing our understanding of cellular biology and improving clinical care.
Collapse
Affiliation(s)
- Tilman Werner
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre Freiburg, University of Freiburg, Breisacher Str. 115a, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Matthias Fahrner
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre Freiburg, University of Freiburg, Breisacher Str. 115a, 79106, Freiburg, Germany
- German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Faculty of Medicine, University Medical Centre Freiburg, University of Freiburg, Breisacher Str. 115a, 79106, Freiburg, Germany.
- German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany.
| |
Collapse
|
41
|
Le Tourneau C, André F, Helland Å, Mileshkin L, Minnaard W, Schiel A, Taskén K, Thomas DM, Veronese ML, Durán-Pacheco G, Leyens L, Rufibach K, Thomas M, Krämer A. Modified study designs to expand treatment options in personalised oncology: a multistakeholder view. Eur J Cancer 2023; 194:113278. [PMID: 37820553 DOI: 10.1016/j.ejca.2023.113278] [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: 05/16/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 10/13/2023]
Abstract
Personalised oncology, whereby patients are given therapies based on their molecular tumour profile, is rapidly becoming an essential part of optimal clinical care, at least partly facilitated by recent advances in next-generation sequencing-based technology using liquid- and tissue-based biopsies. Consequently, clinical trials have shifted in approach, from traditional studies evaluating cytotoxic chemotherapy in largely histology-based populations to modified, biomarker-driven studies (e.g. basket, umbrella, platform) of molecularly guided therapies and cancer immunotherapies in selected patient subsets. Such modified study designs may assess, within the same trial structure, multiple cancer types and treatments, and should incorporate a multistakeholder perspective. This is key to generating complementary, fit-for-purpose and timely evidence for molecularly guided therapies that can be used as proof-of-concept to inform further study designs, lead to approval by regulatory authorities and be used as confirmation of clinical benefit for health technology assessment bodies. In general, the future of cancer clinical trials requires a framework for the application of innovative technologies and dynamic design methodologies, in order to efficiently transform scientific discoveries into clinical utility. Next-generation, modified studies that involve the joint efforts of all key stakeholders will offer individualised strategies that ultimately contribute to globalised knowledge and collective learning. In this review, we outline the background and purpose of such modified study designs and detail key aspects from a multistakeholder perspective. We also provide methodological considerations for designing the studies and highlight how insights from already-ongoing studies may address current challenges and opportunities in the era of personalised oncology.
Collapse
Affiliation(s)
- Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, INSERM U900 Research Unit, Paris-Saclay University, Paris, France
| | | | - Åslaug Helland
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linda Mileshkin
- Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | | | | | - Kjetil Taskén
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - David M Thomas
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - Lada Leyens
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
42
|
Mock A, Teleanu MV, Kreutzfeldt S, Heilig CE, Hüllein J, Möhrmann L, Jahn A, Hanf D, Kerle IA, Singh HM, Hutter B, Uhrig S, Fröhlich M, Neumann O, Hartig A, Brückmann S, Hirsch S, Grund K, Dikow N, Lipka DB, Renner M, Bhatti IA, Apostolidis L, Schlenk RF, Schaaf CP, Stenzinger A, Schröck E, Hübschmann D, Heining C, Horak P, Glimm H, Fröhling S. NCT/DKFZ MASTER handbook of interpreting whole-genome, transcriptome, and methylome data for precision oncology. NPJ Precis Oncol 2023; 7:109. [PMID: 37884744 PMCID: PMC10603123 DOI: 10.1038/s41698-023-00458-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Analysis of selected cancer genes has become an important tool in precision oncology but cannot fully capture the molecular features and, most importantly, vulnerabilities of individual tumors. Observational and interventional studies have shown that decision-making based on comprehensive molecular characterization adds significant clinical value. However, the complexity and heterogeneity of the resulting data are major challenges for disciplines involved in interpretation and recommendations for individualized care, and limited information exists on how to approach multilayered tumor profiles in clinical routine. We report our experience with the practical use of data from whole-genome or exome and RNA sequencing and DNA methylation profiling within the MASTER (Molecularly Aided Stratification for Tumor Eradication Research) program of the National Center for Tumor Diseases (NCT) Heidelberg and Dresden and the German Cancer Research Center (DKFZ). We cover all relevant steps of an end-to-end precision oncology workflow, from sample collection, molecular analysis, and variant prioritization to assigning treatment recommendations and discussion in the molecular tumor board. To provide insight into our approach to multidimensional tumor profiles and guidance on interpreting their biological impact and diagnostic and therapeutic implications, we present case studies from the NCT/DKFZ molecular tumor board that illustrate our daily practice. This manual is intended to be useful for physicians, biologists, and bioinformaticians involved in the clinical interpretation of genome-wide molecular information.
Collapse
Affiliation(s)
- Andreas Mock
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Maria-Veronica Teleanu
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Rheumatology, Heidelberg Unversity Hospital, Heidelberg, Germany
| | - Simon Kreutzfeldt
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph E Heilig
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jennifer Hüllein
- Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Lino Möhrmann
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases/University Cancer Center (NCT/UCC) Dresden, Dresden, Germany
- DKFZ, Heidelberg, Germany
| | - Arne Jahn
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus, Technische Universität Dresden and Hereditary Cancer Syndrome Center Dresden, Dresden, Germany
| | - Dorothea Hanf
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases/University Cancer Center (NCT/UCC) Dresden, Dresden, Germany
- DKFZ, Heidelberg, Germany
| | - Irina A Kerle
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases/University Cancer Center (NCT/UCC) Dresden, Dresden, Germany
- DKFZ, Heidelberg, Germany
| | - Hans Martin Singh
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany
| | - Barbara Hutter
- Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Sebastian Uhrig
- Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Martina Fröhlich
- Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Olaf Neumann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Hartig
- Institute of Pathology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sascha Brückmann
- Institute of Pathology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Steffen Hirsch
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Kerstin Grund
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Nicola Dikow
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel B Lipka
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Translational Cancer Epigenomics, Division of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Marcus Renner
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Irfan Ahmed Bhatti
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany
| | - Leonidas Apostolidis
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- Department of Hematology, Oncology and Rheumatology, Heidelberg Unversity Hospital, Heidelberg, Germany
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany
- NCT Trial Center, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Christian P Schaaf
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Evelin Schröck
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus, Technische Universität Dresden and Hereditary Cancer Syndrome Center Dresden, Dresden, Germany
| | - Daniel Hübschmann
- Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Christoph Heining
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases/University Cancer Center (NCT/UCC) Dresden, Dresden, Germany
- DKFZ, Heidelberg, Germany
| | - Peter Horak
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hanno Glimm
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases/University Cancer Center (NCT/UCC) Dresden, Dresden, Germany
- DKFZ, Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
43
|
Ballatore Z, Bozzi F, Cardea S, Savino FD, Migliore A, Tarantino V, Chiodi N, Ambrosini E, Bianchi F, Goteri G, Filosa A, Barbisan F, Bartoli E, Papa R, Berardi R. Molecular Tumour Board (MTB): From Standard Therapy to Precision Medicine. J Clin Med 2023; 12:6666. [PMID: 37892804 PMCID: PMC10607087 DOI: 10.3390/jcm12206666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Background: In the metastatic setting, cancer patients may not benefit from standard care regimes and their diseases undergo drug resistance due to tumour cell heterogeneity and genomic landscape complexity. In recent years, there have been several attempts to personalise the diagnostic-therapeutic path and to propose novel strategies based on not only histological test results but also on each patient's clinical history and molecular biology. Profiling molecular tests allows physicians to investigate the single tumour genomic landscape and to promote targeted approaches. The Molecular Tumour Board (MTB) is a multidisciplinary committee dedicated to selecting individualised and targeted therapeutic strategies appropriate for patients suffering from diseases that present resistance to standard care. Materials and Methods: Our MTB settled in "Azienda Ospedaliero Universitaria delle Marche", Ancona (AN), Italy, and includes oncologists, molecular biologists, geneticists, and other specialists. Clinical cases are referred by physicians to the MTB, through the Cancer and Research Centre of the Marche Region (CORM), through a telemedicine platform. Four possible molecular profiles are available: FoundationOne® CDx e FoundationOne®Liquid CDx and two local Next Generation Sequencing (NGS) panels, with 16 DNA genes and 10 RNA genes respectively. The resulting genetic mutations and their analyses are evaluated by all the members of the Board and a report for each patient is provided with medical recommendations. Results: from June 2021 to May 2023, we collected data from 97 referral patients (M: 49, F: 48). The mean age was 60.6 years (range 22-83 years). 90 cases were approved for testing. Only seven patients were not eligible for genomic profiling. In two patients who were eligible, molecular profiling was not performed because a tissue sample was not available. Off-label therapy was recommended for three patients. 5% of cases (5/88) showed addressable driver mutations associated with an existing targeted therapy and were immediately enrolled. Conclusions: MTB presents a powerful tool for offering precise medical goals. Our Department of Clinical Oncology also takes advantage of the important role of multidisciplinary teams, through the establishment of CORM and MTB meetings, within which there is the chance to perform NGS-based analyses. It will be important in the future to implement the use of genomic profiling to improve personalised care and to guide the choice of suitable therapies and more appropriate management of patients.
Collapse
Affiliation(s)
- Zelmira Ballatore
- Department of Medical Oncology, AOU delle Marche, 60126 Ancona, Italy; (Z.B.); (V.T.)
| | - Francesco Bozzi
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Sara Cardea
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Francesco Domenico Savino
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Antonella Migliore
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Valentina Tarantino
- Department of Medical Oncology, AOU delle Marche, 60126 Ancona, Italy; (Z.B.); (V.T.)
| | - Natalia Chiodi
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Elisa Ambrosini
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Francesca Bianchi
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Gaia Goteri
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Alessandra Filosa
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Francesca Barbisan
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Elisa Bartoli
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Roberto Papa
- Quality, Risk Management and Health Technology Innovation Unit, Department of Staff, AOU delle Marche, 60126 Ancona, Italy;
| | - Rossana Berardi
- Department of Medical Oncology, AOU delle Marche, 60126 Ancona, Italy; (Z.B.); (V.T.)
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| |
Collapse
|
44
|
Rodriguez Castells M, Baraibar I, Ros J, Saoudi N, Salvà F, García A, Alcaraz A, Tabernero J, Élez E. The impact of clinical and translational research on the quality of life during the metastatic colorectal cancer patient journey. Front Oncol 2023; 13:1272561. [PMID: 37909013 PMCID: PMC10614292 DOI: 10.3389/fonc.2023.1272561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/30/2023] [Indexed: 11/02/2023] Open
Abstract
The journey of metastatic colorectal cancer patients is complex and challenging, requiring coordination and collaboration between multiple healthcare providers. Understanding patients' needs, fears, feelings, concerns, and behaviors is essential for providing individualized patient-centered care. In recent years, mCRC patients have experienced improvements in clinical outcomes, from 16 months of overall survival to 32 months, thanks to research. However, there is still room for improvement, and integrating clinical and translational research into routine practice can help patients benefit from treatments and techniques that would not be an option. In the Journey of mCRC patients, living well with cancer and quality of life becomes a priority given the outcomes of the disease. Patient reported outcomes (PRO) and Patient Reported Outcome Measures (PROMs) are becoming therefore new estimands in Oncology. Patient advocates represent important figures in this process by prioritizing issues and research questions; evaluating research designs and the performance of the research; the analysis and interpretation of data; and how results are disseminated. Multidisciplinary Tumor Boards and shared decision-making is essential for designing treatment strategies for individual patients. Quality of Life is often prioritized only when it comes to refractory advanced disease and end-of-life care, but it has to be integrated from the beginning, as the emotional impact of diagnosis leads to a vulnerable situation where patients' needs and preferences can be easily overseen. First-line treatment will be chosen among more treatment options than subsequent lines, with longer progression-free survival and a bigger impact on the outcomes. Practicing patient-centered care and optimizing first-line treatment for colorectal cancer patients requires a comprehensive understanding of patient experience and treatment outcomes, which can guide clinical practice and inform regulatory decisions for the benefit of patients.
Collapse
Affiliation(s)
- Marta Rodriguez Castells
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Iosune Baraibar
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Javier Ros
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Nadia Saoudi
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Francesc Salvà
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Ariadna García
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Adriana Alcaraz
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Josep Tabernero
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Elena Élez
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
45
|
Giacomini P, Valenti F, Allegretti M, Pallocca M, De Nicola F, Ciuffreda L, Fanciulli M, Scalera S, Buglioni S, Melucci E, Casini B, Carosi M, Pescarmona E, Giordani E, Sperati F, Jannitti N, Betti M, Maugeri-Saccà M, Cecere FL, Villani V, Pace A, Appetecchia M, Vici P, Savarese A, Krasniqi E, Ferraresi V, Russillo M, Fabi A, Landi L, Minuti G, Cappuzzo F, Zeuli M, Ciliberto G. The Molecular Tumor Board of the Regina Elena National Cancer Institute: from accrual to treatment in real-world. J Transl Med 2023; 21:725. [PMID: 37845764 PMCID: PMC10577953 DOI: 10.1186/s12967-023-04595-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Molecular Tumor Boards (MTB) operating in real-world have generated limited consensus on good practices for accrual, actionable alteration mapping, and outcome metrics. These topics are addressed herein in 124 MTB patients, all real-world accrued at progression, and lacking approved therapy options. METHODS Actionable genomic alterations identified by tumor DNA (tDNA) and circulating tumor DNA (ctDNA) profiling were mapped by customized OncoKB criteria to reflect diagnostic/therapeutic indications as approved in Europe. Alterations were considered non-SoC when mapped at either OncoKB level 3, regardless of tDNA/ctDNA origin, or at OncoKB levels 1/2, provided they were undetectable in matched tDNA, and had not been exploited in previous therapy lines. RESULTS Altogether, actionable alterations were detected in 54/124 (43.5%) MTB patients, but only in 39 cases (31%) were these alterations (25 from tDNA, 14 from ctDNA) actionable/unexploited, e.g. they had not resulted in the assignment of pre-MTB treatments. Interestingly, actionable and actionable/unexploited alterations both decreased (37.5% and 22.7% respectively) in a subset of 88 MTB patients profiled by tDNA-only, but increased considerably (77.7% and 66.7%) in 18 distinct patients undergoing combined tDNA/ctDNA testing, approaching the potential treatment opportunities (76.9%) in 147 treatment-naïve patients undergoing routine tDNA profiling for the first time. Non-SoC therapy was MTB-recommended to all 39 patients with actionable/unexploited alterations, but only 22 (56%) accessed the applicable drug, mainly due to clinical deterioration, lengthy drug-gathering procedures, and geographical distance from recruiting clinical trials. Partial response and stable disease were recorded in 8 and 7 of 19 evaluable patients, respectively. The time to progression (TTP) ratio (MTB-recommended treatment vs last pre-MTB treatment) exceeded the conventional Von Hoff 1.3 cut-off in 9/19 cases, high absolute TTP and Von Hoff values coinciding in 3 cases. Retrospectively, 8 patients receiving post-MTB treatment(s) as per physician's choice were noted to have a much longer overall survival from MTB accrual than 11 patients who had received no further treatment (35.09 vs 6.67 months, p = 0.006). CONCLUSIONS MTB-recommended/non-SoC treatments are effective, including those assigned by ctDNA-only alterations. However, real-world MTBs may inadvertently recruit patients electively susceptible to diverse and/or multiple treatments.
Collapse
Affiliation(s)
- Patrizio Giacomini
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy.
| | - Fabio Valenti
- UOC Translational Oncology Research, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Matteo Allegretti
- UOC Translational Oncology Research, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Matteo Pallocca
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Francesca De Nicola
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Ludovica Ciuffreda
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Maurizio Fanciulli
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Stefano Scalera
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Elisa Melucci
- Department of Pathology, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Beatrice Casini
- Department of Pathology, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Mariantonia Carosi
- Department of Pathology, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Elena Giordani
- UOC Translational Oncology Research, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Francesca Sperati
- Clinical Trial Center, Biostatistics and Bioinformatics, San Gallicano Dermatological Institute IRCCS, 00144, Rome, Italy
| | - Nicoletta Jannitti
- Pharmacy Unit, Medical Direction, IRCCS-Regina Elena National Cancer Institute and San Gallicano Institute, 00144, Rome, Italy
| | - Martina Betti
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Marcello Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
- Medical Oncology 2, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | | | - Veronica Villani
- Neuro-Oncology Unit, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Andrea Pace
- Neuro-Oncology Unit, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Patrizia Vici
- Phase IV Studies, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Antonella Savarese
- Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Eriseld Krasniqi
- Phase IV Studies, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Virginia Ferraresi
- Sarcomas and Rare Tumors Departmental Unit, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Michelangelo Russillo
- Sarcomas and Rare Tumors Departmental Unit, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Alessandra Fabi
- Precision Medicine Unit in Senology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Lorenza Landi
- Clinical Trial Center: Phase 1 and Precision Medicine, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Gabriele Minuti
- Clinical Trial Center: Phase 1 and Precision Medicine, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Federico Cappuzzo
- Medical Oncology 2, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Massimo Zeuli
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
- Medical Oncology 1, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS-Regina Elena National Cancer Institute, 00144, Rome, Italy
| |
Collapse
|
46
|
Vodicska B, Déri J, Tihanyi D, Várkondi E, Kispéter E, Dóczi R, Lakatos D, Dirner A, Vidermann M, Filotás P, Szalkai-Dénes R, Szegedi I, Bartyik K, Gábor KM, Simon R, Hauser P, Péter G, Kiss C, Garami M, Peták I. Real-world performance analysis of a novel computational method in the precision oncology of pediatric tumors. World J Pediatr 2023; 19:992-1008. [PMID: 36914906 PMCID: PMC10497647 DOI: 10.1007/s12519-023-00700-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/31/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND The utility of routine extensive molecular profiling of pediatric tumors is a matter of debate due to the high number of genetic alterations of unknown significance or low evidence and the lack of standardized and personalized decision support methods. Digital drug assignment (DDA) is a novel computational method to prioritize treatment options by aggregating numerous evidence-based associations between multiple drivers, targets, and targeted agents. DDA has been validated to improve personalized treatment decisions based on the outcome data of adult patients treated in the SHIVA01 clinical trial. The aim of this study was to evaluate the utility of DDA in pediatric oncology. METHODS Between 2017 and 2020, 103 high-risk pediatric cancer patients (< 21 years) were involved in our precision oncology program, and samples from 100 patients were eligible for further analysis. Tissue or blood samples were analyzed by whole-exome (WES) or targeted panel sequencing and other molecular diagnostic modalities and processed by a software system using the DDA algorithm for therapeutic decision support. Finally, a molecular tumor board (MTB) evaluated the results to provide therapy recommendations. RESULTS Of the 100 cases with comprehensive molecular diagnostic data, 88 yielded WES and 12 panel sequencing results. DDA identified matching off-label targeted treatment options (actionability) in 72/100 cases (72%), while 57/100 (57%) showed potential drug resistance. Actionability reached 88% (29/33) by 2020 due to the continuous updates of the evidence database. MTB approved the clinical use of a DDA-top-listed treatment in 56 of 72 actionable cases (78%). The approved therapies had significantly higher aggregated evidence levels (AELs) than dismissed therapies. Filtering of WES results for targeted panels missed important mutations affecting therapy selection. CONCLUSIONS DDA is a promising approach to overcome challenges associated with the interpretation of extensive molecular profiling in the routine care of high-risk pediatric cancers. Knowledgebase updates enable automatic interpretation of a continuously expanding gene set, a "virtual" panel, filtered out from genome-wide analysis to always maximize the performance of precision treatment planning.
Collapse
Affiliation(s)
- Barbara Vodicska
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Júlia Déri
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Dóra Tihanyi
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Edit Várkondi
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Enikő Kispéter
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Róbert Dóczi
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Dóra Lakatos
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Anna Dirner
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Mátyás Vidermann
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | - Péter Filotás
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary
| | | | - István Szegedi
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Bartyik
- Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Krisztina Míta Gábor
- Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Réka Simon
- Onco-Hematology Department, Velkey László Paediatric Health Centre, Miskolc, Hungary
| | - Péter Hauser
- Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - György Péter
- Onco-Hematology Department, Heim Pál Children's Hospital, Budapest, Hungary
| | - Csongor Kiss
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Garami
- Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - István Peták
- Oncompass Medicine Hungary Kft, Retek Str. 34, Budapest, 1024, Hungary.
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, USA.
- Genomate Health, Cambridge, MA, USA.
| |
Collapse
|
47
|
Witte HM, Riedl J, Künstner A, Fähnrich A, Ketzer J, Fliedner SMJ, Reimer N, Bernard V, von Bubnoff N, Merz H, Busch H, Feller A, Gebauer N. Molecularly Stratified Treatment Options in Primary Refractory DLBCL/HGBL with MYC and BCL2 or BCL6 Rearrangements (HGBL, NOS with MYC/BCL6). Target Oncol 2023; 18:749-765. [PMID: 37488307 PMCID: PMC10517902 DOI: 10.1007/s11523-023-00983-5] [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] [Accepted: 06/25/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND There is growing evidence supporting multidisciplinary molecular tumor boards (MTB) in solid tumors whereas hematologic malignancies remain underrepresented in this regard. OBJECTIVE The present study aimed to assess the clinical relevance of MTBs in primary refractory diffuse large B-cell lymphomas/high-grade B-cell lymphomas with MYC and BCL2 rearrangements (prDLBCL/HGBL-MYC/BCL2) (n = 13) and HGBL, not otherwise specified (NOS), with MYC and BCL6 rearrangements (prHGBL, NOS-MYC/BCL6) (n = 6) based on our previously published whole-exome sequencing (WES) cohort. PATIENTS AND METHODS For genomic analysis, the institutional MTB WES pipeline (University Cancer Center Schleswig-Holstein: UCCSH), certified for routine clinical diagnostics, was employed and supplemented by a comprehensive immunohistochemical work-up. Consecutive database research and annotation according to established evidence levels for molecularly stratified therapies was performed (NCT-DKTK/ESCAT). RESULTS Molecularly tailored treatment options with NCT-DKTK evidence level of at least m2A were identified in each case. We classified mutations in accordance with biomarker/treatment baskets and detected a heterogeneous spectrum of targetable alterations affecting immune evasion (IE; n = 30), B-cell targets (BCT; n = 26), DNA damage repair (DDR; n = 20), tyrosine kinases (TK; n = 13), cell cycle (CC; n = 7), PI3K-MTOR-AKT pathway (PAM; n = 2), RAF-MEK-ERK cascade (RME; n = 1), and others (OTH; n = 11). CONCLUSION Our virtual MTB approach identified potential molecularly targeted treatment options alongside targetable genomic signatures for both prDLBCL/HGBL-MYC/BCL2 and prHGBL, NOS-MYC/BCL6. These results underline the potential of MTB consultations in difficult-to-treat lymphomas early in the treatment sequence.
Collapse
Affiliation(s)
- Hanno M Witte
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
- Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany.
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany.
| | - Jörg Riedl
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
| | - Axel Künstner
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Anke Fähnrich
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Julius Ketzer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Department of Pediatrics, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Stephanie M J Fliedner
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Niklas Reimer
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Veronica Bernard
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
| | - Hartmut Merz
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Hauke Busch
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Alfred Feller
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
| |
Collapse
|
48
|
Fasola G, Barducci MC, Tozzi VD, Cavanna L, Cinieri S, Perrone F, Pinto C, Russo A, Sapino A, Grossi F, Aprile G. Implementation of Precision Oncology in the National Healthcare System: A Statement Proposal Endorsed by Italian Scientific Societies. JCO Precis Oncol 2023; 7:e2300166. [PMID: 37944071 PMCID: PMC10645411 DOI: 10.1200/po.23.00166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/15/2023] [Accepted: 09/08/2023] [Indexed: 11/12/2023] Open
Abstract
PURPOSE Precision oncology (PO) promises positive results for patients. To date, in Italy, the effort to implement PO has been made autonomously by regional health institutions in a top-down fashion. This approach is not very efficient and jeopardizes patients' equal access to PO. Similar outcomes have been recorded in other Western countries. We tested a method of collaboration among professionals, scientific societies, and government institutions to facilitate the delivery of PO innovation to patients' bedsides. METHOD We designed an organizational research project on the basis of a bottom-up approach. We started by observing PO-related activities in five health care authorities (HCAs) in one Italian region. We then compared the issues that emerged with those of three additional HCAs in other Italian regions. Using the results of the initial observation and adopting validated multiple-step consensus methods, we finally derived 14 statements that were approved by the four main scientific societies of oncology and pathology at the national level. RESULTS The 14 statements addressed the main issues linked to the implementation of PO in clinical practice. The strong professional consensus advocated for prompt adoption within the national healthcare system. CONCLUSIONS The consensus on the statements that were obtained shows the importance of a synergistic effort among professionals, scientific societies, and health care institutions in defining homogeneous solutions for innovation implementation within the health care system.
Collapse
Affiliation(s)
- Gianpiero Fasola
- Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Maria C. Barducci
- Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Valeria D. Tozzi
- Centre for Research on Health and Social Care Management (CERGAS), SDA Bocconi School of Management, Milan, Italy
| | - Luigi Cavanna
- Piacenza General Hospital, AUSL Piacenza, Piacenza, Italy
- Collegio Italiano dei Primari Oncologi Medici Ospedalieri (CIPOMO), Genova, Italy
| | - Saverio Cinieri
- Complex Medical Oncology Unit, ASL Brindisi Senatore Antonio Perrino Hospital, Brindisi, Italy
- Associazione Italiana Oncologia Medica (AIOM), Milano, Italy
| | - Francesco Perrone
- Associazione Italiana Oncologia Medica (AIOM), Milano, Italy
- Clinical Trials Unit, Istituto Nazionale Tumori-IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Carmine Pinto
- Medical Oncology, Comprehensive Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
- Collegio degli Oncologi Medici Universitari (COMU), University of Palermo, Palermo, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Società Italiana di Anatomia Patologica e Citologia Diagnostica (SIAPeC), Milano, Italy
| | | | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Vicenza, Italy
| |
Collapse
|
49
|
Bonilla CE, Montenegro P, O’Connor JM, Hernando-Requejo O, Aranda E, Pinto Llerena J, Llontop A, Gallardo Escobar J, Díaz Romero MDC, Bautista Hernández Y, Graña Suárez B, Batagelj EJ, Wali Mushtaq A, García-Foncillas J. Ibero-American Consensus Review and Incorporation of New Biomarkers for Clinical Practice in Colorectal Cancer. Cancers (Basel) 2023; 15:4373. [PMID: 37686649 PMCID: PMC10487247 DOI: 10.3390/cancers15174373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Advances in genomic technologies have significantly improved the management of colorectal cancer (CRC). Several biomarkers have been identified in CRC that enable personalization in the use of biologic agents that have shown to enhance the clinical outcomes of patients. However, technologies used for their determination generate massive amounts of information that can be difficult for the clinician to interpret and use adequately. Through several discussion meetings, a group of oncology experts from Spain and several Latin American countries reviewed the latest literature to provide practical recommendations on the determination of biomarkers in CRC based on their clinical experience. The article also describes the importance of looking for additional prognostic biomarkers and the use of histopathology to establish an adequate molecular classification. Present and future of immunotherapy biomarkers in CRC patients are also discussed, together with several techniques for marker determination, including liquid biopsy, next-generation sequencing (NGS), polymerase chain reaction (PCR), and fecal immunohistochemical tests. Finally, the role of Molecular Tumor Boards in the diagnosis and treatment of CRC is described. All of this information will allow us to highlight the importance of biomarker determination in CRC.
Collapse
Affiliation(s)
- Carlos Eduardo Bonilla
- Fundación CTIC—Centro de Tratamiento e Investigación sobre Cáncer, Bogotá 1681442, Colombia
| | - Paola Montenegro
- Institución AUNA OncoSalud e Instituto Nacional de Enfermedades Neoplásicas, Lima 15023, Peru
| | | | | | - Enrique Aranda
- Departamento de Oncología Médica, Hospital Reina Sofía, IMIBIC, UCO, CIBERONC, 14004 Cordoba, Spain;
| | | | - Alejandra Llontop
- Instituto de Oncología Ángel H. Roffo, Ciudad Autónoma de Buenos Aires C1437FBG, Argentina
| | | | | | | | - Begoña Graña Suárez
- Servicio de Oncología Médica, Hospital Universitario de A Coruña, Servicio Galego de Saúde (SERGAS), 15006 A Coruña, Spain;
| | | | | | - Jesús García-Foncillas
- Hospital Universitario Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
| |
Collapse
|
50
|
Zhang D, Dorman K, Heinrich K, Weiss L, Boukovala M, Haas M, Greif PA, Ziemann F, Beyer G, Roessler D, Goni E, Renz B, D'Haese JG, Kunz WG, Seidensticker M, Corradini S, Niyazi M, Ormanns S, Kumbrink J, Jung A, Mock A, Rudelius M, Klauschen F, Werner J, Mayerle J, von Bergwelt-Baildon M, Boeck S, Heinemann V, Westphalen CB. A Retrospective Analysis of Biliary Tract Cancer Patients Presented to the Molecular Tumor Board at the Comprehensive Cancer Center Munich. Target Oncol 2023; 18:767-776. [PMID: 37594677 PMCID: PMC10517894 DOI: 10.1007/s11523-023-00985-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND AND OBJECTIVE With the rising importance of precision oncology in biliary tract cancer (BTC), the aim of this retrospective single-center analysis was to describe the clinical and molecular characteristics of patients with BTC who underwent comprehensive genomic profiling (CGP) and were discussed in the CCCMunichLMU molecular tumor board (MTB). PATIENTS AND METHODS In this single-center observational study, we included BTC patients with intrahepatic cholangiocarcinoma (iCCA), extrahepatic CCA (eCCA), and gallbladder cancer (GB), who had been discussed in the institutional MTB from May 29, 2017, to July 25, 2022. Patients were followed up until 31 January 2023. Data were retrospectively collected by review of medical charts, and MTB recommendation. RESULTS In total, 153 cases were registered to the MTB with a median follow-up of 15 months. Testing was successful in 81.7% of the patients. CGP detected targetable alterations in 35.3% of our BTC patients (most commonly ARID1A/ERBB2/IDH1/PIK3CA/BRAF-mutations and FGFR2-fusions). Recommendations for molecularly guided therapy were given in 46.4%. Of those, treatment implementation of targeted therapy followed in 19.4%. In patients receiving the recommended treatment, response rate was 57% and median overall survival was 19 months (vs 8 months in the untreated cohort). The progression-free survival ratio of 1.45 suggest a clinical benefit of molecularly guided treatment. CONCLUSIONS In line with previous work, our series demonstrates feasibility and clinical utility of comprehensive genomic profiling in BTC patients. With the growing number of targeted agents with clinical activity in BTC, CGP should become standard of care in the management of this group of patients.
Collapse
Affiliation(s)
- Danmei Zhang
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Klara Dorman
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Lena Weiss
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Myrto Boukovala
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Michael Haas
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Philipp A Greif
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Frank Ziemann
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Georg Beyer
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Daniel Roessler
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Elisabetta Goni
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Bernhard Renz
- Department of General, Visceral and Transplant Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Jan G D'Haese
- Department of General, Visceral and Transplant Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Corradini
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | | | - Jörg Kumbrink
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Pathology, LMU Munich, Munich, Germany
| | - Andreas Jung
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Pathology, LMU Munich, Munich, Germany
| | - Andreas Mock
- Institute of Pathology, LMU Munich, Munich, Germany
| | | | - Frederick Klauschen
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Institute of Pathology, LMU Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Julia Mayerle
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Stefan Boeck
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Volker Heinemann
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - C Benedikt Westphalen
- Department of Medicine III and Comprehensive Cancer Center, University Hospital, Klinikum Grosshadern, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| |
Collapse
|