1
|
Ballin N, Ott A, Seibel-Kelemen O, Bonzheim I, Nann D, Beha J, Spahn S, Singer S, Ossowski S, Roggia C, Schroeder C, Bitzer M, Armeanu-Ebinger S. Case Report: FGFR2 inhibitor resistance via PIK3CA and CDKN2A/B in an intrahepatic cholangiocarcinoma patient with FGFR2-SH3GLB1 fusion. Front Oncol 2025; 15:1527484. [PMID: 40260297 PMCID: PMC12009697 DOI: 10.3389/fonc.2025.1527484] [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: 11/13/2024] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
FGFR2 fusions occur in up to 14% of patients with intrahepatic cholangiocarcinoma (iCCA) and have been considered as therapeutic target for FGFR inhibitors (FGFRi). However, response to targeted treatment may be limited due to the emergence of various resistance mechanisms. We report a case of recurrent iCCA in a 43-year-old patient with a FGFR2 fusion, who was treated with Lenvatinib. Next-generation sequencing (NGS) of tumor-normal DNA and tumor RNA under Lenvatinib treatment confirmed the FGFR2 fusion, however no further molecular resistance mutation was observed. After failure of FGFRi treatment (Lenvatinib and Infigratinib) ten months later, repeated NGS analysis revealed a new gain-of-function mutation in PIK3CA and a homozygous deletion of CDKN2A/B, potentially representing an acquired resistance mechanism. The emerging acquired resistance to FGFR inhibitor treatment has implications for subsequent treatment strategies.
Collapse
Affiliation(s)
- Nadja Ballin
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Alexander Ott
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Olga Seibel-Kelemen
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Irina Bonzheim
- Department of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Dominik Nann
- Department of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Janina Beha
- Center for Personalized Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Spahn
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Singer
- Department of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Stephan Ossowski
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Cristiana Roggia
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| | - Michael Bitzer
- Center for Personalized Medicine, University Hospital Tübingen, Tübingen, Germany
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Sorin Armeanu-Ebinger
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
2
|
Lörsch AM, Jung J, Lange S, Pfarr N, Mogler C, Illert AL. [Personalized medicine in oncology]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:180-189. [PMID: 38568256 DOI: 10.1007/s00292-024-01315-8] [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: 02/16/2024] [Indexed: 04/26/2024]
Abstract
Due to the considerable technological progress in molecular and genetic diagnostics as well as increasing insights into the molecular pathogenesis of diseases, there has been a fundamental paradigm shift in the past two decades from a "one-size-fits-all approach" to personalized, molecularly informed treatment strategies. Personalized medicine or precision medicine focuses on the genetic, physiological, molecular, and biochemical differences between individuals and considers their effects on the development, prevention, and treatment of diseases. As a pioneer of personalized medicine, the field of oncology is particularly noteworthy, where personalized diagnostics and treatment have led to lasting change in the treatment of cancer patients in recent years. In this article, the significant change towards personalized treatment concepts, especially in the field of personalized oncology, will be discussed and examined in more detail.
Collapse
Affiliation(s)
- Alisa Martina Lörsch
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
| | - Johannes Jung
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
| | - Sebastian Lange
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
| | - Nicole Pfarr
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, München, Deutschland
| | - Carolin Mogler
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, München, Deutschland
| | - Anna Lena Illert
- Zentrum für Personalisierte Medizin (ZPM), Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, München, Deutschland.
- Klinik und Poliklinik für Innere Medizin III, Hämatologie und Onkologie, Klinikum rechts der Isar, Technische Universität München, München, Deutschland.
- Bayerisches Zentrum für Krebsforschung (BZKF), Standort Technische Universität München, München, Deutschland.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Standort München, München, Deutschland.
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, München, Deutschland.
- Klinik für Innere Medizin I, Abteilung für Hämatologie, Onkologie und Stammzelltransplantation, Universitätsklinikum Freiburg, Freiburg, Deutschland.
| |
Collapse
|
3
|
Spahn S, Kleinhenz F, Shevchenko E, Stahl A, Rasen Y, Geisler C, Ruhm K, Klaumuenzer M, Kronenberger T, Laufer SA, Sundberg-Malek H, Bui KC, Horger M, Biskup S, Schulze-Osthoff K, Templin M, Malek NP, Poso A, Bitzer M. The molecular interaction pattern of lenvatinib enables inhibition of wild-type or kinase-mutated FGFR2-driven cholangiocarcinoma. Nat Commun 2024; 15:1287. [PMID: 38346946 PMCID: PMC10861557 DOI: 10.1038/s41467-024-45247-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 01/18/2024] [Indexed: 02/15/2024] Open
Abstract
Fibroblast growth factor receptor (FGFR)-2 can be inhibited by FGFR-selective or non-selective tyrosine kinase inhibitors (TKIs). Selective TKIs are approved for cholangiocarcinoma (CCA) with FGFR2 fusions; however, their application is limited by a characteristic pattern of adverse events or evocation of kinase domain mutations. A comprehensive characterization of a patient cohort treated with the non-selective TKI lenvatinib reveals promising efficacy in FGFR2-driven CCA. In a bed-to-bench approach, we investigate FGFR2 fusion proteins bearing critical tumor-relevant point mutations. These mutations confer growth advantage of tumor cells and increased resistance to selective TKIs but remain intriguingly sensitive to lenvatinib. In line with clinical observations, in-silico analyses reveal a more favorable interaction pattern of lenvatinib with FGFR2, including an increased flexibility and ligand efficacy, compared to FGFR-selective TKIs. Finally, the treatment of a patient with progressive disease and a newly developed kinase mutation during therapy with a selective inhibitor results in a striking response to lenvatinib. Our in vitro, in silico, and clinical data suggest that lenvatinib is a promising treatment option for FGFR2-driven CCA, especially when insurmountable adverse reactions of selective TKIs or acquired kinase mutations occur.
Collapse
Affiliation(s)
- Stephan Spahn
- Department of Internal Medicine I, University Hospital Tuebingen, 72076, Tuebingen, Germany.
| | - Fabian Kleinhenz
- Department of Internal Medicine I, University Hospital Tuebingen, 72076, Tuebingen, Germany
| | - Ekaterina Shevchenko
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-University, 72076, Tuebingen, Germany
- Tuebingen Center for Academic Drug Discovery & Development (TüCAD2), 72076, Tuebingen, Germany
| | - Aaron Stahl
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| | - Yvonne Rasen
- Department of Internal Medicine I, University Hospital Tuebingen, 72076, Tuebingen, Germany
| | - Christine Geisler
- Department of Internal Medicine I, University Hospital Tuebingen, 72076, Tuebingen, Germany
| | - Kristina Ruhm
- Center for Personalized Medicine, Eberhard-Karls University, 72076, Tuebingen, Germany
| | | | - Thales Kronenberger
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-University, 72076, Tuebingen, Germany
- Tuebingen Center for Academic Drug Discovery & Development (TüCAD2), 72076, Tuebingen, Germany
| | - Stefan A Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-University, 72076, Tuebingen, Germany
- Tuebingen Center for Academic Drug Discovery & Development (TüCAD2), 72076, Tuebingen, Germany
- Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, 72076, Tuebingen, Germany
| | - Holly Sundberg-Malek
- Center for Personalized Medicine, Eberhard-Karls University, 72076, Tuebingen, Germany
| | - Khac Cuong Bui
- Department of Internal Medicine I, University Hospital Tuebingen, 72076, Tuebingen, Germany
| | - Marius Horger
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls University, 72076, Tuebingen, Germany
| | - Saskia Biskup
- CeGaT GmbH and Praxis für Humangenetik, 72076, Tuebingen, Germany
| | - Klaus Schulze-Osthoff
- Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, 72076, Tuebingen, Germany
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard-Karls University, 72076, Tuebingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Markus Templin
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| | - Nisar P Malek
- Department of Internal Medicine I, University Hospital Tuebingen, 72076, Tuebingen, Germany
- Center for Personalized Medicine, Eberhard-Karls University, 72076, Tuebingen, Germany
- Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, 72076, Tuebingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, 72076, Tuebingen, Germany
| | - Antti Poso
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-University, 72076, Tuebingen, Germany
- Tuebingen Center for Academic Drug Discovery & Development (TüCAD2), 72076, Tuebingen, Germany
- Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, 72076, Tuebingen, Germany
- School of Pharmacy, University of Eastern Finland, 70210, Kuopio, Finland
| | - Michael Bitzer
- Department of Internal Medicine I, University Hospital Tuebingen, 72076, Tuebingen, Germany.
- Center for Personalized Medicine, Eberhard-Karls University, 72076, Tuebingen, Germany.
- Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, 72076, Tuebingen, Germany.
- M3-Research Center for Malignome, Metabolome and Microbiome, Eberhard-Karls University, 72076, Tuebingen, Germany.
| |
Collapse
|
4
|
Einhaus J, Rochwarger A, Mattern S, Gaudillière B, Schürch CM. High-multiplex tissue imaging in routine pathology-are we there yet? Virchows Arch 2023; 482:801-812. [PMID: 36757500 PMCID: PMC10156760 DOI: 10.1007/s00428-023-03509-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/22/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023]
Abstract
High-multiplex tissue imaging (HMTI) approaches comprise several novel immunohistological methods that enable in-depth, spatial single-cell analysis. Over recent years, studies in tumor biology, infectious diseases, and autoimmune conditions have demonstrated the information gain accessible when mapping complex tissues with HMTI. Tumor biology has been a focus of innovative multiparametric approaches, as the tumor microenvironment (TME) contains great informative value for accurate diagnosis and targeted therapeutic approaches: unraveling the cellular composition and structural organization of the TME using sophisticated computational tools for spatial analysis has produced histopathologic biomarkers for outcomes in breast cancer, predictors of positive immunotherapy response in melanoma, and histological subgroups of colorectal carcinoma. Integration of HMTI technologies into existing clinical workflows such as molecular tumor boards will contribute to improve patient outcomes through personalized treatments tailored to the specific heterogeneous pathological fingerprint of cancer, autoimmunity, or infection. Here, we review the advantages and limitations of existing HMTI technologies and outline how spatial single-cell data can improve our understanding of pathological disease mechanisms and determinants of treatment success. We provide an overview of the analytic processing and interpretation and discuss how HMTI can improve future routine clinical diagnostic and therapeutic processes.
Collapse
Affiliation(s)
- Jakob Einhaus
- Department of Anaesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Alexander Rochwarger
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Sven Mattern
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Brice Gaudillière
- Department of Anaesthesiology, Perioperative & Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Perelló-Reus CM, Rubio-Tomás T, Cisneros-Barroso E, Ibargüen-González L, Segura-Sampedro JJ, Morales-Soriano R, Barceló C. Challenges in precision medicine in pancreatic cancer: A focus in cancer stem cells and microbiota. Front Oncol 2022; 12:995357. [PMID: 36531066 PMCID: PMC9751445 DOI: 10.3389/fonc.2022.995357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Pancreatic cancer adenocarcinoma (PDAC) is a lethal disease, with the lowest 5-years survival rate of all cancers due to late diagnosis. Despite the advance and success of precision oncology in gastrointestinal cancers, the frequency of molecular-informed therapy decisions in PDAC is currently neglectable. The reasons for this dismal situation are mainly the absence of effective early diagnostic biomarkers and therapy resistance. PDAC cancer stem cells (PDAC-SC), which are regarded as essential for tumor initiation, relapse and drug resistance, are highly dependent on their niche i.e. microanatomical structures of the tumor microenvironment. There is an altered microbiome in PDAC patients embedded within the highly desmoplastic tumor microenvironment, which is known to determine therapeutic responses and affecting survival in PDAC patients. We consider that understanding the communication network that exists between the microbiome and the PDAC-SC niche by co-culture of patient-derived organoids (PDOs) with TME microbiota would recapitulate the complexity of PDAC paving the way towards a precision oncology treatment-response prediction.
Collapse
Affiliation(s)
- Catalina M. Perelló-Reus
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases (HUSE), Palma de Mallorca, Spain
| | | | | | - Lesly Ibargüen-González
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases (HUSE), Palma de Mallorca, Spain
| | - Juan José Segura-Sampedro
- Advanced Oncological Surgery, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
- General and Digestive Surgery Unit, Hospital Universitari Son Espases, School of Medicine, Balearic Islands Health Research Institute, University of Balearic Islands, Palma de Mallorca, Spain
| | - Rafael Morales-Soriano
- Advanced Oncological Surgery, Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
- General and Digestive Surgery Unit, Hospital Universitari Son Espases, School of Medicine, Balearic Islands Health Research Institute, University of Balearic Islands, Palma de Mallorca, Spain
| | - Carles Barceló
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases (HUSE), Palma de Mallorca, Spain
| |
Collapse
|
6
|
Heinrich K, Miller-Phillips L, Ziemann F, Hasselmann K, Rühlmann K, Flach M, Biro D, von Bergwelt-Baildon M, Holch J, Herold T, von Baumgarten L, Greif PA, Jeremias I, Wuerstlein R, Casuscelli J, Spitzweg C, Seidensticker M, Renz B, Corradini S, Baumeister P, Goni E, Tufman A, Jung A, Kumbrink J, Kirchner T, Klauschen F, Metzeler KH, Heinemann V, Westphalen CB. Lessons learned: the first consecutive 1000 patients of the CCCMunich LMU Molecular Tumor Board. J Cancer Res Clin Oncol 2022; 149:1905-1915. [PMID: 35796778 PMCID: PMC9261163 DOI: 10.1007/s00432-022-04165-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/18/2022] [Indexed: 12/04/2022]
Abstract
Purpose In 2016, the University of Munich Molecular Tumor Board (MTB) was implemented to initiate a precision oncology program. This review of cases was conducted to assess clinical implications and functionality of the program, to identify current limitations and to inform future directions of these efforts. Methods Charts, molecular profiles, and tumor board decisions of the first 1000 consecutive cases (01/2016–03/2020) were reviewed. Descriptive statistics were applied to describe relevant findings. Results Of the first 1000 patients presented to the MTB; 914 patients received comprehensive genomic profiling. Median age of patients was 56 years and 58% were female. The most prevalent diagnoses were breast (16%) and colorectal cancer (10%). Different types of targeted or genome-wide sequencing assays were used; most of them offered by the local department of pathology. Testing was technically successful in 88%. In 41% of cases, a genomic alteration triggered a therapeutic recommendation. The fraction of patients receiving a tumor board recommendation differed significantly between malignancies ranging from over 50% in breast or biliary tract to less than 30% in pancreatic cancers. Based on a retrospective chart review, 17% of patients with an MTB recommendation received appropriate treatment. Conclusion Based on these retrospective analyses, patients with certain malignancies (breast and biliary tract cancer) tend to be more likely to have actionable variants. The low rate of therapeutic implementation (17% of patients receiving a tumor board recommendation) underscores the importance of meticulous follow-up for these patients and ensuring broad access to innovative therapies for patients receiving molecular tumor profiling. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-022-04165-0.
Collapse
Affiliation(s)
- Kathrin Heinrich
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| | - Lisa Miller-Phillips
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Frank Ziemann
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Korbinian Hasselmann
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Katharina Rühlmann
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - Madeleine Flach
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - Dorottya Biro
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Julian Holch
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Tobias Herold
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurology and Comprehensive Cancer Center (CCC Munich LMU), Ludwig Maximilians University, Munich, Germany
| | - Philipp A Greif
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Irmela Jeremias
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, German Center for Environmental Health (HMGU), Munich, Germany
- Department of Pediatrics, Dr Von Hauner Children's Hospital, LMU, Munich, Germany
| | - Rachel Wuerstlein
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Jozefina Casuscelli
- Department of Urology and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Christine Spitzweg
- Department of Medicine IV and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Bernhard Renz
- Department of General, Visceral und Transplantation Surgery and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Corradini
- Department of Radiation Oncology and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Philipp Baumeister
- Department of Otorhinolaryngology, Head and Neck Surgery and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Elisabetta Goni
- Department of Medicine II and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Amanda Tufman
- Department of Medicine V and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Munich, Germany
| | - Andreas Jung
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Institute of Pathology, Ludwig Maximilians University (LMU), Munich, Germany
| | - Jörg Kumbrink
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Institute of Pathology, Ludwig Maximilians University (LMU), Munich, Germany
| | - Thomas Kirchner
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Institute of Pathology, Ludwig Maximilians University (LMU), Munich, Germany
| | - Frederick Klauschen
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
- Institute of Pathology, Ludwig Maximilians University (LMU), Munich, Germany
| | - Klaus H Metzeler
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Hematology, Cell Therapy and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Volker Heinemann
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - C Benedikt Westphalen
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
- Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany.
| |
Collapse
|
7
|
Reizine N, O’Donnell PH. Modern developments in germline pharmacogenomics for oncology prescribing. CA Cancer J Clin 2022; 72:315-332. [PMID: 35302652 PMCID: PMC9262778 DOI: 10.3322/caac.21722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
The integration of genomic data into personalized treatment planning has revolutionized oncology care. Despite this, patients with cancer remain vulnerable to high rates of adverse drug events and medication inefficacy, affecting prognosis and quality of life. Pharmacogenomics is a field seeking to identify germline genetic variants that contribute to an individual's unique drug response. Although there is widespread integration of genomic information in oncology, somatic platforms, rather than germline biomarkers, have dominated the attention of cancer providers. Patients with cancer potentially stand to benefit from improved integration of both somatic and germline genomic information, especially because the latter may complement treatment planning by informing toxicity risk for drugs with treatment-limiting tolerabilities and narrow therapeutic indices. Although certain germline pharmacogenes, such as TPMT, UGT1A1, and DPYD, have been recognized for decades, recent attention has illuminated modern potential dosing implications for a whole new set of anticancer agents, including targeted therapies and antibody-drug conjugates, as well as the discovery of additional genetic variants and newly relevant pharmacogenes. Some of this information has risen to the level of directing clinical action, with US Food and Drug Administration label guidance and recommendations by international societies and governing bodies. This review is focused on key new pharmacogenomic evidence and oncology-specific dosing recommendations. Personalized oncology care through integrated pharmacogenomics represents a unique multidisciplinary collaboration between oncologists, laboratory science, bioinformatics, pharmacists, clinical pharmacologists, and genetic counselors, among others. The authors posit that expanded consideration of germline genetic information can further transform the safe and effective practice of oncology in 2022 and beyond.
Collapse
Affiliation(s)
- Natalie Reizine
- Division of Hematology and Oncology, Department of Medicine, The University of Illinois at Chicago
| | - Peter H. O’Donnell
- Section of Hematology/Oncology, Department of Medicine, Center for Personalized Therapeutics, and Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago
- Correspondence to: Dr. Peter H. O’Donnell, Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue, MC2115, Chicago, IL 60637, USA. ()
| |
Collapse
|
8
|
Sawada K, Nakayama K, Nakamura K, Yoshimura Y, Razia S, Ishikawa M, Yamashita H, Ishibashi T, Sato S, Kyo S. Clinical Outcomes of Genotype-Matched Therapy for Recurrent Gynecological Cancers: A Single Institutional Experience. Healthcare (Basel) 2021; 9:healthcare9101395. [PMID: 34683075 PMCID: PMC8535840 DOI: 10.3390/healthcare9101395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022] Open
Abstract
Recent advances in next-generation sequencing and genome medicine have contributed to treatment decisions in patients with cancer. Most advanced gynecological cancers develop resistance to chemotherapy and have a poor prognosis. Therefore, we conducted genomic tests in gynecological tumors to examine the efficacy and clinical feasibility of genotype-matched therapy. Target sequencing was performed in 20 cases of gynecological cancers (cervical cancer, 6; endometrial cancer, 6; and ovarian cancer, 6). Both actionable and druggable genes were identified in 95% (19/20) of the cases. Among them, seven patients (35%) received genotype-matched therapy, which was effective in three patients. Of the three patients, one patient with a PTEN mutation received everolimus, another patient with a TSC2 mutation received everolimus and letrozole, and the patient with a BRIP1 mutation received olaparib. Subsequently, disease control in these three patients lasted for more than half a year. However, all patients relapsed between 9 and 13 months after the initiation of genotype-matched therapy. In this study, the response rate of genotype-matched therapy was 43% (3/7), which may have contributed to improved prognoses. Therefore, genotype-matched therapies may help patients with refractory gynecological cancers achieve better outcomes.
Collapse
|
9
|
Niogret J, Dalens L, Truntzer C, Chevrier S, Favier L, Lagrange A, Coudert B, Fraisse C, Foucher P, Zouak A, Westeel V, Goussot V, Dérangère V, Albuisson J, Arnould L, Boidot R, Kaderbhai CG, Ghiringhelli F. Does large NGS panel analysed using exome tumour sequencing improve the management of advanced non-small-cell lung cancers? Lung Cancer 2021; 161:98-107. [PMID: 34560426 DOI: 10.1016/j.lungcan.2021.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/02/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Non-small-cell lung cancer (NSCLC) is one of the most common and deadly cancers. Several molecular drivers of oncogene addiction are now known to be strong predictive biomarkers for target therapies. Advances in large Next Generation Sequencing (LNGS) have improved the ability to detect potentially targetable mutations. However, the integration of LNGS into clinical management in an individualized manner remains challenging. METHODS In this single-center observational study we included all patients with advanced NSCLC who underwent LNGS. Somatic and germline exome analysis was performed with a restriction on 323 cancer related genes. Variants were classified and Molecular Tumour Board (MTB) made therapeutic propositions. RESULTS We performed LNGS analysis in 281 patients with advanced NSCLC between March 2015 and January 2018. Technical failure occurred in only 3% of cases. Three hundred and fifty-six targetable mutations were detected. At least one targetable mutation was found in 209 patients. For all these patients, the MTB was able to recommend treatment with a targeted agent based on the evaluation of the tumour's genetic profile and treatment history. Twenty-nine patients (13.9%) were subsequently treated with an MTB-recommended targeted therapy. We did not observe any improvement in terms of clinical benefit for these patients. CONCLUSIONS In this case series, we show that including LNGS into routine clinical management was feasible but does not appear to provide clinical benefit in the management of patients with advanced NSCLC.
Collapse
Affiliation(s)
- Julie Niogret
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France
| | - Lorraine Dalens
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France
| | - Caroline Truntzer
- Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France
| | - Sandy Chevrier
- Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Laure Favier
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Aurélie Lagrange
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Bruno Coudert
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Cléa Fraisse
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Pascal Foucher
- Department of Thoracic Oncology, Dijon University Hospital, 14 rue Paul Gaffarel, 21000 Dijon, France
| | - Ayoub Zouak
- Department of Thoracic Oncology, Dijon University Hospital, 14 rue Paul Gaffarel, 21000 Dijon, France
| | - Virginie Westeel
- Department of Pneumology, Besançon University Hospital, 3 Boulevard Alexandre Fleming, 25000 Besançon, France
| | - Vincent Goussot
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Valentin Dérangère
- University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France; Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; UMR INSERM 1231, 7 Boulevard Jeanne d'Arc, 21000 Dijon, France
| | - Juliette Albuisson
- Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Laurent Arnould
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Romain Boidot
- University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France; Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; UMR INSERM 1231, 7 Boulevard Jeanne d'Arc, 21000 Dijon, France
| | - Courèche-Guillaume Kaderbhai
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - François Ghiringhelli
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France; Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France; UMR INSERM 1231, 7 Boulevard Jeanne d'Arc, 21000 Dijon, France.
| |
Collapse
|
10
|
Bitzer M, Spahn S, Babaei S, Horger M, Singer S, Schulze-Osthoff K, Missios P, Gatidis S, Nann D, Mattern S, Scheble V, Nikolaou K, Armeanu-Ebinger S, Schulze M, Schroeder C, Biskup S, Beha J, Claassen M, Ruhm K, Poso A, Malek NP. Targeting extracellular and juxtamembrane FGFR2 mutations in chemotherapy-refractory cholangiocarcinoma. NPJ Precis Oncol 2021; 5:80. [PMID: 34480077 PMCID: PMC8417271 DOI: 10.1038/s41698-021-00220-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/14/2021] [Indexed: 12/17/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) has emerged as a promising candidate for precision medicine, especially in the case of activating FGFR2 gene fusions. In addition to fusions, a considerable fraction of iCCA patients reveals FGFR2 mutations, which might lead to uncontrolled activation of the FGFR2 pathway but are mostly of unknown functional significance. A current challenge for molecular tumor boards (MTB) is to predict the functional consequences of such FGFR2 alterations to guide potential treatment decisions. We report two iCCA patients with extracellular and juxtamembrane FGFR2 mutations. After in silico investigation of the alterations and identification of activated FGFR2 downstream targets in tumor specimens by immunohistochemistry and transcriptome analysis, the MTB recommended treatment with an FGFR-inhibiting tyrosine kinase inhibitor. Both patients developed a rapidly detectable and prolonged partial response to treatment. These two cases suggest an approach to characterize further detected FGFR2 mutations in iCCA to enable patients´ selection for a successful application of the FGFR -inhibiting drugs.
Collapse
Affiliation(s)
- Michael Bitzer
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany. .,Center for Personalized Medicine, Eberhard-Karls University, Tübingen, Germany. .,Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, Tübingen, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Stephan Spahn
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany
| | - Sepideh Babaei
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany
| | - Marius Horger
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls University, Tübingen, Germany
| | - Stephan Singer
- Institute of Pathology and Neuropathology, Eberhard-Karls University, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Molecular Medicine, Interfaculty Institute for Biochemistry, Eberhard-Karls University, Tübingen, Germany
| | - Pavlos Missios
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls University, Tübingen, Germany
| | - Dominik Nann
- Institute of Pathology and Neuropathology, Eberhard-Karls University, Tübingen, Germany
| | - Sven Mattern
- Institute of Pathology and Neuropathology, Eberhard-Karls University, Tübingen, Germany
| | - Veit Scheble
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, Eberhard-Karls University, Tübingen, Germany
| | - Sorin Armeanu-Ebinger
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University, Tübingen, Germany
| | - Martin Schulze
- CeGaT GmbH and Praxis für Humangenetik, Tübingen, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University, Tübingen, Germany
| | - Saskia Biskup
- CeGaT GmbH and Praxis für Humangenetik, Tübingen, Germany
| | - Janina Beha
- Center for Personalized Medicine, Eberhard-Karls University, Tübingen, Germany
| | - Manfred Claassen
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany
| | - Kristina Ruhm
- Center for Personalized Medicine, Eberhard-Karls University, Tübingen, Germany
| | - Antti Poso
- Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nisar P Malek
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany.,Center for Personalized Medicine, Eberhard-Karls University, Tübingen, Germany.,Cluster of Excellence, Image Guided and Functionally Instructed Tumor Therapies, Eberhard-Karls University, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Abstract
BACKGROUND New diagnostic tools in the field of oncology that became available with introduction of the next generation sequencing call for adjustments in the current clinical workflow. To ensure correct interpretation, newly collected data need to be processed and categorized properly. Thus, current experts in oncology need to be trained and new experts from other fields need to be recruited. OBJECTIVES The molecular tumor board was introduced to bring experts from various specialties together. The goal is to discuss and assess complex oncological cases in the context of new molecular diagnostics and give recommendations regarding individualized therapy. RESULTS After the introduction of the molecular tumor board 2 years ago, the number of cases processed within the molecular tumor board has increased steadily. Of these patients, 70% exhibit molecular alterations that are relevant to therapy. Preliminary results indicate positive responses to the applied therapies and clear improvements in the progression-free and overall survival of patients who would have been considered "untreatable" in the classical clinical setting. CONCLUSION The introduction of new molecular diagnostics makes the establishment of advanced clinical structures mandatory. In this regard, the molecular tumor board continues to gain in importance. Preliminary results point towards a significant impact on the therapy of advanced malignancies. The advancements in sequencing and newly established insights into the interpretation of sequencing results will lead to new therapeutic routes. Inevitably, this will make the molecular tumor board indispensable in the future.
Collapse
|
12
|
Melas M, Subbiah S, Saadat S, Rajurkar S, McDonnell KJ. The Community Oncology and Academic Medical Center Alliance in the Age of Precision Medicine: Cancer Genetics and Genomics Considerations. J Clin Med 2020; 9:E2125. [PMID: 32640668 PMCID: PMC7408957 DOI: 10.3390/jcm9072125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Recent public policy, governmental regulatory and economic trends have motivated the establishment and deepening of community health and academic medical center alliances. Accordingly, community oncology practices now deliver a significant portion of their oncology care in association with academic cancer centers. In the age of precision medicine, this alliance has acquired critical importance; novel advances in nucleic acid sequencing, the generation and analysis of immense data sets, the changing clinical landscape of hereditary cancer predisposition and ongoing discovery of novel, targeted therapies challenge community-based oncologists to deliver molecularly-informed health care. The active engagement of community oncology practices with academic partners helps with meeting these challenges; community/academic alliances result in improved cancer patient care and provider efficacy. Here, we review the community oncology and academic medical center alliance. We examine how practitioners may leverage academic center precision medicine-based cancer genetics and genomics programs to advance their patients' needs. We highlight a number of project initiatives at the City of Hope Comprehensive Cancer Center that seek to optimize community oncology and academic cancer center precision medicine interactions.
Collapse
Affiliation(s)
- Marilena Melas
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Shanmuga Subbiah
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Glendora, CA 91741, USA;
| | - Siamak Saadat
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Colton, CA 92324, USA;
| | - Swapnil Rajurkar
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Upland, CA 91786, USA;
| | - Kevin J. McDonnell
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA 91010, USA
- Center for Precision Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|