1
|
Loth AG, Wild PJ. [Individualization and standardization in head and neck pathology]. HNO 2025:10.1007/s00106-025-01627-y. [PMID: 40237827 DOI: 10.1007/s00106-025-01627-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
Individualization and standardization are seemingly contradictory requirements in medicine. In the treatment of head and neck cancer, both terms have a direct influence on diagnostic procedures, which are usually carried out in pathology institutes. The current article examines the conflicting requirements arising from various technical analyses, regulatory requirements, structural changes due to digitalization, and the advent of personalized medicine. On the one hand, the goal is to promote interdisciplinary exchange by understanding the challenges and, on the other, to provide the otorhinolaryngologist with a practical understanding of the common and current pathological diagnostic tests. Using pathology as an example, it can be shown that standardization of procedures ultimately serves to improve individualized treatment. At the same time, however, the following challenges are also apparent: despite comprehensive regulations and a laboratory environment with digital support, standardization is very time consuming and costly. If similar standardization approaches are to be implemented in an operative environment such as, e.g., ENT surgery, the effort involved can be expected to be equivalent or higher due to the human factor.
Collapse
Affiliation(s)
- Andreas G Loth
- Universitätsklinikum Frankfurt, Klinik für Hals‑, Nasen- und Ohrenheilkunde, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60450, Frankfurt am Main, Deutschland.
| | - Peter J Wild
- Universitätsklinikum Frankfurt, Dr. Senckenbergisches Institut für Pathologie und Humangenetik, Goethe-Universität Frankfurt, Frankfurt am Main, Deutschland
| |
Collapse
|
2
|
Bezerra OCL, Rodger M, Munsch G, Kovacs MJ, Le Gal G, Morange PE, Trégouët DA, Greenwood CMT, Gagnon F. Sex-specific DNA methylation marks associated with sex-biased risk of recurrence in unprovoked venous thromboembolism. J Thromb Haemost 2025; 23:1379-1392. [PMID: 39848545 DOI: 10.1016/j.jtha.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/01/2024] [Accepted: 01/06/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Whether to stop oral anticoagulants after a first unprovoked venous thromboembolism (VTE) is challenging, partially due to an intriguingly higher risk of VTE recurrence (rVTE) in men after therapy discontinuation. DNA methylation (DNAm) differences between men and women might underlie this sex-biased rVTE risk difference. OBJECTIVES To investigate sex-specific associations between DNAm at cytosine-phosphate-guanine (CpG) sites and rVTE. METHODS In 417 unprovoked VTE patients, including 101 experiencing recurrences over a 5-year follow-up (REcurrent VEnous thromboembolism Risk Stratification Evaluation [REVERSE] I), we analyzed blood DNAm using the Illumina EPIC array and performed a sex-stratified epigenome-wide association study. We further examined 181 major provoked VTE patients, including 36 recurrences over a 14-year follow-up (the MARseille THrombosis Association [MARTHA]), to investigate whether DNAm is a risk factor for rVTE after anticoagulation therapy. RESULTS Hypomethylated CpGs at genes TBC1D22B-cg01060850 and ZHX2-cg07808424 in men and DIP2B-ch.12.1038646R and DENND3-cg03401656 in women were associated with rVTE at genome-wide level (P < 7x10-8). Though not statistically significant, DENND3-cg03401656 had the same direction of effect in MARTHA women. Sensitivity analysis confirmed the robustness of the estimates, including potential confounders, adaptations of the Cox model, non-Europeans, and proximal methylation quantitative trait loci in the association. The associated CpGs were situated at genes for membrane trafficking, corroborating the participation of Rab regulatory proteins in rVTE and transcription factors. CONCLUSION We identified DNAm marks as potential risk factors for sex-biased recurrence in unprovoked VTE. Further replication and experimental validation could refine our understanding of the regulation of the identified DNAm sites and help optimize personalized decision-making for long-term anticoagulation after a first VTE.
Collapse
Affiliation(s)
- Ohanna C L Bezerra
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada. https://twitter.com/ohannaclb
| | - Marc Rodger
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Gaëlle Munsch
- Bordeaux Population Health Research Center, Institut national de la santé et de la recherche médicale (INSERM), Unité Mixte de Recherche (UMR) 1219, F-33000, University of Bordeaux, Bordeaux, France
| | - Michael J Kovacs
- Department of Medicine, Western University, London, Ontario, Canada
| | - Grégoire Le Gal
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, Institut national de la santé et de la recherche médicale (INSERM), Unité Mixte de Recherche (UMR) 1219, F-33000, University of Bordeaux, Bordeaux, France
| | - Celia M T Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - France Gagnon
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada; Office of the Vice-Principal of Research and Innovation, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| |
Collapse
|
3
|
Inoue S, Takami H, Tanaka S, Nomura M, Takayanagi S, Saito Y, Kikuta S, Kondo K, Matsuura R, Ikemura M, Yamazawa S, Matsutani M, Nishikawa R, Matsushita Y, Ichimura K, Saito N. Nasal immature teratoma in an elderly patient: Clinicopathological and epigenetic analogies with central nervous system counterparts, alongside genomic divergences. Neuropathology 2025; 45:100-108. [PMID: 39359021 PMCID: PMC11962582 DOI: 10.1111/neup.13008] [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: 04/16/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
Germ cell tumors (GCTs) are categorized as gonadal or extra-gonadal, based on the origin. Extra-gonadal GCTs predominantly manifest within the central nervous system (CNS), mediastinum, retroperitoneum, and sacrococcygeal region. These malignancies are most frequently diagnosed in the pediatric, adolescent, and young adult demographics. Incidences of GCT within the nasal cavity are notably scarce, with only six cases documented. This report details the case of a 70-year-old man who presented with a left nasal mass ultimately diagnosed as immature teratoma. A remarkable aspect of this case was the detection of SMARCA4 (BRG1) loss through immunohistochemical analysis. In addition, methylation profiling aligned this case with CNS GCTs, specifically those classified as non-germinomatous GCTs. This molecular characterization informed a tailored therapeutic strategy incorporating carboplatin and etoposide, alongside localized irradiation. This individualized treatment regimen achieved favorable outcomes, with the patient remaining recurrence free for over three years. This highlights the need for precise therapeutic approaches in the management of extragonadal GCTs, particularly those arising in atypical anatomical locations. The present case accentuates the significance of thorough diagnostic evaluations and customized treatment plans for rare GCT presentations. Further empirical and clinical investigations are warranted to enhance our understanding of and refine therapeutic protocols for such exceptional cases.
Collapse
Affiliation(s)
- Shintaro Inoue
- Department of NeurosurgeryThe University of Tokyo HospitalTokyoJapan
| | - Hirokazu Takami
- Department of NeurosurgeryThe University of Tokyo HospitalTokyoJapan
| | - Shota Tanaka
- Department of NeurosurgeryThe University of Tokyo HospitalTokyoJapan
| | - Masashi Nomura
- Department of NeurosurgeryThe University of Tokyo HospitalTokyoJapan
| | | | - Yuki Saito
- Department Otolaryngology, Head and Neck SurgeryThe University of Tokyo HospitalTokyoJapan
| | - Shu Kikuta
- Department Otolaryngology, Head and Neck SurgeryThe University of Tokyo HospitalTokyoJapan
| | - Kenji Kondo
- Department Otolaryngology, Head and Neck SurgeryThe University of Tokyo HospitalTokyoJapan
| | - Reiko Matsuura
- Department of NeurosurgeryThe University of Tokyo HospitalTokyoJapan
| | - Masako Ikemura
- Department PathologyThe University of Tokyo HospitalTokyoJapan
| | - Sho Yamazawa
- Department PathologyThe University of Tokyo HospitalTokyoJapan
| | - Masao Matsutani
- Department of Neuro‐Oncology/NeurosurgerySaitama Medical University International Medical CenterSaitamaJapan
| | - Ryo Nishikawa
- Department of Neuro‐Oncology/NeurosurgerySaitama Medical University International Medical CenterSaitamaJapan
| | - Yuko Matsushita
- Department of Brain Disease Translational ResearchJuntendo University Faculty of MedicineTokyoJapan
| | - Koichi Ichimura
- Department of Brain Disease Translational ResearchJuntendo University Faculty of MedicineTokyoJapan
| | - Nobuhito Saito
- Department of NeurosurgeryThe University of Tokyo HospitalTokyoJapan
| |
Collapse
|
4
|
Benfatto S, Sill M, Jones DTW, Pfister SM, Sahm F, von Deimling A, Capper D, Hovestadt V. Explainable artificial intelligence of DNA methylation-based brain tumor diagnostics. Nat Commun 2025; 16:1787. [PMID: 39979307 PMCID: PMC11842776 DOI: 10.1038/s41467-025-57078-0] [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: 01/08/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
We have recently developed a machine learning classifier that enables fast, accurate, and affordable classification of brain tumors based on genome-wide DNA methylation profiles that is widely employed in the clinic. Neuro-oncology research would benefit greatly from understanding the underlying artificial intelligence decision process, which currently remains unclear. Here, we describe an interpretable framework to explain the classifier's decisions. We show that functional genomic regions of various sizes are predominantly employed to distinguish between different tumor classes, ranging from enhancers and CpG islands to large-scale heterochromatic domains. We detect a high degree of genomic redundancy, with many genes distinguishing individual tumor classes, explaining the robustness of the classifier and revealing potential targets for further therapeutic investigation. We anticipate that our resource will build up trust in machine learning in clinical settings, foster biomarker discovery and development of compact point-of-care assays, and enable further epigenome research of brain tumors. Our interpretable framework is accessible to the research community via an interactive web application ( https://hovestadtlab.shinyapps.io/shinyMNP/ ).
Collapse
Affiliation(s)
- Salvatore Benfatto
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martin Sill
- Division of Pediatric Neurooncology, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - David T W Jones
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - David Capper
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Hovestadt
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
5
|
Ghanem A, Finlay JB, Jang DW, Goldstein BJ, Abi Hachem R. Recent developments in olfactory neuroblastoma research. Curr Opin Otolaryngol Head Neck Surg 2025; 33:50-55. [PMID: 39607836 DOI: 10.1097/moo.0000000000001027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
PURPOSE OF REVIEW Olfactory neuroblastoma (ONB) is a rare malignancy originating from olfactory neuroepithelial cells. Given its uncommon nature and complex clinical presentation, this comprehensive review highlights recent findings and treatment approaches for advancing clinical practice and research. RECENT FINDINGS Recent literature emphasizes significant advancements in the genomic profiling and molecular classification of ONB. Emerging targeted therapies include somatostatin analogs and programmed death-ligand 1 (PD-L1) inhibitors. In addition, the development of genetically engineered mouse models has provided valuable platforms for testing new treatment strategies, revealing similarities between ONB and small cell lung cancer, which may inform future therapeutic approaches. SUMMARY These findings have profound implications on clinical practice. Improved diagnostic accuracy through advanced imaging and genomic profiling in addition to identifying specific mutations for targeted therapy can lead to personalized treatments of patients with ONB. Developments in genetically engineered mouse models and multiinstitutional collaborative efforts are vital for advancing research and standardizing molecular testing. The integration of advanced imaging techniques, genomic profiling, and targeted therapies holds promise for improving patient outcomes and understanding this rare malignancy.
Collapse
Affiliation(s)
- Anthony Ghanem
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine
| | - John B Finlay
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine
- Duke University, School of Medicine, Durham, North Carolina, USA
| | - David W Jang
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine
| | - Bradley J Goldstein
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine
| | - Ralph Abi Hachem
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine
| |
Collapse
|
6
|
Walker A, Fang CS, Schroff C, Serrano J, Vasudevaraja V, Yang Y, Belakhoua S, Faustin A, William CM, Zagzag D, Chiang S, Acosta AM, Movahed-Ezazi M, Park K, Moreira AL, Darvishian F, Galbraith K, Snuderl M. Deep learning-based classifier for carcinoma of unknown primary using methylation quantitative trait loci. J Neuropathol Exp Neurol 2025; 84:147-154. [PMID: 39607989 PMCID: PMC11747144 DOI: 10.1093/jnen/nlae123] [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] [Indexed: 11/30/2024] Open
Abstract
Cancer of unknown primary (CUP) constitutes between 2% and 5% of human malignancies and is among the most common causes of cancer death in the United States. Brain metastases are often the first clinical presentation of CUP; despite extensive pathological and imaging studies, 20%-45% of CUP are never assigned a primary site. DNA methylation array profiling is a reliable method for tumor classification but tumor-type-specific classifier development requires many reference samples. This is difficult to accomplish for CUP as many cases are never assigned a specific diagnosis. Recent studies identified subsets of methylation quantitative trait loci (mQTLs) unique to specific organs, which could help increase classifier accuracy while requiring fewer samples. We performed a retrospective genome-wide methylation analysis of 759 carcinoma samples from formalin-fixed paraffin-embedded tissue samples using Illumina EPIC array. Utilizing mQTL specific for breast, lung, ovarian/gynecologic, colon, kidney, or testis (BLOCKT) (185k total probes), we developed a deep learning-based methylation classifier that achieved 93.12% average accuracy and 93.04% average F1-score across a 10-fold validation for BLOCKT organs. Our findings indicate that our organ-based DNA methylation classifier can assist pathologists in identifying the site of origin, providing oncologists insight on a diagnosis to administer appropriate therapy, improving patient outcomes.
Collapse
Affiliation(s)
- Adam Walker
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Camila S Fang
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | - Chanel Schroff
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Jonathan Serrano
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Varshini Vasudevaraja
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Yiying Yang
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Sarra Belakhoua
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Arline Faustin
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Christopher M William
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - David Zagzag
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Sarah Chiang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Misha Movahed-Ezazi
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Kyung Park
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Andre L Moreira
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Farbod Darvishian
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Kristyn Galbraith
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health and NYU Grossman School of Medicine, New York, NY, United States
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| |
Collapse
|
7
|
Geisenberger C, Chimal E, Jurmeister P, Klauschen F. A cost-effective and scalable approach for DNA extraction from FFPE tissues. Biol Methods Protoc 2025; 10:bpaf003. [PMID: 39995602 PMCID: PMC11849955 DOI: 10.1093/biomethods/bpaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 02/26/2025] Open
Abstract
Genomic profiling of cancer plays an increasingly vital role for diagnosis and therapy planning. In addition, research of novel diagnostic applications such as DNA methylation profiling requires large training and validation cohorts. Currently, most diagnostic cases processed in pathology departments are stored as formalin-fixed and paraffin embedded tissue blocks (FFPE). Consequently, there is a growing demand for high-throughput extraction of nucleic acids from FFPE tissue samples. While proprietary kits are available, they are expensive and offer little flexibility. Here, we present ht-HiTE, a high-throughput implementation of a recently published and highly efficient DNA extraction protocol. This approach enables manual and automated processing of 96-well plates with a liquid handler, offers two options for purification and utilizes off-the-shelf reagents. Finally, we show that NGS and DNA methylation microarray data obtained from DNA processed with ht-HiTE are of equivalent quality as compared to a manual, kit-based approach.
Collapse
Affiliation(s)
- Christoph Geisenberger
- Institute of Pathology, Ludwig-Maximilians-Universität München, Thalkirchnerstr. 36, Munich, 80337, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital Munich, Pettenkoferstr. 8a, Munich, 80336, Germany
| | - Edgar Chimal
- Institute of Pathology, Ludwig-Maximilians-Universität München, Thalkirchnerstr. 36, Munich, 80337, Germany
| | - Philipp Jurmeister
- Institute of Pathology, Ludwig-Maximilians-Universität München, Thalkirchnerstr. 36, Munich, 80337, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital Munich, Pettenkoferstr. 8a, Munich, 80336, Germany
- BIFOLD—Berlin Institute for the Foundations of Learning and Data, Einsteinufer 17, Berlin, 10587, Germany
| | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-Universität München, Thalkirchnerstr. 36, Munich, 80337, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital Munich, Pettenkoferstr. 8a, Munich, 80336, Germany
- BIFOLD—Berlin Institute for the Foundations of Learning and Data, Einsteinufer 17, Berlin, 10587, Germany
| |
Collapse
|
8
|
Aldape K, Capper D, von Deimling A, Giannini C, Gilbert MR, Hawkins C, Hench J, Jacques TS, Jones D, Louis DN, Mueller S, Orr BA, Nasrallah M, Pfister SM, Sahm F, Snuderl M, Solomon D, Varlet P, Wesseling P. cIMPACT-NOW update 9: Recommendations on utilization of genome-wide DNA methylation profiling for central nervous system tumor diagnostics. Neurooncol Adv 2025; 7:vdae228. [PMID: 39902391 PMCID: PMC11788596 DOI: 10.1093/noajnl/vdae228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025] Open
Abstract
Genome-wide DNA methylation signatures correlate with and distinguish central nervous system (CNS) tumor types. Since the publication of the initial CNS tumor DNA methylation classifier in 2018, this platform has been increasingly used as a diagnostic tool for CNS tumors, with multiple studies showing the value and utility of DNA methylation-based classification of CNS tumors. A Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT-NOW) Working Group was therefore convened to describe the current state of the field and to provide advice based on lessons learned to date. Here, we provide recommendations for the use of DNA methylation-based classification in CNS tumor diagnostics, emphasizing the attributes and limitations of the modality. We emphasize that the methylation classifier is one diagnostic tool to be used alongside previously established diagnostic tools in a fully integrated fashion. In addition, we provide examples of the inclusion of DNA methylation data within the layered diagnostic reporting format endorsed by the World Health Organization (WHO) and the International Collaboration on Cancer Reporting. We emphasize the need for backward compatibility of future platforms to enable accumulated data to be compatible with new versions of the array. Finally, we outline the specific connections between methylation classes and CNS WHO tumor types to aid in the interpretation of classifier results. It is hoped that this update will assist the neuro-oncology community in the interpretation of DNA methylation classifier results to facilitate the accurate diagnosis of CNS tumors and thereby help guide patient management.
Collapse
Affiliation(s)
- Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MarylandUSA
| | - David Capper
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas von Deimling
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Caterina Giannini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum, Bologna, Italy
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jürgen Hench
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Basel, Switzerland
| | - Thomas S Jacques
- Department of Histopathology, Great Ormond Street Hospital for Children, London, UK
- Paediatric Neuropathology, University College London, UCL GOS Institute of Child Health, London, UK
| | - David Jones
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - David N Louis
- Department of Pathology, Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston Massachusetts, USA
| | - Sabine Mueller
- Department of Pediatric, University of Zurich, Zürich, Switzerland
- Department of Neurology, Neurosurgery, and Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - MacLean Nasrallah
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stefan M Pfister
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Hopp Children´s Cancer Center Heidelberg (KiTZ), Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Felix Sahm
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York, USA
| | - David Solomon
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Pascale Varlet
- Department of Neuropathology, GHU Paris - Psychiatry and Neuroscience, Sainte-Anne Hospital, Paris, France
| | - Pieter Wesseling
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, Amsterdam University Medical Centers/VU University, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Panthong W, Pientong C, Nukpook T, Roytrakul S, Yingchutrakul Y, Teeramatwanich W, Aromseree S, Ekalaksananan T. Integrated Proteomics and Machine Learning Approach Reveals PYCR1 as a Novel Biomarker to Predict Prognosis of Sinonasal Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:13234. [PMID: 39768999 PMCID: PMC11675701 DOI: 10.3390/ijms252413234] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Sinonasal squamous cell carcinoma (SNSCC) is a rare tumor with a high 5-year mortality rate. However, proteomic technologies have not yet been utilized to identify SNSCC-associated proteins, which could be used as biomarkers. In this study, we aimed to discover a biomarker to predict SNSCC patients using proteomic analysis integrated with machine learning models. Support vector machine (SVM), logistic regression (LR), random forest (RF), and gradient boost (GB) classifiers were developed to predict SNSCC based on proteomic profiles of SNSCC compared with nasal polyps (NP) as control. Seventeen feature proteins were found in all models, indicating possible biomarkers for SNSCC. Analysis of gene expression across multiple cancer types and their associations with cancer stage and patient survival in the TCGA-HNSC dataset identified a PYCR1 and MYO1B gene that could be a potential tumor-associated marker. The expression of PYCR1 was confirmed by RT-qPCR in SNSCC tissues, and its high expression was associated with poor overall survival, indicating PYCR1 as a potential tumor-associated biomarker to predict the prognosis of SNSCC.
Collapse
Affiliation(s)
- Watcharapong Panthong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Thawaree Nukpook
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Khlong Luang, Pathum Thani 12120, Thailand; (S.R.); (Y.Y.)
| | - Yodying Yingchutrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Khlong Luang, Pathum Thani 12120, Thailand; (S.R.); (Y.Y.)
| | - Watchareporn Teeramatwanich
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand;
| | - Sirinart Aromseree
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand; (W.P.); (T.N.); (S.A.)
- HPV&EBV and Carcinogenesis (HEC) Research Group, Faculty of Medicine, Khon Kaen University, Mueang Khon Kaen, Khon Kaen 40002, Thailand
| |
Collapse
|
10
|
Woods R, Scholfield D, Axiotakis L, Fitzgerald C, Adilbay D, Cracchiolo J, Patel S, Shah J, Dunn L, Pfister D, Lee N, Dogan S, Ganly I, Cohen M. Outcomes of SMARCB1-deficient sinonasal carcinoma: Largest single-center cross-sectional study. Head Neck 2024; 46:3076-3084. [PMID: 39044555 DOI: 10.1002/hed.27859] [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: 04/15/2024] [Revised: 06/12/2024] [Accepted: 06/23/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND We evaluate outcomes of SMARCB1-deficient sinonasal carcinomas in the largest single-institution study. METHODS Retrospective cross-sectional study of patients with SMARCB1-deficient sinonasal carcinoma between 1998 and 2024. Disease-specific survival (DSS) and recurrence-free probability (RFP) at 1 and 5 years were measured by Kaplan-Meier method. RESULTS There were 47 patients with a median age of 53. Initial pathological diagnosis was altered in 33%. Twelve (34%) patients received neoadjuvant chemotherapy, with one partial response. Curative surgical approach was undertaken in 73%. Definitive chemoradiation was administered in 20%. DSS at 1 and 5 years was 93% and 45%, respectively. RFP at 1 and 5 years was 73% and 33%, respectively. On multivariate analysis, cranial nerve involvement (p = 0.01 for DSS) remained significantly worse for DSS and overall survival. CONCLUSIONS SMARCB1-deficient tumors had limited response to neoadjuvant chemotherapy. Cranial nerve involvement was associated with worse prognosis. Optimal treatment is unclear. Surgery should be offered to patients with resectable disease.
Collapse
Affiliation(s)
- Robbie Woods
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Daniel Scholfield
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lucas Axiotakis
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Conall Fitzgerald
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dauren Adilbay
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jennifer Cracchiolo
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Snehal Patel
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jatin Shah
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lara Dunn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David Pfister
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nancy Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Snjezana Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ian Ganly
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Marc Cohen
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
11
|
Jurmeister P, Leitheiser M, Arnold A, Capilla EP, Mochmann LH, Zhdanovic Y, Schleich K, Jung N, Chimal EC, Jung A, Kumbrink J, Harter P, Prenißl N, Elezkurtaj S, Brcic L, Deigendesch N, Frank S, Hench J, Försch S, Breimer G, van Engen van Grunsven I, Lassche G, van Herpen C, Zhou F, Snuderl M, Agaimy A, Müller KR, von Deimling A, Capper D, Klauschen F, Ihrler S. DNA Methylation Profiling of Salivary Gland Tumors Supports and Expands Conventional Classification. Mod Pathol 2024; 37:100625. [PMID: 39332710 DOI: 10.1016/j.modpat.2024.100625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/05/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024]
Abstract
Tumors of the major and minor salivary glands histologically encompass a diverse and partly overlapping spectrum of frequent diagnostically challenging neoplasms. Despite recent advances in molecular testing and the identification of tumor-specific mutations or gene fusions, there is an unmet need to identify additional diagnostic biomarkers for entities lacking specific alterations. In this study, we collected a comprehensive cohort of 363 cases encompassing 20 different salivary gland tumor entities and explored the potential of DNA methylation to classify these tumors. We were able to show that most entities show specific epigenetic signatures and present a machine learning algorithm that achieved a mean balanced accuracy of 0.991. Of note, we showed that cribriform adenocarcinoma is epigenetically distinct from classical polymorphous adenocarcinoma, which could support risk stratification of these tumors. Myoepithelioma and pleomorphic adenoma form a uniform epigenetic class, supporting the theory of a single entity with a broad but continuous morphologic spectrum. Furthermore, we identified a histomorphologically heterogeneous but epigenetically distinct class that could represent a novel tumor entity. In conclusion, our study provides a comprehensive resource of the DNA methylation landscape of salivary gland tumors. Our data provide novel insight into disputed entities and show the potential of DNA methylation to identify new tumor classes. Furthermore, in future, our machine learning classifier could support the histopathologic diagnosis of salivary gland tumors.
Collapse
Affiliation(s)
- Philipp Jurmeister
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | | | - Alexander Arnold
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Emma Payá Capilla
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Liliana H Mochmann
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Yauheniya Zhdanovic
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Konstanze Schleich
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nina Jung
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Andreas Jung
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Patrick Harter
- German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Neuropathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Niklas Prenißl
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sefer Elezkurtaj
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Nikolaus Deigendesch
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stephan Frank
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jürgen Hench
- Department of Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastian Försch
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gerben Breimer
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Gerben Lassche
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fang Zhou
- Department of Pathology, New York University Langone Health, School of Medicine, New York, New York
| | - Matija Snuderl
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nurnberg, University Hospital Erlangen, Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nurnberg, University Hospital Erlangen, Erlangen, Germany
| | - Klaus-Robert Müller
- Machine Learning Group, Department of Software Engineering and Theoretical Computer Science, Technical University of Berlin, Berlin, Germany; Department of Artificial Intelligence, Korea University, Seoul, South Korea; Max Planck Institute for Informatics, Saarbrucken, Germany; BIFOLD-Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Capper
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany; BIFOLD-Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | |
Collapse
|
12
|
Mikula MW, Rooper LM. Olfactory Neuroblastoma and Olfactory Carcinoma: A Practical Review. Surg Pathol Clin 2024; 17:637-652. [PMID: 39489554 DOI: 10.1016/j.path.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
In the sinonasal tract, two tumor types are defined by neuroectodermal differentiation. Olfactory neuroblastoma (ONB), the traditional member of this category, is a keratin-negative neuroectodermal neoplasm that has lobulated architecture and variable neurofibrillary matrix. Olfactory carcinoma (OC), a newly-recognized diagnosis in the sinonasal tract, has keratin-positive neuroectodermal elements frequently intermixed with well-formed glands. These two neuroectodermal entities can show substantial overlap with other sinonasal small round blue cell tumors that express neuroendocrine markers. This review provides a practical overview of the key clinical and diagnostic features of ONB and OC and their differential diagnosis.
Collapse
Affiliation(s)
- Michael W Mikula
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa M Rooper
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
13
|
Xue E, Bracken-Clarke D, Krause H, Adeyelu T, Evans MG, Akbulut D, Quezado M, Gandhi N, Farrell A, Soares HP, Lou E, Phan M, Patel R, Vanderwalde AM, Elliott A, Steuer CE, Saba NF, Lubin DJ, London NR, Gulley JL, Floudas CS. Characterization of Somatostatin Receptor 2 Gene Expression and Immune Landscape in Sinonasal Malignancies. Cancers (Basel) 2024; 16:3931. [PMID: 39682120 DOI: 10.3390/cancers16233931] [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: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Olfactory neuroblastoma (ONB), sinonasal undifferentiated carcinoma (SNUC), and sinonasal neuroendocrine carcinoma (SNEC) are rare malignancies arising from the sinonasal tract with limited therapeutic options. The expression of the somatostatin receptor 2 gene (SSTR2), which is expressed in other neuroendocrine neoplasms and is therapeutically actionable, has been reported in these tumors. Here, we analyzed SSTR2 gene expression and its associations with genomic features, established biomarkers predicting of immune response, and the tumor immune microenvironment in a cohort of ONB, SNUC, and SNEC tumor samples (26, 13, and 8 samples, respectively) from a real-world database. SSTR2 gene expression was high in neural-type ONB and low in basal-type ONB and in most of the SNUC and SNEC cases; there was no difference in expression between primary and metastatic tumors. The T cell-inflamed (TCI) score analysis classified 38.5% of SNUC cases as T cell-inflamed compared to only 3.9% of ONB and 0% of SNEC cases; 26.9% of ONB cases were classified as intermediate TCI; and SNEC had the lowest relative immune cell infiltration by deconvolution. In high SSTR2-expressing ONB, there was a higher proportion of infiltrating of Natural Killer cells and dendritic cells by deconvolution. Additionally, high SSTR2-expressing ONB was enriched for proliferation pathways, including E2F and Myc targets and G2M checkpoints. In conclusion, our findings delineate significant differences between these three types of sinonasal malignancies that were examined. In ONB, relative to SNUC and SNEC, the SSTR2 expression profile, combined with its immune profiles, indicates potential novel therapeutic strategies and combinations for this unmet clinical need. Conversely, the inflammatory microenvironment of SNUC may be targetable using immuno-oncologic therapies.
Collapse
Affiliation(s)
- Elisabetta Xue
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | - Dara Bracken-Clarke
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | | | | | | | - Dilara Akbulut
- Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | | | | | - Heloisa P Soares
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Emil Lou
- Department of Medicine, Division of Hematology Oncology and Transplant, University of Minnesota, Minneapolis, MN 55414, USA
| | - Minh Phan
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Rusha Patel
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
| | | | | | - Conor E Steuer
- Department of Hematology-Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA
| | - Nabil F Saba
- Department of Hematology-Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA
| | - Daniel J Lubin
- Department of Hematology-Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30308, USA
| | - Nyall R London
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Sinonasal and Skull Base Tumor Program, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - James L Gulley
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| | - Charalampos S Floudas
- Center for Immuno-Oncology, National Cancer Institute, National Institute of Health, Bethesda, MD 20894, USA
| |
Collapse
|
14
|
Calina TG, Perez E, Grafenhorst E, Benhamida J, Schallenberg S, Popescu A, Koch I, Janik T, Chen B, Ihlow J, Roessler S, Goeppert B, Sinn B, Bahra M, Calin GA, Taube ET, Pelzer U, Neumann CCM, Horst D, Knutsen E, Capper D, Dragomir MP. DNA methylation classifier to diagnose pancreatic ductal adenocarcinoma metastases from different anatomical sites. Clin Epigenetics 2024; 16:156. [PMID: 39523345 PMCID: PMC11550539 DOI: 10.1186/s13148-024-01768-x] [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: 03/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND We have recently constructed a DNA methylation classifier that can discriminate between pancreatic ductal adenocarcinoma (PAAD) liver metastasis and intrahepatic cholangiocarcinoma (iCCA) with high accuracy (PAAD-iCCA-Classifier). PAAD is one of the leading causes of cancer of unknown primary and diagnosis is based on exclusion of other malignancies. Therefore, our focus was to investigate whether the PAAD-iCCA-Classifier can be used to diagnose PAAD metastases from other sites. METHODS For this scope, the anomaly detection filter of the initial classifier was expanded by 8 additional mimicker carcinomas, amounting to a total of 10 carcinomas in the negative class. We validated the updated version of the classifier on a validation set, which consisted of a biological cohort (n = 3579) and a technical one (n = 15). We then assessed the performance of the classifier on a test set, which included a positive control cohort of 16 PAAD metastases from various sites and a cohort of 124 negative control samples consisting of 96 breast cancer metastases from 18 anatomical sites and 28 carcinoma metastases to the brain. RESULTS The updated PAAD-iCCA-Classifier achieved 98.21% accuracy on the biological validation samples, and on the technical validation ones it reached 100%. The classifier also correctly identified 15/16 (93.75%) metastases of the positive control as PAAD, and on the negative control, it correctly classified 122/124 samples (98.39%) for a 97.85% overall accuracy on the test set. We used this DNA methylation dataset to explore the organotropism of PAAD metastases and observed that PAAD liver metastases are distinct from PAAD peritoneal carcinomatosis and primary PAAD, and are characterized by specific copy number alterations and hypomethylation of enhancers involved in epithelial-mesenchymal-transition. CONCLUSIONS The updated PAAD-iCCA-Classifier (available at https://classifier.tgc-research.de/ ) can accurately classify PAAD samples from various metastatic sites and it can serve as a diagnostic aid.
Collapse
Affiliation(s)
- Teodor G Calina
- TGC Ventures UG, Berlin, Germany
- Faculty of Physics, Babeș-Bolyai University, Cluj-Napoca, Romania
| | - Eilís Perez
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin School of Integrative Oncology (BSIO), Charite - Universitätsmedizin Berlin (CVK), Berlin, Germany
| | - Elena Grafenhorst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jamal Benhamida
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Ines Koch
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Tobias Janik
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - BaoQing Chen
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China
- Guangdong Esophageal Cancer Research Institute, Guangzhou, Guangdong, People's Republic of China
| | - Jana Ihlow
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Benjamin Goeppert
- Institute of Pathology and Neuropathology, Hospital RKH Kliniken Ludwigsburg, Ludwigsburg, Germany
- Institute of Tissue Medicine and Pathology (ITMP), University Bern, Bern, Switzerland
| | - Bruno Sinn
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus Bahra
- Department of Surgical Oncology and Robotics, Krankenhaus Waldfriede, Lehrkrankenhaus der Charité, Berlin, Germany
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The RNA Interference and Non-Coding RNA Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eliane T Taube
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Uwe Pelzer
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christopher C M Neumann
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT The Artic University of Norway, Tromsø, Norway
| | - David Capper
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mihnea P Dragomir
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
15
|
Gubbiotti MA, LiVolsi V, Montone KT. Update on Sinonasal Tract Malignancies: Advances in Diagnostic Modalities. Arch Pathol Lab Med 2024; 148:1082-1091. [PMID: 36920001 DOI: 10.5858/arpa.2022-0447-ra] [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: 12/20/2022] [Indexed: 03/16/2023]
Abstract
CONTEXT.— Sinonasal tract malignancies are rare cancers with frequent morphologic overlap. Given the similar histologic profiles seen in many of these entities, they often present a diagnostic challenge to the practicing pathologist. OBJECTIVE.— To provide a streamlined algorithm using histologic clues, immunohistochemical profiles, and molecular assays to aid in diagnosis of these lesions. DATA SOURCES.— Sources were the World Health Organization Tumor Classification, literature review, and institutional experience. CONCLUSIONS.— Although many sinonasal tract malignancies show similar histology, distinct immunohistochemical and molecular profiles can help parse out differences, thereby facilitating diagnosis for the pathologist.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- From the Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia. Gubbiotti is now located at the Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Virginia LiVolsi
- From the Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia. Gubbiotti is now located at the Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Kathleen T Montone
- From the Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia. Gubbiotti is now located at the Department of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
16
|
Jia Y, Liu Y, Yang H, Yao F. Adenoid cystic carcinoma: insights from molecular characterization and therapeutic advances. MedComm (Beijing) 2024; 5:e734. [PMID: 39263605 PMCID: PMC11387731 DOI: 10.1002/mco2.734] [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: 03/06/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
Adenoid cystic carcinoma (ACC) is a malignant tumor primarily originating from the salivary glands, capable of affecting multiple organs. Although ACC typically exhibits slow growth, it is notorious for its propensity for neural invasion, local recurrence, and distant metastasis, making it a particularly challenging cancer to treat. The complexity of ACC's histological and molecular features poses significant challenges to current treatment modalities, which often show limited effectiveness. Recent advancements in single-cell RNA-sequencing (scRNA-seq) have begun to unravel unprecedented insights into the heterogeneity and subpopulation diversity within ACC, revealing distinct cellular phenotypes and origins. This review delves into the intricate pathological and molecular characteristics of ACC, focusing on recent therapeutic advancements. We particularly emphasize the insights gained from scRNA-seq studies that shed light on the cellular landscape of ACC, underscoring its heterogeneity and pathobiology. Moreover, by integrating analyses from public databases, this review proposes novel perspectives for advancing treatment strategies in ACC. This review contributes to the academic understanding of ACC by proposing novel therapeutic approaches informed by cutting-edge molecular insights, paving the way for more effective, personalized therapeutic approaches for this challenging malignancy.
Collapse
Affiliation(s)
- Yunxuan Jia
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yupeng Liu
- Department of Thoracic Surgery Tumor Hospital Affiliated to Nantong University Nantong Tumor Hospital Nantong China
| | - Haitang Yang
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Feng Yao
- Department of Thoracic Surgery Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
17
|
Carosi F, Broseghini E, Fabbri L, Corradi G, Gili R, Forte V, Roncarati R, Filippini DM, Ferracin M. Targeting Isocitrate Dehydrogenase (IDH) in Solid Tumors: Current Evidence and Future Perspectives. Cancers (Basel) 2024; 16:2752. [PMID: 39123479 PMCID: PMC11311780 DOI: 10.3390/cancers16152752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
The isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) enzymes are involved in key metabolic processes in human cells, regulating differentiation, proliferation, and oxidative damage response. IDH mutations have been associated with tumor development and progression in various solid tumors such as glioma, cholangiocarcinoma, chondrosarcoma, and other tumor types and have become crucial markers in molecular classification and prognostic assessment. The intratumoral and serum levels of D-2-hydroxyglutarate (D-2-HG) could serve as diagnostic biomarkers for identifying IDH mutant (IDHmut) tumors. As a result, an increasing number of clinical trials are evaluating targeted treatments for IDH1/IDH2 mutations. Recent studies have shown that the focus of these new therapeutic strategies is not only the neomorphic activity of the IDHmut enzymes but also the epigenetic shift induced by IDH mutations and the potential role of combination treatments. Here, we provide an overview of the current knowledge about IDH mutations in solid tumors, with a particular focus on available IDH-targeted treatments and emerging results from clinical trials aiming to explore IDHmut tumor-specific features and to identify the clinical benefit of IDH-targeted therapies and their combination strategies. An insight into future perspectives and the emerging roles of circulating biomarkers and radiomic features is also included.
Collapse
Affiliation(s)
- Francesca Carosi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
| | | | - Laura Fabbri
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
| | - Giacomo Corradi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
| | - Riccardo Gili
- Medical Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Valentina Forte
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Roberta Roncarati
- Istituto di Genetica Molecolare “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche (CNR), 40136 Bologna, Italy;
| | - Daria Maria Filippini
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Manuela Ferracin
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
18
|
Fu X, Ma W, Zuo Q, Qi Y, Zhang S, Zhao Y. Application of machine learning for high-throughput tumor marker screening. Life Sci 2024; 348:122634. [PMID: 38685558 DOI: 10.1016/j.lfs.2024.122634] [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: 01/16/2024] [Revised: 03/26/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
High-throughput sequencing and multiomics technologies have allowed increasing numbers of biomarkers to be mined and used for disease diagnosis, risk stratification, efficacy assessment, and prognosis prediction. However, the large number and complexity of tumor markers make screening them a substantial challenge. Machine learning (ML) offers new and effective ways to solve the screening problem. ML goes beyond mere data processing and is instrumental in recognizing intricate patterns within data. ML also has a crucial role in modeling dynamic changes associated with diseases. Used together, ML techniques have been included in automatic pipelines for tumor marker screening, thereby enhancing the efficiency and accuracy of the screening process. In this review, we discuss the general processes and common ML algorithms, and highlight recent applications of ML in tumor marker screening of genomic, transcriptomic, proteomic, and metabolomic data of patients with various types of cancers. Finally, the challenges and future prospects of the application of ML in tumor therapy are discussed.
Collapse
Affiliation(s)
- Xingxing Fu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Wanting Ma
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Qi Zuo
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Yanfei Qi
- Centenary Institute, The University of Sydney, Sydney, NSW 2050, Australia
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| |
Collapse
|
19
|
Hoang DT, Shulman ED, Turakulov R, Abdullaev Z, Singh O, Campagnolo EM, Lalchungnunga H, Stone EA, Nasrallah MP, Ruppin E, Aldape K. Prediction of DNA methylation-based tumor types from histopathology in central nervous system tumors with deep learning. Nat Med 2024; 30:1952-1961. [PMID: 38760587 DOI: 10.1038/s41591-024-02995-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/11/2024] [Indexed: 05/19/2024]
Abstract
Precision in the diagnosis of diverse central nervous system (CNS) tumor types is crucial for optimal treatment. DNA methylation profiles, which capture the methylation status of thousands of individual CpG sites, are state-of-the-art data-driven means to enhance diagnostic accuracy but are also time consuming and not widely available. Here, to address these limitations, we developed Deep lEarning from histoPathoLOgy and methYlation (DEPLOY), a deep learning model that classifies CNS tumors to ten major categories from histopathology. DEPLOY integrates three distinct components: the first classifies CNS tumors directly from slide images ('direct model'), the second initially generates predictions for DNA methylation beta values, which are subsequently used for tumor classification ('indirect model'), and the third classifies tumor types directly from routinely available patient demographics. First, we find that DEPLOY accurately predicts beta values from histopathology images. Second, using a ten-class model trained on an internal dataset of 1,796 patients, we predict the tumor categories in three independent external test datasets including 2,156 patients, achieving an overall accuracy of 95% and balanced accuracy of 91% on samples that are predicted with high confidence. These results showcase the potential future use of DEPLOY to assist pathologists in diagnosing CNS tumors within a clinically relevant short time frame.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Eldad D Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Rust Turakulov
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Omkar Singh
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Emma M Campagnolo
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - H Lalchungnunga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Eric A Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - MacLean P Nasrallah
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
20
|
Burgermeister S, Stoykova S, Krebs FS, Zoete V, Mbefo M, Egervari K, Reinhard A, Bisig B, Hewer E. Methylation-Based Characterization of a New IDH2 Mutation in Sinonasal Undifferentiated Carcinoma. Int J Mol Sci 2024; 25:6518. [PMID: 38928223 PMCID: PMC11204065 DOI: 10.3390/ijms25126518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Mutations affecting codon 172 of the isocitrate dehydrogenase 2 (IDH2) gene define a subgroup of sinonasal undifferentiated carcinomas (SNUCs) with a relatively favorable prognosis and a globally hypermethylated phenotype. They are also recurrent (along with IDH1 mutations) in gliomas, acute myeloid leukemia, and intrahepatic cholangiocarcinoma. Commonly reported mutations, all associated with aberrant IDH2 enzymatic activity, include R172K, R172S, R172T, R172G, and R172M. We present a case of SNUC with a never-before-described IDH2 mutation, R172A. Our report compares the methylation pattern of our sample to other cases from the Gene Expression Omnibus database. Hierarchical clustering suggests a strong association between our sample and other IDH-mutant SNUCs and a clear distinction between sinonasal normal tissues and tumors. Principal component analysis (PCA), using 100 principal components explaining 94.5% of the variance, showed the position of our sample to be within 1.02 standard deviation of the other IDH-mutant SNUCs. A molecular modeling analysis of the IDH2 R172A versus other R172 variants provides a structural explanation to how they affect the protein active site. Our findings thus suggest that the R172A mutation in IDH2 confers a gain of function similar to other R172 mutations in IDH2, resulting in a similar hypermethylated profile.
Collapse
Affiliation(s)
- Simon Burgermeister
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Simona Stoykova
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Fanny S. Krebs
- Computer-Aided Molecular Engineering, Department of Oncology UNIL-CHUV, University of Lausanne, 1066 Epalinges, Switzerland; (F.S.K.); (V.Z.)
- Ludwig Institute for Cancer Research, 1066 Epalinges, Switzerland
| | - Vincent Zoete
- Computer-Aided Molecular Engineering, Department of Oncology UNIL-CHUV, University of Lausanne, 1066 Epalinges, Switzerland; (F.S.K.); (V.Z.)
- Ludwig Institute for Cancer Research, 1066 Epalinges, Switzerland
- Molecular Modelling Group, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Martial Mbefo
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Kristof Egervari
- Service of Clinical Pathology, Department of Diagnostics, Geneva University Hospitals, 1206 Geneva, Switzerland;
| | - Antoine Reinhard
- Department of Otorhinolaryngology-Head and Neck Surgery, Lausanne University Hospital, 1011 Lausanne, Switzerland;
| | - Bettina Bisig
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Ekkehard Hewer
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| |
Collapse
|
21
|
Hoke ATK, Takahashi Y, Padget MR, Gomez J, Amit M, Burks J, Bell D, Xie T, Soon-Shiong P, Hodge JW, Hanna EY, London NR. Targeting sinonasal undifferentiated carcinoma with a combinatory immunotherapy approach. Transl Oncol 2024; 44:101943. [PMID: 38593586 PMCID: PMC11024348 DOI: 10.1016/j.tranon.2024.101943] [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: 01/28/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
PURPOSE Sinonasal undifferentiated carcinoma (SNUC) is a rare, aggressive malignancy of the sinonasal cavity with poor prognosis and limited treatment options. To investigate the potential for SNUC sensitivity to combinatory immunotherapy, we performed in vitro studies with SNUC cell lines and used multi-spectral immunofluorescence to characterize the in vivo patient SNUC tumor immune microenvironment (TIME). EXPERIMENTAL DESIGN Human-derived SNUC cell lines were used for in vitro studies of tumor cell susceptibility to natural killer (NK) cell-based immunotherapeutic strategies. Tumor samples from 14 treatment naïve SNUC patients were examined via multi-spectral immunofluorescence and clinical correlations assessed. RESULTS Anti-PD-L1 blockade enhanced NK cell lysis of SNUC cell lines ∼5.4 fold (P ≤ 0.0001). This effect was blocked by a CD16 neutralizing antibody demonstrating activity through an antibody-dependent cellular cytotoxicity (ADCC) mediated pathway. ADCC-dependent lysis of SNUC cells was further enhanced by upregulation of PD-L1 on tumor cells by exogenous interferon-gamma (IFN-γ) administration or interleukin-15 (IL-15) stimulated IFN-γ release from NK cells. Combination treatment with anti-PD-L1 blockade and IL-15 superagonism enhanced NK-cell killing of SNUC cells 9.6-fold (P ≤ 0.0001). Untreated SNUC patient tumor samples were found to have an NK cell infiltrate and PD-L1+ tumor cells at a median of 5.4 cells per mm2. A striking 55.7-fold increase in CKlow tumor cell/NK cell interactions was observed in patients without disease recurrence after treatment (P = 0.022). Patients with higher CD3+CD8+ in the stroma had a significantly improved 5-year overall survival (P = 0.0029) and a significant increase in CKlow tumor cell/CD8+ cytotoxic T cell interactions was noted in long-term survivors (P = 0.0225). CONCLUSION These data provide the pre-clinical rationale for ongoing investigation into combinatory immunotherapy approaches for SNUC.
Collapse
Affiliation(s)
- Austin T K Hoke
- Sinonasal and Skull Base Tumor Program, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yoko Takahashi
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michelle R Padget
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Javier Gomez
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jared Burks
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diana Bell
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, United States
| | - Tongxin Xie
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - James W Hodge
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ehab Y Hanna
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nyall R London
- Sinonasal and Skull Base Tumor Program, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States; Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
22
|
Rooper LM, Agaimy A, Bell D, Gagan J, Gallia GL, Jo VY, Lewis JS, London NR, Nishino M, Stoehr R, Thompson LDR, Din NU, Wenig BM, Westra WH, Bishop JA. Recurrent Wnt Pathway and ARID1A Alterations in Sinonasal Olfactory Carcinoma. Mod Pathol 2024; 37:100448. [PMID: 38369189 DOI: 10.1016/j.modpat.2024.100448] [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: 05/28/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/20/2024]
Abstract
Sinonasal tumors with neuroepithelial differentiation, defined by neuroectodermal elements reminiscent of olfactory neuroblastoma (ONB) and epithelial features such as keratin expression or gland formation, are a diagnostically challenging group that has never been formally included in sinonasal tumor classifications. Recently, we documented that most of these neuroepithelial neoplasms have distinctive histologic and immunohistochemical findings and proposed the term "olfactory carcinoma" to describe these tumors. However, the molecular characteristics of olfactory carcinoma have not yet been evaluated. In this study, we performed targeted molecular profiling of 23 sinonasal olfactory carcinomas to further clarify their pathogenesis and classification. All tumors included in this study were composed of high-grade neuroectodermal cells that were positive for pankeratin and at least 1 specific neuroendocrine marker. A significant subset of cases also displayed rosettes and neurofibrillary matrix, intermixed glands with variable cilia, peripheral p63/p40 expression, and S100 protein-positive sustentacular cells. Recurrent oncogenic molecular alterations were identified in 20 tumors, including Wnt pathway alterations affecting CTNNB1 (n = 8) and PPP2R1A (n = 2), ARID1A inactivation (n = 5), RUNX1 mutations (n = 3), and IDH2 hotspot mutations (n = 2). Overall, these findings do demonstrate the presence of recurrent molecular alterations in olfactory carcinoma, although this group of tumors does not appear to be defined by any single mutation. Minimal overlap with alterations previously reported in ONB also adds to histologic and immunohistochemical separation between ONB and olfactory carcinoma. Conversely, these molecular findings enhance the overlap between olfactory carcinoma and sinonasal neuroendocrine carcinomas. A small subset of neuroepithelial tumors might better fit into the superseding molecular category of IDH2-mutant sinonasal carcinoma. At this point, sinonasal neuroendocrine and neuroepithelial tumors may best be regarded as a histologic and molecular spectrum that includes core groups of ONB, olfactory carcinoma, neuroendocrine carcinoma, and IDH2-mutant sinonasal carcinoma.
Collapse
Affiliation(s)
- Lisa M Rooper
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital, Erlangen, Germany
| | - Diana Bell
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Gary L Gallia
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vickie Y Jo
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - James S Lewis
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nyall R London
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michiya Nishino
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Robert Stoehr
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital, Erlangen, Germany
| | | | - Nasir Ud Din
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Bruce M Wenig
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - William H Westra
- Department of Pathology, Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
23
|
Fattori A, Reita D, Brinkert D, Willaume T, Gantzer J, Karanian M, Lhermitte B, Wolf T, De Pinieux G, Weingertner N. Coxal giant cell-rich tumour harbouring an NUTM1 gene fusion: a new molecular subtype of giant cell tumour of bone? Histopathology 2024; 84:1072-1075. [PMID: 38229224 DOI: 10.1111/his.15139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/30/2023] [Indexed: 01/18/2024]
Affiliation(s)
- Antonin Fattori
- Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | - Damien Reita
- Department of Cancer Molecular Genetics, Laboratory of Biochemistry and Molecular Biology, Strasbourg University Hospital, Strasbourg, France
| | - David Brinkert
- Department of Orthopedic Surgery and Traumatology, Strasbourg University Hospital, Strasbourg, France
| | - Thibault Willaume
- Department of Radiology, Strasbourg University Hospital, Strasbourg, France
| | - Justine Gantzer
- Department of Medical Oncology, Strasbourg-Europe Cancer Institute, Strasbourg, France
| | - Marie Karanian
- Department of Biopathology, Centre Léon Bérard, Lyon, France
| | - Benoit Lhermitte
- Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | - Thibaut Wolf
- Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| | | | - Noëlle Weingertner
- Department of Pathology, Strasbourg University Hospital, Strasbourg, France
| |
Collapse
|
24
|
Hermsen M, Bossi P, Capper D, Fleming J, Haybaeck J, Martinez-Balibrea E, Nuyts S, Skalova A, Thomson D, Trama A, Turri-Zanoni M, Verillaud B, Woods R, von Buchwald C, Lechner M. The European Network for Sinonasal Cancer Research (EUSICA) - A pan-European initiative targeting a group of orphan tumours. Eur J Cancer 2024; 202:113939. [PMID: 38447380 DOI: 10.1016/j.ejca.2024.113939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Affiliation(s)
- Mario Hermsen
- Dept Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.
| | - Paolo Bossi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan 20072, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - David Capper
- Department of Neuropathology, Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin), Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Johannes Haybaeck
- Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, Graz, Styria, Austria; Department of Pathology, Saint Vincent Hospital Zams, Zams, Tyrol, Austria; Department of Pathology, University Medical Centre Maribor, Maribor, Styria, Slovenia
| | - Eva Martinez-Balibrea
- ProCURE program, Catalan Institute of Oncology and CARE program, Germans Trias i Pujol Research Institute (IGTP) Ctra. De Can Ruti, cami de les escoles s/n, Badalona 08916, Spain
| | - Sandra Nuyts
- Laboratory of Experimental Radiotherapy, Department of Oncology, KU Leuven, Department of Radiation Oncology, Leuven Cancer Institute, University Hospitals, Leuven 3000, Belgium
| | - Alena Skalova
- Department of Pathology, Charles University, Faculty of Medicine in Plzen, Plzen, Czech Republic
| | - David Thomson
- The Christie NHS Foundation Trust, Manchester, UK; The University of Manchester, Manchester, UK
| | - Annalisa Trama
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mario Turri-Zanoni
- Unit of Otorhinolaryngology and Head & Neck Surgery, Department of Biotechnology and Life Sciences, ASST Lariana, University of Insubria, Como, Italy
| | | | - Robbie Woods
- Department of Otolaryngology - Head and Neck Surgery, Beaumont Hospital / Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Christian von Buchwald
- Department of ORL, Head & Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Matt Lechner
- Division of Surgery and Interventional Science and UCL Cancer Institute, University College London, London, UK.
| |
Collapse
|
25
|
Raj A, Petreaca RC, Mirzaei G. Multi-Omics Integration for Liver Cancer Using Regression Analysis. Curr Issues Mol Biol 2024; 46:3551-3562. [PMID: 38666952 PMCID: PMC11049490 DOI: 10.3390/cimb46040222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Genetic biomarkers have played a pivotal role in the classification, prognostication, and guidance of clinical cancer therapies. Large-scale and multi-dimensional analyses of entire cancer genomes, as exemplified by projects like The Cancer Genome Atlas (TCGA), have yielded an extensive repository of data that holds the potential to unveil the underlying biology of these malignancies. Mutations stand out as the principal catalysts of cellular transformation. Nonetheless, other global genomic processes, such as alterations in gene expression and chromosomal re-arrangements, also play crucial roles in conferring cellular immortality. The incorporation of multi-omics data specific to cancer has demonstrated the capacity to enhance our comprehension of the molecular mechanisms underpinning carcinogenesis. This report elucidates how the integration of comprehensive data on methylation, gene expression, and copy number variations can effectively facilitate the unsupervised clustering of cancer samples. We have identified regressors that can effectively classify tumor and normal samples with an optimal integration of RNA sequencing, DNA methylation, and copy number variation while also achieving significant p-values. Further, these regressors were trained using linear and logistic regression with k-means clustering. For comparison, we employed autoencoder- and stacking-based omics integration and computed silhouette scores to evaluate the clusters. The proof of concept is illustrated using liver cancer data. Our analysis serves to underscore the feasibility of unsupervised cancer classification by considering genetic markers beyond mutations, thereby emphasizing the clinical relevance of additional global cellular parameters that contribute to the transformative process in cells. This work is clinically relevant because changes in gene expression and genomic re-arrangements have been shown to be signatures of cellular transformation across cancers, as well as in liver cancers.
Collapse
Affiliation(s)
- Aditya Raj
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, OH 43210, USA;
| | - Ruben C. Petreaca
- Department of Molecular Genetics, The Ohio State University, Marion, OH 43302, USA;
- Cancer Biology Program, The Ohio State University James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Golrokh Mirzaei
- Department of Computer Science and Engineering, The Ohio State University, Marion, OH 43302, USA
| |
Collapse
|
26
|
Hench J, Hultschig C, Brugger J, Mariani L, Guzman R, Soleman J, Leu S, Benton M, Stec IM, Hench IB, Hoffmann P, Harter P, Weber KJ, Albers A, Thomas C, Hasselblatt M, Schüller U, Restelli L, Capper D, Hewer E, Diebold J, Kolenc D, Schneider UC, Rushing E, Della Monica R, Chiariotti L, Sill M, Schrimpf D, von Deimling A, Sahm F, Kölsche C, Tolnay M, Frank S. EpiDiP/NanoDiP: a versatile unsupervised machine learning edge computing platform for epigenomic tumour diagnostics. Acta Neuropathol Commun 2024; 12:51. [PMID: 38576030 PMCID: PMC10993614 DOI: 10.1186/s40478-024-01759-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/11/2024] [Indexed: 04/06/2024] Open
Abstract
DNA methylation analysis based on supervised machine learning algorithms with static reference data, allowing diagnostic tumour typing with unprecedented precision, has quickly become a new standard of care. Whereas genome-wide diagnostic methylation profiling is mostly performed on microarrays, an increasing number of institutions additionally employ nanopore sequencing as a faster alternative. In addition, methylation-specific parallel sequencing can generate methylation and genomic copy number data. Given these diverse approaches to methylation profiling, to date, there is no single tool that allows (1) classification and interpretation of microarray, nanopore and parallel sequencing data, (2) direct control of nanopore sequencers, and (3) the integration of microarray-based methylation reference data. Furthermore, no software capable of entirely running in routine diagnostic laboratory environments lacking high-performance computing and network infrastructure exists. To overcome these shortcomings, we present EpiDiP/NanoDiP as an open-source DNA methylation and copy number profiling suite, which has been benchmarked against an established supervised machine learning approach using in-house routine diagnostics data obtained between 2019 and 2021. Running locally on portable, cost- and energy-saving system-on-chip as well as gpGPU-augmented edge computing devices, NanoDiP works in offline mode, ensuring data privacy. It does not require the rigid training data annotation of supervised approaches. Furthermore, NanoDiP is the core of our public, free-of-charge EpiDiP web service which enables comparative methylation data analysis against an extensive reference data collection. We envision this versatile platform as a useful resource not only for neuropathologists and surgical pathologists but also for the tumour epigenetics research community. In daily diagnostic routine, analysis of native, unfixed biopsies by NanoDiP delivers molecular tumour classification in an intraoperative time frame.
Collapse
Affiliation(s)
- Jürgen Hench
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland.
| | - Claus Hultschig
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland
| | - Jon Brugger
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland
| | - Luigi Mariani
- Klinik für Neurochirurgie, Universitätsspital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Raphael Guzman
- Klinik für Neurochirurgie, Universitätsspital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Jehuda Soleman
- Klinik für Neurochirurgie, Universitätsspital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Severina Leu
- Klinik für Neurochirurgie, Universitätsspital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Miles Benton
- Human Genomics, Institute of Environmental Science and Research (ESR), 5022, Porirua, Wellington, New Zealand
| | - Irenäus Maria Stec
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland
| | - Ivana Bratic Hench
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland
| | - Per Hoffmann
- Life&Brain GmbH, Venusberg-Campus 1, Gebäude 76, 53127, Bonn, Germany
| | - Patrick Harter
- Institute of Neuropathology, Center for Neuropathology and Prion Research, Feodor- Lynen-Str. 23, 81377, München, Germany
| | - Katharina J Weber
- Neurological Institute (Edinger Institute), University Hospital, Heinrich-Hoffmann- Straße 7, 60528, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Anne Albers
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Christian Thomas
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Pottkamp 2, 48149, Münster, Germany
| | - Ulrich Schüller
- Forschungsinstitut Kinderkrebszentrum, Martinistrasse 52, 20251, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg- Eppendorf, Hamburg, Germany
- Institute of Neuropathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Department of Neuropathology, Department of Neuropathology, Charité- Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Lisa Restelli
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland
| | - David Capper
- , 15. Luzerner Kantonsspital, Pathologie, Haus 27, 6000, Spitalstrasse, Luzern 16, Switzerland
| | - Ekkehard Hewer
- Institut universitaire de pathologie, Lausanne University Hospital (CHUV), University of Lausanne, Rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Joachim Diebold
- , 15. Luzerner Kantonsspital, Pathologie, Haus 27, 6000, Spitalstrasse, Luzern 16, Switzerland
| | - Danijela Kolenc
- , 15. Luzerner Kantonsspital, Pathologie, Haus 27, 6000, Spitalstrasse, Luzern 16, Switzerland
| | - Ulf C Schneider
- Klinik für Neurochirurgie, Luzerner Kantonsspital, Haus 31, 6000, 16, Spitalstrasse, Luzern, Switzerland
| | - Elisabeth Rushing
- , 15. Luzerner Kantonsspital, Pathologie, Haus 27, 6000, Spitalstrasse, Luzern 16, Switzerland
- Medica Pathologie Zentrum Zürich, Hottingerstrasse 9 / 11, 8032, Zürich, Switzerland
| | - Rosa Della Monica
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486 - 80145, Napoli, Italy
| | - Lorenzo Chiariotti
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486 - 80145, Napoli, Italy
| | - Martin Sill
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Daniel Schrimpf
- Department of Neuropathology, Institute of Neuropathology, University Hospital Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Neuropathology, University Hospital Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Institute of Neuropathology, University Hospital Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- CCU Neuropathology, German Consortium for Translational Cancer Research (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- , 23. DKFZ, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Christian Kölsche
- Pathologisches Institut der LMU, Thalkirchner Str. 36, 80337, München, Germany
| | - Markus Tolnay
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland
| | - Stephan Frank
- Institut für Medizinische Genetik und Pathologie, Universitätsspital Basel, Schönbeinstr. 40, 4031, Basel, Switzerland.
| |
Collapse
|
27
|
Hoch CC, Knoedler L, Knoedler S, Bashiri Dezfouli A, Schmidl B, Trill A, Douglas JE, Adappa ND, Stögbauer F, Wollenberg B. Integrated Molecular and Histological Insights for Targeted Therapies in Mesenchymal Sinonasal Tract Tumors. Curr Oncol Rep 2024; 26:272-291. [PMID: 38376625 PMCID: PMC10920452 DOI: 10.1007/s11912-024-01506-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide a comprehensive overview of mesenchymal sinonasal tract tumors (STTs), a distinct subset of STTs. Despite their rarity, mesenchymal STTs represent a unique clinical challenge, characterized by their rarity, often slow progression, and frequently subtle or overlooked symptoms. The complex anatomy of the sinonasal area, which includes critical structures such as the orbit, brain, and cranial nerves, further complicates surgical treatment options. This underscores an urgent need for more advanced and specialized therapeutic approaches. RECENT FINDINGS Advancements in molecular diagnostics, particularly in next-generation sequencing, have significantly enhanced our understanding of STTs. Consequently, the World Health Organization has updated its tumor classification to better reflect the distinct histological and molecular profiles of these tumors, as well as to categorize mesenchymal STTs with greater accuracy. The growing understanding of the molecular characteristics of mesenchymal STTs opens new possibilities for targeted therapeutic interventions, marking a significant shift in treatment paradigms. This review article concentrates on mesenchymal STTs, specifically addressing sinonasal tract angiofibroma, sinonasal glomangiopericytoma, biphenotypic sinonasal sarcoma, and skull base chordoma. These entities are marked by unique histopathological and molecular features, which challenge conventional treatment approaches and simultaneously open avenues for novel targeted therapies. Our discussion is geared towards delineating the molecular underpinnings of mesenchymal STTs, with the objective of enhancing therapeutic strategies and addressing the existing shortcomings in the management of these intricate tumors.
Collapse
Affiliation(s)
- Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Ismaningerstrasse 22, 81675, Munich, Germany
| | - Leonard Knoedler
- Department of Surgery, Division of Plastic Surgery, Yale School of Medicine, New Haven, CT, USA
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Samuel Knoedler
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum Munich, Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Ismaningerstrasse 22, 81675, Munich, Germany
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany
| | - Benedikt Schmidl
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Ismaningerstrasse 22, 81675, Munich, Germany
| | - Anskar Trill
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Ismaningerstrasse 22, 81675, Munich, Germany
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany
| | - Jennifer E Douglas
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Nithin D Adappa
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Fabian Stögbauer
- Institute of Pathology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine and Health, Technical University of Munich (TUM), Ismaningerstrasse 22, 81675, Munich, Germany.
| |
Collapse
|
28
|
Klauschen F, Dippel J, Keyl P, Jurmeister P, Bockmayr M, Mock A, Buchstab O, Alber M, Ruff L, Montavon G, Müller KR. [Explainable artificial intelligence in pathology]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:133-139. [PMID: 38315198 DOI: 10.1007/s00292-024-01308-7] [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: 01/19/2024] [Indexed: 02/07/2024]
Abstract
With the advancements in precision medicine, the demands on pathological diagnostics have increased, requiring standardized, quantitative, and integrated assessments of histomorphological and molecular pathological data. Great hopes are placed in artificial intelligence (AI) methods, which have demonstrated the ability to analyze complex clinical, histological, and molecular data for disease classification, biomarker quantification, and prognosis estimation. This paper provides an overview of the latest developments in pathology AI, discusses the limitations, particularly concerning the black box character of AI, and describes solutions to make decision processes more transparent using methods of so-called explainable AI (XAI).
Collapse
Affiliation(s)
- Frederick Klauschen
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland.
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Berlin, Deutschland.
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Deutschland.
- Deutsches Krebsforschungszentrum (DKTK/DKFZ), Partnerstandort München, München, Deutschland.
| | - Jonas Dippel
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Deutschland
- Machine Learning Group, Fachbereich Elektrotechnik und Informatik, Technische Universität Berlin, Berlin, Deutschland
| | - Philipp Keyl
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
| | - Philipp Jurmeister
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
- Deutsches Krebsforschungszentrum (DKTK/DKFZ), Partnerstandort München, München, Deutschland
| | - Michael Bockmayr
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
- Pädiatrische Hämatologie und Onkologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
- Forschungsinstitut Kinderkrebs-Zentrum Hamburg, Hamburg, Deutschland
| | - Andreas Mock
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
- Deutsches Krebsforschungszentrum (DKTK/DKFZ), Partnerstandort München, München, Deutschland
| | - Oliver Buchstab
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
| | - Maximilian Alber
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
- Aignostics GmbH, Berlin, Deutschland
| | | | - Grégoire Montavon
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Deutschland
- Machine Learning Group, Fachbereich Elektrotechnik und Informatik, Technische Universität Berlin, Berlin, Deutschland
- Fachbereich Mathematik und Informatik, Freie Universität Berlin, Berlin, Deutschland
| | - Klaus-Robert Müller
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Deutschland.
- Machine Learning Group, Fachbereich Elektrotechnik und Informatik, Technische Universität Berlin, Berlin, Deutschland.
- Department of Artificial Intelligence, Korea University, Seoul, Südkorea.
- Max-Planck-Institut für Informatik, Saarbrücken, Deutschland.
- Machine Learning/Intelligent Data Analysis (IDA), Technische Universität Berlin, Marchstr. 23, 10587, Berlin, Deutschland.
| |
Collapse
|
29
|
Adamane S, Weir J. A diagnostic approach to small round cell epithelial and neuroepithelial tumours of the sinonasal tract. DIAGNOSTIC HISTOPATHOLOGY 2024; 30:179-187. [DOI: 10.1016/j.mpdhp.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Sun M, Xu B, Chen C, Zhu Y, Li X, Chen K. Tissue of origin prediction for cancer of unknown primary using a targeted methylation sequencing panel. Clin Epigenetics 2024; 16:25. [PMID: 38336771 PMCID: PMC10854167 DOI: 10.1186/s13148-024-01638-6] [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: 11/20/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
RATIONALE Cancer of unknown primary (CUP) is a group of rare malignancies with poor prognosis and unidentifiable tissue-of-origin. Distinct DNA methylation patterns in different tissues and cancer types enable the identification of the tissue of origin in CUP patients, which could help risk assessment and guide site-directed therapy. METHODS Using genome-wide DNA methylation profile datasets from The Cancer Genome Atlas (TCGA) and machine learning methods, we developed a 200-CpG methylation feature classifier for CUP tissue of origin prediction (MFCUP). MFCUP was further validated with public-available methylation array data of 2977 specimens and targeted methylation sequencing of 78 Formalin-fixed paraffin-embedded (FFPE) samples from a single center. RESULTS MFCUP achieved an accuracy of 97.2% in a validation cohort (n = 5923) representing 25 cancer types. When applied to an Infinium 450 K array dataset (n = 1052) and an Infinium EPIC (850 K) array dataset (n = 1925), MFCUP achieved an overall accuracy of 93.4% and 84.8%, respectively. Based on MFCUP, we established a targeted bisulfite sequencing panel and validated it with FFPE sections from 78 patients of 20 cancer types. This methylation sequencing panel correctly identified tissue of origin in 88.5% (69/78) of samples. We also found that the methylation levels of specific CpGs can distinguish one cancer type from others, indicating their potential as biomarkers for cancer diagnosis and screening. CONCLUSION Our methylation-based cancer classifier and targeted methylation sequencing panel can predict tissue of origin in diverse cancer types with high accuracy.
Collapse
Affiliation(s)
- Miaomiao Sun
- Department of Pathology, Henan Key Laboratory of Tumor Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo Xu
- Research and Development Division, Oriomics Biotech Inc, Hangzhou, China
| | - Chao Chen
- Department of Pathology, Henan Key Laboratory of Tumor Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Youjie Zhu
- Research and Development Division, Oriomics Biotech Inc, Hangzhou, China
| | - Xiaomo Li
- Research and Development Division, Oriomics Biotech Inc, Hangzhou, China.
| | - Kuisheng Chen
- Department of Pathology, Henan Key Laboratory of Tumor Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
31
|
Klauschen F, Dippel J, Keyl P, Jurmeister P, Bockmayr M, Mock A, Buchstab O, Alber M, Ruff L, Montavon G, Müller KR. Toward Explainable Artificial Intelligence for Precision Pathology. ANNUAL REVIEW OF PATHOLOGY 2024; 19:541-570. [PMID: 37871132 DOI: 10.1146/annurev-pathmechdis-051222-113147] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The rapid development of precision medicine in recent years has started to challenge diagnostic pathology with respect to its ability to analyze histological images and increasingly large molecular profiling data in a quantitative, integrative, and standardized way. Artificial intelligence (AI) and, more precisely, deep learning technologies have recently demonstrated the potential to facilitate complex data analysis tasks, including clinical, histological, and molecular data for disease classification; tissue biomarker quantification; and clinical outcome prediction. This review provides a general introduction to AI and describes recent developments with a focus on applications in diagnostic pathology and beyond. We explain limitations including the black-box character of conventional AI and describe solutions to make machine learning decisions more transparent with so-called explainable AI. The purpose of the review is to foster a mutual understanding of both the biomedical and the AI side. To that end, in addition to providing an overview of the relevant foundations in pathology and machine learning, we present worked-through examples for a better practical understanding of what AI can achieve and how it should be done.
Collapse
Affiliation(s)
- Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- German Cancer Consortium, German Cancer Research Center (DKTK/DKFZ), Munich Partner Site, Munich, Germany
| | - Jonas Dippel
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- Machine Learning Group, Department of Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany;
| | - Philipp Keyl
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
| | - Philipp Jurmeister
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
- German Cancer Consortium, German Cancer Research Center (DKTK/DKFZ), Munich Partner Site, Munich, Germany
| | - Michael Bockmayr
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Andreas Mock
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
- German Cancer Consortium, German Cancer Research Center (DKTK/DKFZ), Munich Partner Site, Munich, Germany
| | - Oliver Buchstab
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
| | - Maximilian Alber
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Aignostics, Berlin, Germany
| | | | - Grégoire Montavon
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- Machine Learning Group, Department of Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany;
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Klaus-Robert Müller
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- Machine Learning Group, Department of Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany;
- Department of Artificial Intelligence, Korea University, Seoul, Korea
- Max Planck Institute for Informatics, Saarbrücken, Germany
| |
Collapse
|
32
|
Lorenzo-Guerra SL, Codina-Martínez H, Suárez-Fernández L, Cabal VN, García-Marín R, Riobello C, Vivanco B, Blanco-Lorenzo V, Sánchez-Fernández P, López F, Llorente JL, Hermsen MA. Characterization of a Preclinical In Vitro Model Derived from a SMARCA4-Mutated Sinonasal Teratocarcinosarcoma. Cells 2023; 13:81. [PMID: 38201285 PMCID: PMC10778008 DOI: 10.3390/cells13010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Sinonasal teratocarcinosarcoma (TCS) is a rare tumor that displays a variable histology with admixtures of epithelial, mesenchymal, neuroendocrine and germ cell elements. Facing a very poor prognosis, patients with TCS are in need of new options for treatment. Recently identified recurrent mutations in SMARCA4 may serve as target for modern therapies with EZH1/2 and CDK4/6 inhibitors. Here, we present the first in vitro cell line TCS627, established from a previously untreated primary TCS originating in the ethmoid sinus with invasion into the brain. The cultured cells expressed immunohistochemical markers, indicating differentiation of epithelial, neuroepithelial, sarcomatous and teratomatous components. Whole-exome sequencing revealed 99 somatic mutations including SMARCA4, ARID2, TET2, CDKN2A, WNT7A, NOTCH3 and STAG2, all present both in the primary tumor and in the cell line. Focusing on mutated SMARCA4 as the therapeutic target, growth inhibition assays showed a strong response to the CDK4/6 inhibitor palbociclib, but much less to the EZH1/2 inhibitor valemetostat. In conclusion, cell line TCS627 carries both histologic and genetic features characteristic of TCS and is a valuable model for both basic research and preclinical testing of new therapeutic options for treatment of TCS patients.
Collapse
Affiliation(s)
- Sara Lucila Lorenzo-Guerra
- Department of Head and Neck Cancer, Health Research Institute of the Principality of Asturias, 33011 Oviedo, Spain; (S.L.L.-G.); (H.C.-M.); (L.S.-F.); (V.N.C.); (R.G.-M.); (C.R.)
| | - Helena Codina-Martínez
- Department of Head and Neck Cancer, Health Research Institute of the Principality of Asturias, 33011 Oviedo, Spain; (S.L.L.-G.); (H.C.-M.); (L.S.-F.); (V.N.C.); (R.G.-M.); (C.R.)
| | - Laura Suárez-Fernández
- Department of Head and Neck Cancer, Health Research Institute of the Principality of Asturias, 33011 Oviedo, Spain; (S.L.L.-G.); (H.C.-M.); (L.S.-F.); (V.N.C.); (R.G.-M.); (C.R.)
| | - Virginia N. Cabal
- Department of Head and Neck Cancer, Health Research Institute of the Principality of Asturias, 33011 Oviedo, Spain; (S.L.L.-G.); (H.C.-M.); (L.S.-F.); (V.N.C.); (R.G.-M.); (C.R.)
| | - Rocío García-Marín
- Department of Head and Neck Cancer, Health Research Institute of the Principality of Asturias, 33011 Oviedo, Spain; (S.L.L.-G.); (H.C.-M.); (L.S.-F.); (V.N.C.); (R.G.-M.); (C.R.)
| | - Cristina Riobello
- Department of Head and Neck Cancer, Health Research Institute of the Principality of Asturias, 33011 Oviedo, Spain; (S.L.L.-G.); (H.C.-M.); (L.S.-F.); (V.N.C.); (R.G.-M.); (C.R.)
| | - Blanca Vivanco
- Department of Pathology, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (B.V.)
| | - Verónica Blanco-Lorenzo
- Department of Pathology, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (B.V.)
| | - Paula Sánchez-Fernández
- Department of Otolaryngology, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (P.S.-F.); (F.L.); (J.L.L.)
| | - Fernando López
- Department of Otolaryngology, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (P.S.-F.); (F.L.); (J.L.L.)
| | - Jóse Luis Llorente
- Department of Otolaryngology, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (P.S.-F.); (F.L.); (J.L.L.)
| | - Mario A. Hermsen
- Department of Head and Neck Cancer, Health Research Institute of the Principality of Asturias, 33011 Oviedo, Spain; (S.L.L.-G.); (H.C.-M.); (L.S.-F.); (V.N.C.); (R.G.-M.); (C.R.)
| |
Collapse
|
33
|
Hermsen MA, Lechner M, Oliveira Ferrer L, Trama A, Eriksen PRG, Martinez-Balibrea E, von Buchwald C. EUSICA/COST IMMUNO-model workshop fostering collaboration to advance sinonasal cancer research: A meeting report. Oral Oncol 2023; 146:106543. [PMID: 37573683 DOI: 10.1016/j.oraloncology.2023.106543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Sinonasal cancer is a clinically and histologically heterogeneous group of rare tumors with generally poor clinical outcomes. Their low incidence hampers the advancement of clinical management as well as translational research, and calls for multicenter and multinational collaboration between physicians and researchers. This report describes the proceedings of a two-day conference organized by the European Network for Sinonasal Cancer Research (EUSICA) and COST Action 'IMMUNO-model', fostering such collaboration and focusing on preclinical tumor and immuno models, surgical and radio-oncological treatments, core facilities for genetic characterization and molecular tumor classification, and cancer registry.
Collapse
Affiliation(s)
- Mario A Hermsen
- Dept Head and Neck Cancer, Health Research Institute of the Principality of Asturias, Oviedo, Spain.
| | - Matt Lechner
- CL Division of Surgery and Interventional Science and UCL Cancer Institute, University College London, London, UK
| | | | - Annalisa Trama
- Fondazione IRCCS Istituto Nazionale dei Tumori di milano, Milan, Italy
| | - Patrick René Gerhard Eriksen
- Dept Otorhinolaryngology, Head and Neck Surgery & Audiology, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| | - Eva Martinez-Balibrea
- Procure Program, Catalan Institue of Oncology and CARE Program, Germans Trias I Pujol Research Institute, Badalona, Spain
| | - Christian von Buchwald
- Dept Otorhinolaryngology, Head and Neck Surgery & Audiology, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
34
|
Edsjö A, Holmquist L, Geoerger B, Nowak F, Gomon G, Alix-Panabières C, Ploeger C, Lassen U, Le Tourneau C, Lehtiö J, Ott PA, von Deimling A, Fröhling S, Voest E, Klauschen F, Dienstmann R, Alshibany A, Siu LL, Stenzinger A. Precision cancer medicine: Concepts, current practice, and future developments. J Intern Med 2023; 294:455-481. [PMID: 37641393 DOI: 10.1111/joim.13709] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Precision cancer medicine is a multidisciplinary team effort that requires involvement and commitment of many stakeholders including the society at large. Building on the success of significant advances in precision therapy for oncological patients over the last two decades, future developments will be significantly shaped by improvements in scalable molecular diagnostics in which increasingly complex multilayered datasets require transformation into clinically useful information guiding patient management at fast turnaround times. Adaptive profiling strategies involving tissue- and liquid-based testing that account for the immense plasticity of cancer during the patient's journey and also include early detection approaches are already finding their way into clinical routine and will become paramount. A second major driver is the development of smart clinical trials and trial concepts which, complemented by real-world evidence, rapidly broaden the spectrum of therapeutic options. Tight coordination with regulatory agencies and health technology assessment bodies is crucial in this context. Multicentric networks operating nationally and internationally are key in implementing precision oncology in clinical practice and support developing and improving the ecosystem and framework needed to turn invocation into benefits for patients. The review provides an overview of the diagnostic tools, innovative clinical studies, and collaborative efforts needed to realize precision cancer medicine.
Collapse
Affiliation(s)
- Anders Edsjö
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
- Genomic Medicine Sweden (GMS), Kristianstad, Sweden
| | - Louise Holmquist
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Genomic Medicine Sweden (GMS), Kristianstad, Sweden
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | | | - Georgy Gomon
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France
- CREEC, MIVEGEC, University of Montpellier, Montpellier, France
| | - Carolin Ploeger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
| | - Ulrik Lassen
- Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- INSERM U900 Research Unit, Saint-Cloud, France
- Faculty of Medicine, Paris-Saclay University, Paris, France
| | - Janne Lehtiö
- Department of Oncology Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas von Deimling
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Emile Voest
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frederick Klauschen
- Institute of Pathology, Charite - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Munich Partner Site, Heidelberg, Germany
| | | | | | - Lillian L Siu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
| |
Collapse
|
35
|
Kazdal D, Menzel M, Budczies J, Stenzinger A. [Molecular tumor diagnostics as the driving force behind precision oncology]. Dtsch Med Wochenschr 2023; 148:1157-1165. [PMID: 37657453 DOI: 10.1055/a-1937-0347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
Molecular pathological diagnostics plays a central role in personalized oncology and requires multidisciplinary teamwork. It is just as relevant for the individual patient who is being treated with an approved therapy method or an individual treatment attempt as it is for prospective clinical studies that require the identification of specific therapeutic target structures or complex biomarkers for study inclusion. It is also of crucial importance for the generation of real-world data, which is becoming increasingly important for drug development. Future developments will be significantly shaped by improvements in scalable molecular diagnostics, in which increasingly complex and multi-layered data sets must be quickly converted into clinically useful information. One focus will be on the development of adaptive diagnostic strategies in order to be able to depict the enormous plasticity of a cancer disease over time.
Collapse
|
36
|
Dragomir MP, Calina TG, Perez E, Schallenberg S, Chen M, Albrecht T, Koch I, Wolkenstein P, Goeppert B, Roessler S, Calin GA, Sers C, Horst D, Roßner F, Capper D. DNA methylation-based classifier differentiates intrahepatic pancreato-biliary tumours. EBioMedicine 2023; 93:104657. [PMID: 37348162 DOI: 10.1016/j.ebiom.2023.104657] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 05/21/2023] [Accepted: 06/02/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Differentiating intrahepatic cholangiocarcinomas (iCCA) from hepatic metastases of pancreatic ductal adenocarcinoma (PAAD) is challenging. Both tumours have similar morphological and immunohistochemical pattern and share multiple driver mutations. We hypothesised that DNA methylation-based machine-learning algorithms may help perform this task. METHODS We assembled genome-wide DNA methylation data for iCCA (n = 259), PAAD (n = 431), and normal bile duct (n = 70) from publicly available sources. We split this cohort into a reference (n = 399) and a validation set (n = 361). Using the reference cohort, we trained three machine learning models to differentiate between these entities. Furthermore, we validated the classifiers on the technical validation set and used an internal cohort (n = 72) to test our classifier. FINDINGS On the validation cohort, the neural network, support vector machine, and the random forest classifiers reached accuracies of 97.68%, 95.62%, and 96.5%, respectively. Filtering by anomaly detection and thresholds improved the accuracy to 99.07% (37 samples excluded by filtering), 96.22% (17 samples excluded), and 100% (44 samples excluded) for the neural network, support vector machine and random forest, respectively. Because of best balance between accuracy and number of predictable cases we tested the neural network with applied filters on the in-house cohort, obtaining an accuracy of 95.45%. INTERPRETATION We developed a classifier that can differentiate between iCCAs, intrahepatic metastases of a PAAD, and normal bile duct tissue with high accuracy. This tool can be used for improving the diagnosis of pancreato-biliary cancers of the liver. FUNDING This work was supported by Berlin Institute of Health (JCS Program), DKTK Berlin (Young Investigator Grant 2022), German Research Foundation (493697503 and 314905040 - SFB/TRR 209 Liver Cancer B01), and German Cancer Aid (70113922).
Collapse
Affiliation(s)
- Mihnea P Dragomir
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Berlin Institute of Health, Berlin, Germany.
| | | | - Eilís Perez
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Berlin School of Integrative Oncology (BSIO), Charite - Universitätsmedizin Berlin (CVK), Berlin, Germany
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Meng Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Albrecht
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ines Koch
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Peggy Wolkenstein
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benjamin Goeppert
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Institute of Pathology and Neuropathology, Hospital RKH Kliniken Ludwigsburg, 71640 Ludwigsburg, Germany
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine Sers
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Florian Roßner
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Capper
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
37
|
Rooper LM. Proceedings of the 2023 North American Society of Head and Neck Pathology Companion Meeting, New Orleans, LA, March 12, 2023: Navigating New Developments in High Grade Sinonasal Neuroendocrine and Neuroectodermal Neoplasms. Head Neck Pathol 2023; 17:299-312. [PMID: 37184733 PMCID: PMC10293143 DOI: 10.1007/s12105-023-01548-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/18/2023] [Indexed: 05/16/2023]
Abstract
Although the definitions of sinonasal neuroendocrine and neuroectodermal neoplasms did not change substantially in the 5th edition WHO Classification of Head and Neck Tumours, the diagnosis of olfactory neuroblastoma (ONB), small cell neuroendocrine carcinoma, and large cell neuroendocrine carcinoma remains quite challenging in practice. Ambiguities surrounding the amount of keratin expression allowable in ONB and the amount of neuroendocrine differentiation seen in sinonasal undifferentiated carcinoma (SNUC) lead to significant diagnostic discrepancies at the high grade end of this tumor spectrum. Furthermore, a group of problematic neuroepithelial tumors that show overlapping features of ONB and neuroendocrine carcinoma have never been recognized in formal classification schemes. Since publication of the 5th edition WHO, two new tumor entities have been proposed that help resolve these problems. Olfactory carcinoma is defined by high grade keratin-positive neuroectodermal cells with frequent intermixed glands and shows recurrent Wnt pathway, ARID1A, and RUNX1 alterations. IDH2-mutant sinonasal carcinoma is a molecularly-defined category that encompasses tumors with undifferentiated (SNUC), large cell neuroendocrine, and neuroepithelial phenotypes. This review will provide a practical overview of these emerging entities and their application to diagnostic challenges in the post-WHO sinonasal neuroendocrine and neuroectodermal tumor classification.
Collapse
Affiliation(s)
- Lisa M Rooper
- Department of Pathology, The Johns Hopkins University School of Medicine, 401 N. Broadway, Weinberg 2242, Baltimore, MD, 21231, USA.
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
38
|
Cabezas-Camarero S, García-Barberán V, Benítez-Fuentes JD, Sotelo MJ, Plaza JC, Encinas-Bascones A, De-la-Sen Ó, Falahat F, Gimeno-Hernández J, Gómez-Serrano M, Puebla-Díaz F, De-Pedro-Marina M, Iglesias-Moreno M, Pérez-Segura P. Clinical Behavior, Mutational Profile and T-Cell Repertoire of High-Grade Neuroendocrine Tumors of the Head and Neck. Cancers (Basel) 2023; 15:cancers15092431. [PMID: 37173898 PMCID: PMC10177201 DOI: 10.3390/cancers15092431] [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: 03/15/2023] [Revised: 04/15/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Neuroendocrine carcinomas (NECs) of the head and neck (HN) account for <1% of HN cancers (HNCs), with a 5-year overall survival (OS) <20%. This is a retrospective study of HN NECs diagnosed at our institution between 2005 and 2022. Immunohistochemistry and next-generation sequencing (NGS) were used to evaluate neuroendocrine markers, tumor mutational burden (TMB), mutational profiles and T-cell receptor repertoires. Eleven patients with high-grade HN NECs were identified (male:female ratio 6:5; median age 61 (Min-Max: 31-86)): nasoethmoidal (3), parotid gland (3), submaxillary gland (1), larynx (3) and base of tongue (1). Among n = 8 stage II/IVA/B, all received (chemo)radiotherapy with/without prior surgery or induction chemotherapy, with complete response in 7/8 (87.5%). Among n = 6 recurrent/metastatic patients, three received anti-PD1 (nivolumab (2), pembrolizumab (1)): two achieved partial responses lasting 24 and 10 months. After a median follow-up of 30 and 23.5 months since diagnosis and since recurrent/metastatic, median OS was not reached. Median TMB (n = 7) was 6.72 Mut/Mb. The most common pathogenic variants were TP53, HNF1A, SMARCB1, CDKN2A, PIK3CA, RB1 and MYC. There were 224 median TCR clones (n = 5 pts). In one patient, TCR clones increased from 59 to 1446 after nivolumab. HN NECs may achieve long-lasting survival with multimodality treatment. They harbor moderate-high TMBs and large TCR repertoires, which may explain responses to anti-PD1 agents in two patients and justify the study of immunotherapy in this disease.
Collapse
Affiliation(s)
- Santiago Cabezas-Camarero
- Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Vanesa García-Barberán
- Molecular Oncology Laboratory, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Javier David Benítez-Fuentes
- Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Miguel J Sotelo
- Medical Oncology Department, Aliada Cancer Center, Lima 15036, Peru
- Medical Oncology Department, Clínica San Felipe, Lima 15072, Peru
- Medical Oncology Department, Hospital María Auxiliadora, Lima 15801, Peru
| | - José Carlos Plaza
- Pathology Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | | | - Óscar De-la-Sen
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Farzin Falahat
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Jesús Gimeno-Hernández
- Otolaryngology-Head and Neck Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Manuel Gómez-Serrano
- Otolaryngology-Head and Neck Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Fernando Puebla-Díaz
- Radiation Oncology Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Manuel De-Pedro-Marina
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Maricruz Iglesias-Moreno
- Maxillofacial Surgery Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain
| | - Pedro Pérez-Segura
- Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
39
|
Hermsen MA, Bossi P, Franchi A, Lechner M. Sinonasal Cancer: Improving Classification, Stratification and Therapeutic Options. Cancers (Basel) 2023; 15:1675. [PMID: 36980561 PMCID: PMC10046049 DOI: 10.3390/cancers15061675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
The nasal cavities and paranasal sinuses are the site of origin of a wide spectrum of histologically and clinically distinct disease entities [...].
Collapse
Affiliation(s)
- Mario A. Hermsen
- Department Head and Neck Oncology, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain
| | - Paolo Bossi
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Alessandro Franchi
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Matt Lechner
- UCL Cancer Institute, University College London, London WC1E 6BT, UK
- Division of Surgery and Interventional Science, Academic Head and Neck Centre University College London, London WC1E 6BT, UK
| |
Collapse
|
40
|
A Rare Adult Primary Intracranial Sarcoma, DICER1-Mutant Identified by Epigenomic Profiling: A Case Report. Brain Sci 2023; 13:brainsci13020235. [PMID: 36831778 PMCID: PMC9953897 DOI: 10.3390/brainsci13020235] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023] Open
Abstract
Diagnoses of primary malignant mesenchymal brain tumors are a challenge for pathologists. Here, we report the case of a 52-year-old man with a primary brain tumor, histologically diagnosed as a high-grade glioma, not otherwise specified (NOS). The patient underwent two neurosurgeries in several months, followed by radiotherapy and chemotherapy. We re-examined the tumor samples by methylome profiling. Methylome analysis revealed an epi-signature typical of a primary intracranial sarcoma, DICER1-mutant, an extremely rare tumor. The diagnosis was confirmed by DNA sequencing that revealed a mutation in DICER1 exon 25. DICER1 mutations were not found in the patient's blood cells, thus excluding an inherited DICER1 syndrome. The methylome profile of the DICER1 mutant sarcoma was then compared with that of a high-grade glioma, a morphologically similar tumor type. We found that several relevant regions were differentially methylated. Taken together, we report the morphological, epigenetic, and genetic characterization of the sixth described case of an adult primary intracranial sarcoma, DICER1-mutant to-date. Furthermore, this case report underscores the importance of methylome analysis to refine primary brain tumor diagnosis and to avoid misdiagnosis among morphologically similar subtypes.
Collapse
|
41
|
Ren L, Yang Y, Li W, Yang H, Zhang Y, Ge B, Zhang S, Du G, Wang J. Recent advances in epigenetic anticancer therapeutics and future perspectives. Front Genet 2023; 13:1085391. [PMID: 36685834 PMCID: PMC9845602 DOI: 10.3389/fgene.2022.1085391] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Tumor development is frequently accompanied by abnormal expression of multiple genomic genes, which can be broadly viewed as decreased expression of tumor suppressor genes and upregulated expression of oncogenes. In this process, epigenetic regulation plays an essential role in the regulation of gene expression without alteration of DNA or RNA sequence, including DNA methylation, RNA methylation, histone modifications and non-coding RNAs. Therefore, drugs developed for the above epigenetic modulation have entered clinical use or preclinical and clinical research stages, contributing to the development of antitumor drugs greatly. Despite the efficacy of epigenetic drugs in hematologic caners, their therapeutic effects in solid tumors have been less favorable. A growing body of research suggests that epigenetic drugs can be applied in combination with other therapies to increase efficacy and overcome tumor resistance. In this review, the progress of epigenetics in tumor progression and oncology drug development is systematically summarized, as well as its synergy with other oncology therapies. The future directions of epigenetic drug development are described in detail.
Collapse
Affiliation(s)
- Liwen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yihui Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hong Yang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yizhi Zhang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Binbin Ge
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Sen Zhang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, China,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China,*Correspondence: Jinhua Wang,
| |
Collapse
|