1
|
Freitas R, Peixoto A, Santos LL, Ferreira JA. Glycan-based therapeutic approaches for bladder cancer: Overcoming clinical barriers. Biochim Biophys Acta Rev Cancer 2025; 1880:189327. [PMID: 40274080 DOI: 10.1016/j.bbcan.2025.189327] [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: 01/03/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Bladder cancer (BLCA) remains a significant global health concern, being characterized by high incidence, recurrence, and mortality rates. Disease heterogeneity and rapid progression pose major challenges for effective management and identification of actionable biomarkers. Conventional therapies often fail to successfully achieve disease control, urging the development of novel, personalized approaches. In recent years, anti-tumour immunotherapy approaches in both pre-clinical and clinical settings have boomed. However, the efficacy of these strategies has been limited by the low mutational burden in some tumours, which hinders neoantigen presentation and the identification of BLCA-specific signatures. Cancer-associated aberrant glycosylation presents a unique opportunity for identifying BLCA-specific glycosignatures and developing innovative targeted therapeutics. This review provides a comprehensive overview of the clinical challenges in BLCA management and emerging novel therapies. Furthermore, it highlights the potential of glycosylation alterations as a unique opportunity for developing glycan-based therapies, potentially revolutionizing BLCA treatment strategies.
Collapse
Affiliation(s)
- Rui Freitas
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal; Health School of University Fernando Pessoa, 4249-004 Porto, Portugal; Department of Surgical Oncology, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal; CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal..
| |
Collapse
|
2
|
Li M, Gao X, Lin X, Zhang Y, Peng W, Sun T, Shu W, Shi Y, Guan Y, Xia X, Yi X, Li Y, Jia J. Analysis of germline-somatic mutational connections in colorectal cancer reveals differential tumorigenic patterns and a novel predictive marker for germline mutation carriers. Cancer Lett 2025; 620:217637. [PMID: 40118241 DOI: 10.1016/j.canlet.2025.217637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
Colorectal cancer (CRC) genetic testing of regions beyond clinical guidelines has revealed a substantial number of likely pathogenic germline mutations (GMs). It remains largely undetermined whether and how these GMs, typically located in non-mismatch repair (non-MMR) genes, are associated with the tumorigenesis of CRC. This study aimed to identify CRC-predisposing GMs among 93 cancer susceptibility genes and investigate their potential influences on CRC somatic mutational features. We secondarily aimed to investigate whether somatic ERBB2 amplification contributes to identifying GM carriers. This study incorporated a total of 3,240 Chinese CRC patients and 10,588 control individuals. CRC patients were subjected to paired tumor-normal sequencing with a 1,021-gene panel. A case-control analysis was conducted to profile the GM-associated CRC risk. A comprehensive germline-somatic association analysis was performed among 2,405 patients, with key findings subsequently validated in an independent 835-patient cohort and the TCGA CRC cohort. The case-control results supported CRC-predisposing effects of GMs in certain homologous recombination repair (HRR) and DNA damage checkpoint factor (CPF) genes, such as BRCA1/2, RecQ helicase genes, ATM, and CHEK2. HRR GMs were associated with an increased copy number alteration burden, more TP53 clonal mutations, and a higher probability of carrying somatic ERBB2 amplification. CPF GMs were inferred to have synergistic effects with ARID1A and KDM6A somatic mutations in CRC tumorigenesis. Among patients with onset age ≥55 years, stable microsatellites, and no cancer family history, ERBB2 amplification was significantly predictive of GM carriers. Our findings elucidate different germline tumorigenic patterns not driven by deficient MMR. Somatic ERBB2 amplification in CRC can serve as an indicator for germline genetic testing when traditional risk features are absent.
Collapse
Affiliation(s)
- Mintao Li
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xuan Gao
- Geneplus-Shenzhen Clinical Laboratory, Shenzhen, China
| | - Xiangchun Lin
- Department of Gastroenterology, Peking University International Hospital, Beijing, China
| | - Yan Zhang
- Geneplus-Beijing Institute, Beijing, China
| | - Wenying Peng
- The Second Department of Oncology, Yunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Center, Kunming, China
| | - Tao Sun
- General Surgery Department, Peking University Third Hospital, Beijing, China
| | - Weiyang Shu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | | | | | - Xin Yi
- Geneplus-Beijing Institute, Beijing, China.
| | - Yuan Li
- Department of Gastroenterology, Peking University International Hospital, Beijing, China; Department of Gastroenterology, Peking University Third Hospital, Beijing, China.
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China; Center for Statistical Science, Peking University, Beijing, China.
| |
Collapse
|
3
|
Su M, Duan H, Gu Y, Zhao J. Hypoxia-related signatures predicts survival, immunosuppression and PARP inhibitor resistance in HCC. Discov Oncol 2025; 16:1116. [PMID: 40518504 DOI: 10.1007/s12672-025-02923-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Accepted: 06/05/2025] [Indexed: 06/18/2025] Open
Abstract
BACKGROUND Despite extensive research on hypoxia in hepatocellular carcinoma (HCC), previous studies have relied on pre-existing hypoxia gene sets, limiting their specificity. We developed a novel approach using direct comparison of hypoxic versus normoxic HCC cell lines to establish a more precise hypoxia signature. METHODS Through differential gene expression analysis of HCC cells under controlled oxygen conditions (GSE185969) and integration with TCGA-LIHC data, we identified and validated a highly specific 29-gene hypoxia signature. We performed comprehensive immune profiling and genomic instability analyses using multi-omics approaches. RESULTS Our HCC-specific hypoxia signature demonstrated superior prognostic value (AUC: 0.805, 0.805, 0.748 at 1/3/5 years) compared to conventional hypoxia markers. High-risk tumors showed distinct immunosuppressive features including reduced CD8 + T cells and elevated Th2 cells, along with significantly increased expression of immune checkpoints CD274 (PD-1, p < 0.05) and CD276 (B7-H3, r = 0.62, p < 0.001). Notably, we uncovered an unexpected inverse relationship between hypoxia-induced genomic instability and PARP inhibitor sensitivity, challenging current therapeutic paradigms. CONCLUSION Our methodology establishes a more precise hypoxia signature specific to HCC, advancing beyond traditional approaches. The paradoxical finding of reduced PARP inhibitor sensitivity in genomically unstable tumors reveals new complexities in hypoxia-driven treatment resistance, suggesting the need for alternative therapeutic strategies in hypoxic HCC.
Collapse
Affiliation(s)
- Min Su
- Department of Oncology, Cancer Diagnosis and Therapy Research Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Oncology, Guangzhou Development District Hospital, Guangzhou, 510730, China
| | - Haibo Duan
- Department of Oncology, Cancer Diagnosis and Therapy Research Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Breast Cancer, The First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Yu Gu
- Guangzhou Institute of Cancer Research, Affiliated Cancer Hospital,Guangzhou Medical University, Guangzhou, 510000, Guangdong, China.
| | - Jianfu Zhao
- Department of Oncology, Cancer Diagnosis and Therapy Research Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Möhrmann L, Rostock L, Werner M, Oleś M, Arnold JS, Paramasivam N, Jöhrens K, Rupp L, Schmitz M, Richter D, Uhrig S, Fröhlich M, Hutter B, Hüllein J, Jahn A, Arlt M, Möhrmann EE, Hanf D, Gieldon L, Kreutzfeldt S, Heilig CE, Teleanu MV, Lipka DB, Beck K, Baude-Müller A, Mock A, Jelas I, Rieke DT, Wiesweg M, Brandts C, Boerries M, Illert AL, Desuki A, Kindler T, Krackhardt AM, Westphalen CB, Christopoulos P, Apostolidis L, Stenzinger A, Allgäuer M, Neumann O, Kerle IA, Horak P, Heining C, Grosch H, Schröck E, Hübschmann D, Fröhling S, Glimm H. Genomic landscape and molecularly informed therapy in thymic carcinoma and other advanced thymic epithelial tumors. MED 2025; 6:100612. [PMID: 40107270 DOI: 10.1016/j.medj.2025.100612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/12/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Thymic epithelial tumors (TETs) are rare malignancies with limited treatment options and underexplored molecular features. METHODS We examined the genomic landscape and therapeutic outcomes in 81 patients with advanced TETs, including thymic carcinomas (TCs), thymomas, and thymic neuroendocrine neoplasms (TNENs), who were enrolled in the MASTER trial, a prospective observational precision oncology trial. FINDINGS Using whole-genome-sequencing and whole-exome-sequencing analysis, transcriptome analysis, and methylome analysis, we identified distinct molecular features across TET subtypes, including a higher tumor mutational burden in TC and pathogenic germline variants in 18% of cases. We performed transcriptome- and methylome-based unsupervised clustering and were able to divide TCs into immunologically hot and cold subsets, with hot TCs exhibiting higher T cell infiltration and significantly longer overall survival. In 65 out of 76 (86%) patients, we recommended molecularly informed therapies, which were applied in 29 out of 65 (45%) cases, leading to a disease control rate of 62% and an objective response rate of 23% (both n = 26). The progression-free survival ratio (PFSr) was > 1.3 in 8 out of 24 (33%) patients, 7 of them having TC. Among TCs, patients achieved a mean PFSr of 1.4, indicating potential therapeutic advantages in this subgroup. The PFSr between the PFS of immune checkpoint inhibition and preceding therapies was significantly higher in the hot cluster compared to the cold cluster (median 1.7 vs. 0.3; p = 0.01945). CONCLUSIONS Our findings expand the understanding of TET biology and emphasize the role of precision oncology in informing treatment decisions and improving outcomes for patients with advanced TETs, particularly in TCs.
Collapse
Affiliation(s)
- Lino Möhrmann
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, USA; Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany.
| | - Lysann Rostock
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany
| | - Maximilian Werner
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Małgorzata Oleś
- Computational Oncology Group (CO), Molecular Precision Oncology Program (MPOP), German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Jonas S Arnold
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; ERN GENTURIS, Hereditary Cancer Syndrome Center Dresden, Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Nagarajan Paramasivam
- Computational Oncology Group (CO), Molecular Precision Oncology Program (MPOP), German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Korinna Jöhrens
- Department of Pathology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany
| | - Daniela Richter
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany
| | - Sebastian Uhrig
- Computational Oncology Group (CO), Molecular Precision Oncology Program (MPOP), German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Martina Fröhlich
- Computational Oncology Group (CO), Molecular Precision Oncology Program (MPOP), German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Barbara Hutter
- Computational Oncology Group (CO), Molecular Precision Oncology Program (MPOP), German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Jennifer Hüllein
- Computational Oncology Group (CO), Molecular Precision Oncology Program (MPOP), German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Arne Jahn
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; ERN GENTURIS, Hereditary Cancer Syndrome Center Dresden, Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marie Arlt
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; ERN GENTURIS, Hereditary Cancer Syndrome Center Dresden, Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Elena E Möhrmann
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Dorothea Hanf
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Laura Gieldon
- Institute of Medical Genetics, Carl von Ossietzky University, Oldenburg, Germany
| | - Simon Kreutzfeldt
- Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, Heidelberg, Germany
| | - Christoph E Heilig
- Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, Heidelberg, Germany
| | - Maria-Veronica Teleanu
- Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, Heidelberg, Germany
| | - Daniel B Lipka
- Section Translational Cancer Epigenomics, Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Katja Beck
- Institute of Medical Genetics, Carl von Ossietzky University, Oldenburg, Germany
| | - Annika Baude-Müller
- Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, Heidelberg, Germany; Section Translational Cancer Epigenomics, Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Andreas Mock
- Institute of Pathology, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Ivan Jelas
- Charité Comprehensive Cancer Center, Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Damian T Rieke
- Charité Comprehensive Cancer Center, Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcel Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christian Brandts
- Department of Medicine, Hematology/Oncology, University Hospital, Goethe University, Frankfurt, Germany; University Cancer Center Frankfurt (UCT), University Hospital, Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Anna L Illert
- Department of Internal Medicine I, Division of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg im Breisgau, Germany; Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Desuki
- University Cancer Center and Department of Internal Medicine III, University Medical Center Mainz, Mainz, Germany; TRON-Translational Oncology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; DKTK, partner site Frankfurt/Mainz, a partnership between DKFZ and University Medical Center Mainz, Mainz, Germany
| | - Thomas Kindler
- University Cancer Center and Department of Internal Medicine III, University Medical Center Mainz, Mainz, Germany; TRON-Translational Oncology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; DKTK, partner site Frankfurt/Mainz, a partnership between DKFZ and University Medical Center Mainz, Mainz, Germany
| | - Angela M Krackhardt
- Klinik und Poliklinik für Innere Medizin III, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - C Benedikt Westphalen
- Department of Internal Medicine III, University Hospital, LMU Munich and Comprehensive Cancer Center, Munich, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, and NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center, member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Leonidas Apostolidis
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany
| | | | - Michael Allgäuer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Olaf Neumann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Irina A Kerle
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Peter Horak
- Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, Heidelberg, Germany
| | - Christoph Heining
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Heidrun Grosch
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital, and NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center, member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Evelin Schröck
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany; Institute for Clinical Genetics, University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; ERN GENTURIS, Hereditary Cancer Syndrome Center Dresden, Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Daniel Hübschmann
- Computational Oncology Group (CO), Molecular Precision Oncology Program (MPOP), German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; Innovation and Service Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, DKFZ, Heidelberg, Germany; NCT Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Dresden, a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine, and University Hospital Carl Gustav Carus, TU Dresden University of Technology, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany; Translational Functional Cancer Genomics, DKFZ Heidelberg, Heidelberg, Germany.
| |
Collapse
|
5
|
Wei TY, Wu CC, Hsu YT, Lin WH, Tsai LC, Shieh DB, Chang CL. An exploratory analysis of driver gene alterations and their potential prognostic relevance in endometrial cancer with POLE exonuclease domain mutations. Gynecol Oncol 2025; 199:17-26. [PMID: 40516204 DOI: 10.1016/j.ygyno.2025.06.006] [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: 01/11/2025] [Revised: 05/20/2025] [Accepted: 06/05/2025] [Indexed: 06/16/2025]
Abstract
OBJECTIVE Endometrial cancer represents a significant gynecologic malignancy affecting women globally, including Taiwan. This study aimed to analyze mutation patterns in endometrial cancer genes related to POLE exonuclease domain mutations and their association with patient prognosis. METHODS We analyzed 100 endometrial cancer tissue samples from patients at MacKay Memorial Hospital (February 2014-February 2017). DNA sequencing was conducted on an Illumina MiSeq platform with subsequent QIAGEN CLC Workbench analysis. We applied Kaplan-Meier analysis and Fisher's exact test for statistical evaluations. RESULTS Mutations in POLE were primarily located in the exonuclease domain, with 36 missense variants identified, of which 19 were in this specific domain. Tumors with POLE exonuclease domain mutations (POLEmut) exhibited a significantly higher tumor mutation burden (p < 0.0001), and Kaplan-Meier analysis revealed better overall survival for patients with POLEmut tumors compared to POLE wild-type tumors (POLEwt, p < 0.0388; HR: 3.624). In POLEmut cases, tumor suppressor genes showed scattered mutations with higher nonsense rates (TP53, PIK3R1, FBXW7; all p < 0.01), contrasting with oncogenes (PIK3CA, CTNNB1, KRAS). Mutations in tumor suppressor genes (TP53, PIK3R1, FBXW7) conferred significantly better survival outcomes in POLEmut cases compared to their POLEwt counterparts with identical gene mutations (HR: 3.929-6.598, all p < 0.05). CONCLUSION This exploratory study finds that POLE exonuclease domain mutations are associated with distinct mutational patterns that vary between tumor suppressor genes and oncogenes. These observations provide hypothesis-generating insights into potential mechanisms underlying the favorable prognosis of POLEmut tumors and may inform future molecular investigations in endometrial cancer.
Collapse
Affiliation(s)
- Tsai-Yin Wei
- Department of Medical Research, MacKay Memorial Hospital, Tamsui Dist., New Taipei City, Taiwan; Institute of Organic and Polymeric Materials, National Taipei University of Technology, Taipei, Taiwan
| | - Chao-Chih Wu
- Department of Medical Research, MacKay Memorial Hospital, Tamsui Dist., New Taipei City, Taiwan; MacKay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan; Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Yun-Ting Hsu
- Department of Medical Research, MacKay Memorial Hospital, Tamsui Dist., New Taipei City, Taiwan; MacKay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Wen-Hsuan Lin
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Li-Chu Tsai
- Institute of Organic and Polymeric Materials, National Taipei University of Technology, Taipei, Taiwan
| | - Dar-Bin Shieh
- School of Dentistry and Institute of Oral Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Applied Nanomedicine and Core Facility Center, National Cheng Kung University, Tainan, Taiwan; iMANI Center of the National Core Facility for Biopharmaceuticals, National Science and Technology Council, Taipei, Taiwan; Department of Stomatology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chih-Long Chang
- Department of Medical Research, MacKay Memorial Hospital, Tamsui Dist., New Taipei City, Taiwan; Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.
| |
Collapse
|
6
|
Ventura AB, Loconte T, Negri A, Viggiano L, Fiermonte G, Ciavarella S, Guarini A, Volpe G. MAFB: a key regulator of myeloid commitment involved in hematological diseases. Cell Death Discov 2025; 11:276. [PMID: 40506426 PMCID: PMC12162849 DOI: 10.1038/s41420-025-02551-4] [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: 04/01/2025] [Revised: 05/12/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
The MAFB protein, a member of the MAF family of bZip transcription factors, plays a pivotal role in various biological processes, including cell differentiation, development, and homeostasis. Characterized by its selective expression in monocytes and macrophages, MAFB has been shown to play a crucial role during myeloid lineage differentiation, acting as a critical determinant in the transition from multipotent progenitors to fully differentiated monocytes. By modulating the expression of genes associated with immune activation and inflammation, MAFB plays a vital role in maintaining immune homeostasis and responding to pathogenic challenges. Dysregulation of MAFB expression or function has been implicated in several pathological conditions, including hematological malignancies and metabolic disorders. In particular, aberrant MAFB activity has been associated with the progression of diseases such as multiple myeloma and acute myeloid leukemia as well as other solid tumors, where it may contribute to the survival and proliferation of malignant cells, thereby promoting disease progression. MAFB and downstream targets of its transcriptional network are now being regarded as predictive biomarkers for certain types of tumors as well as being considered as potential therapeutic targets for cancer treatment. In this review, we summarize current knowledge on both physiological and pathological roles of MAFB and highlight the impact of its deregulation on hematological cancer initiation and progression.
Collapse
Affiliation(s)
| | - Tiziana Loconte
- Hematology and Cell Therapy Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Antonio Negri
- Hematology and Cell Therapy Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Luigi Viggiano
- Department of Biology, University of Bari "Aldo Moro", Bari, Italy
| | - Giuseppe Fiermonte
- Department of Bioscience, Biotechnology and Environment, University of Bari "Aldo Moro", Bari, Italy
| | - Sabino Ciavarella
- Hematology and Cell Therapy Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Attilio Guarini
- Hematology and Cell Therapy Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Giacomo Volpe
- Hematology and Cell Therapy Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
7
|
Hoda RS, Krings G. Genetic landscapes of breast tumors by next-generation sequencing with focus on less common types and genotype-phenotype correlations. Hum Pathol 2025:105826. [PMID: 40480318 DOI: 10.1016/j.humpath.2025.105826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2025] [Accepted: 06/01/2025] [Indexed: 06/11/2025]
Abstract
Next-generation sequencing (NGS) has transformed our understanding of oncogenic pathways and mutational processes underlying many breast tumors. Although large-scale NGS studies included mostly common invasive breast carcinomas, the genetic landscapes of several less common or rare special histologic types and other breast tumors have now also been elucidated. Many of these lesions harbor highly specific types of mutations or rearrangements/gene fusions, including invasive lobular carcinoma, tall cell carcinoma with reversed polarity, most salivary gland-like neoplasms, fibroepithelial neoplasms, and mesenchymal tumors such as fibromatosis, nodular fasciitis, and dermatofibrosarcoma protuberans. In some cases, surrogate immunohistochemical or RNA in situ hybridization markers evaluable by light microscopy have been shown to correlate with the underlying genetic alterations. Angiosarcomas and other special breast cancer subtypes, such as triple negative apocrine carcinomas, metaplastic carcinomas, and a subset of ER-positive carcinomas (mucinous and micropapillary carcinomas, neuroendocrine neoplasms) have not been associated with specific genetic underpinnings but are enriched for certain genetic features and oncogenic pathways. The identification of characteristic genetic alterations or their molecular surrogates can be useful to establish an accurate diagnosis, and in some cases, may point to potentially actionable therapeutic targets. This review aims to summarize the genetic landscapes of less common benign and malignant breast tumors, with special attention to genotype-phenotype correlations and to the diagnostic utility of genetics and surrogate markers when applicable. BRCA1/2-associated breast carcinomas will also be discussed due to the association of so-called BRCAness with basal-like histology.
Collapse
Affiliation(s)
- Raza S Hoda
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Department of Pathology, Cleveland Clinic, OH, United States
| | - Gregor Krings
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Department of Pathology, Cleveland Clinic, OH, United States.
| |
Collapse
|
8
|
Li Y, Tian Z, Si Z, Wang Y, Song G. Impact of TP53 Alterations on Clinical Outcomes in Penile Squamous Cell Carcinoma. Clin Genitourin Cancer 2025; 23:102323. [PMID: 40240224 DOI: 10.1016/j.clgc.2025.102323] [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: 01/26/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Penile squamous cell carcinoma (PSCC) is a rare, aggressive malignancy with a high risk of mortality due to metastasis. A comprehensive understanding of its mutational landscape is critical for improving early detection and therapeutic strategies. METHODS We analyzed tissue samples from 28 patients with PSCC treated at the Cancer Hospital of the Chinese Academy of Medical Sciences between January 2019 and March 2023. DNA from primary tumors and/or matched inguinal lymph nodes underwent targeted sequencing. Somatic mutations were profiled to compare primary and metastatic lesions. Statistical analyses assessed associations between mutational features, clinical outcomes, and treatment responses. RESULTS A total of 980 mutations were identified across 354 genes. Frequently mutated genes included TP53 (67.5%), TERT (45%), CDKN2A (40%), FAT1 (37.5%), and NOTCH1 (30%). Copy number variations (CNVs) revealed amplifications in EGFR, SOX2, and MSH6 and deletions in CDKN2B and CDKN2A. A strong correlation was observed between mutational profiles of primary and metastatic lesions (r = 0.61, P < .001). Metastatic tumors exhibited higher tumor mutational burden (TMB) than primary tumors (38.9% vs. 9.5%, P = .030) and displayed a greater prevalence of mismatch repair deficiency-associated mutational signatures. Patients with higher TP53 mutation frequencies responded more favorably to immune checkpoint inhibitors (P = .024), with treatment efficacy strongly correlated (AUC = 0.938). CONCLUSION Key mutational alterations in PSCC, including high TMB and TP53 mutations, have significant implications for early diagnosis and personalized therapies. These findings support the potential use of specific genetic markers to guide targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yajian Li
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer /Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziru Tian
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer /Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhannan Si
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer /Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yifan Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer /Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Song
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer /Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
9
|
Feng H, Jin Y, Wu B. Strategies for neoantigen screening and immunogenicity validation in cancer immunotherapy (Review). Int J Oncol 2025; 66:43. [PMID: 40342048 PMCID: PMC12101193 DOI: 10.3892/ijo.2025.5749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer immunotherapy stimulates and enhances antitumor immune responses to eliminate cancer cells. Neoantigens, which originate from specific mutations within tumor cells, are key targets in cancer immunotherapy. Neoantigens manifest as abnormal peptide fragments or protein segments that are uniquely expressed in tumor cells, making them highly immunogenic. As a result, they activate the immune system, particularly T cell‑mediated immune responses, effectively identifying and eliminating tumor cells. Certain tumor‑associated antigens that are abnormally expressed in normal host proteins in cancer cells are promising targets for immunotherapy. Neoantigens derived from mutated proteins in cancer cells offer true cancer specificity and are often highly immunogenic. Furthermore, most neoantigens are unique to each patient, highlighting the need for personalized treatment strategies. The precise identification and screening of neoantigens are key for improving treatment efficacy and developing individualized therapeutic plans. The neoantigen prediction process involves somatic mutation identification, human leukocyte antigen (HLA) typing, peptide processing and peptide‑HLA binding prediction. The present review summarizes the major current methods used for neoantigen screening, available computational tools and the advantages and limitations of various techniques. Additionally, the present review aimed to summarize experimental strategies for validating the immunogenicity of the predicted neoantigens, which will determine whether these neoantigens can effectively trigger immune responses, as well as challenges encountered during neoantigen screening, providing relevant recommendations for the optimization of neoantigen‑based immunotherapy.
Collapse
Affiliation(s)
- Hua Feng
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yuanting Jin
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, P.R. China
| | - Bin Wu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
10
|
Huang C, Qiu Z, Huang H, Xiao X, Du F, Ji J, Xu X, Jiang X, Wang Y, Gao C. Alterations in genomic features and the tumour immune microenvironment predict immunotherapy outcomes in advanced biliary tract cancer patients. Br J Cancer 2025; 132:1072-1082. [PMID: 40211026 PMCID: PMC12119827 DOI: 10.1038/s41416-025-03011-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND The response to immunotherapy is limited in advanced biliary tract cancer (BTC). Response prediction is a serious challenge in the clinic. METHODS This study included 60 patients with advanced BTC who received anti-PD-1 treatment. Among these patients, 30 were subjected to 520 gene panel sequencing, and 50 were subjected to multiplex circulating cytokine testing. The entropy and mutation features were analysed via the optimized pipeline based on our previous work. The repeated LASSO algorithm was used to identify the optimal features. The associations between sequence features and cell communications were explored by analysing single-cell transcriptome data from BTC (GSE125449). Cox regression was used to develop the integrated model. Time-dependent C-index, Kaplan‒Meier, and receiver operating characteristic (ROC) curves were used to assess the prediction performance. RESULTS TP53, NRAS, FBXW7, and APC were identified as prognosis-related genes. The average C-indices of sequence entropy (0.819) and mutation (0.817) for overall survival (OS) were significantly greater than those of tumour mutation burden (TMB, 0.392) and mutation score (0.638). Single-cell transcriptome data revealed that TP53, KRAS, and NRAS were enriched in plasmacytoid dendritic cells (pDCs) and that the communication between pDCs and macrophages was mediated through the CXCL signalling pathway. The integrated model (EM-CXCL10) showed powerful predictive performance for survival status (AUC: 0.863, 95% CI: 0.643-0.972) and objective response rate (AUC: 0.990, 95% CI: 0.822-1.000). CONCLUSIONS This study constructed a multidimensional strategy that might indicate the prognosis of BTC immunotherapy, enabling the recognition of BTC patients who would benefit from immunotherapy, thereby guiding personalized clinical decision-making.
Collapse
Affiliation(s)
- Chenjun Huang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Zhiquan Qiu
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Honglian Huang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiao Xiao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Fei Du
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Jun Ji
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Xuewen Xu
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiaoqing Jiang
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Ying Wang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Chunfang Gao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| |
Collapse
|
11
|
Simpson KL, Rothwell DG, Blackhall F, Dive C. Challenges of small cell lung cancer heterogeneity and phenotypic plasticity. Nat Rev Cancer 2025; 25:447-462. [PMID: 40211072 DOI: 10.1038/s41568-025-00803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with ~7% 5-year overall survival reflecting early metastasis and rapid acquired chemoresistance. Immunotherapy briefly extends overall survival in ~15% cases, yet predictive biomarkers are lacking. Targeted therapies are beginning to show promise, with a recently approved delta-like ligand 3 (DLL3)-targeted therapy impacting the treatment landscape. The increased availability of patient-faithful models, accumulating human tumour biobanks and numerous comprehensive molecular profiling studies have collectively facilitated the mapping and understanding of substantial intertumoural and intratumoural heterogeneity. Beyond the almost ubiquitous loss of wild-type p53 and RB1, SCLC is characterized by heterogeneously mis-regulated expression of MYC family members, yes-associated protein 1 (YAP1), NOTCH pathway signalling, anti-apoptotic BCL2 and epigenetic regulators. Molecular subtypes are based on the neurogenic transcription factors achaete-scute homologue 1 (ASCL1) and neurogenic differentiation factor 1 (NEUROD1), the rarer non-neuroendocrine transcription factor POU class 2 homeobox 3 (POU2F3), and immune- and inflammation-related signatures. Furthermore, SCLC shows phenotypic plasticity, including neuroendocrine-to-non-neuroendocrine transition driven by NOTCH signalling, which is associated with disease progression, chemoresistance and immune modulation and, in mouse models, with metastasis. Although these features pose substantial challenges, understanding the molecular vulnerabilities of transcription factor subtypes, the functional relevance of plasticity and cell cooperation offer opportunities for personalized therapies informed by liquid and tissue biomarkers.
Collapse
Affiliation(s)
- Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Dominic G Rothwell
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Oncology, Christie Hospital National Health Service, Foundation Trust, Manchester, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK.
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK.
- CRUK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
12
|
Wang S, Lv H, Zhou F. AARS1 and AARS2, the L-lactate sensors and universal lactyltransferases. Sci Bull (Beijing) 2025; 70:1547-1549. [PMID: 39730223 DOI: 10.1016/j.scib.2024.11.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Affiliation(s)
- Shuai Wang
- Children's Hospital of Soochow University, Suzhou 215025, China
| | - Haitao Lv
- Children's Hospital of Soochow University, Suzhou 215025, China
| | - Fangfang Zhou
- Children's Hospital of Soochow University, Suzhou 215025, China; Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China.
| |
Collapse
|
13
|
Beffinger M, Schellhammer L, Taskoparan B, Deplazes S, Salazar U, Tatari N, Seehusen F, von Balthazar L, Zinner CP, Spath S, Shekarian T, Ritz MF, McDaid M, Egloff P, Zimmermann I, Okada H, Ward ES, Rohrer J, Seeger MA, Buch T, Hutter G, Vom Berg J. FcRn-silencing of IL-12Fc prevents toxicity of local IL-12 therapy and prolongs survival in experimental glioblastoma. Nat Commun 2025; 16:4751. [PMID: 40404625 PMCID: PMC12098678 DOI: 10.1038/s41467-025-59971-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
Glioblastoma remains a challenging indication for immunotherapy: the blood-brain barrier hampers accessibility for systemic treatments and the immunosuppressive microenvironment impedes immune attack. Intratumoral therapy with the proinflammatory cytokine interleukin-12 (IL-12) can revert immunosuppression but leakage into the circulation causes treatment-limiting toxicity. Here we engineer an IL-12Fc fusion cytokine with reduced binding to the neonatal Fc receptor FcRn. FcRn-silenced IL-12Fc avoids FcRn-mediated brain export, thus exhibits prolonged brain retention and reduced blood levels, which prevents toxicity. In murine glioblastoma, FcRn-silenced IL-12Fc induces more durable responses with negligible systemic cytokine exposure and boosts the efficacy of radio- and chemotherapy. It triggers anti-tumor responses independently of peripheral T cell influx or lymphopenia and leads to inflammatory polarization of the tumor microenvironment in patient-derived glioblastoma explants. FcRn-silencing of IL-12Fc may unlock the full potential of IL-12 for brain cancer therapy and could be further applied to containing the activity of other therapeutics targeting neurological diseases.
Collapse
Affiliation(s)
- Michal Beffinger
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
- InCephalo AG, Allschwil, Switzerland
| | - Linda Schellhammer
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
| | - Betül Taskoparan
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
| | - Sereina Deplazes
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
- InCephalo AG, Allschwil, Switzerland
| | - Ulisse Salazar
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
| | - Nazanin Tatari
- Brain Tumor Immunotherapy and Biology Lab, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Frauke Seehusen
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, University of Zurich, Zurich, Switzerland
| | - Leopold von Balthazar
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Waedenswil, Switzerland
| | - Carl Philipp Zinner
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | | | - Tala Shekarian
- Brain Tumor Immunotherapy and Biology Lab, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Marie-Françoise Ritz
- Brain Tumor Immunotherapy and Biology Lab, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Marta McDaid
- Brain Tumor Immunotherapy and Biology Lab, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Pascal Egloff
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Iwan Zimmermann
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
- Linkster Therapeutics AG, Zurich, Switzerland
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - E Sally Ward
- Cancer Sciences Unit, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Jack Rohrer
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Waedenswil, Switzerland
| | - Markus A Seeger
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland
| | - Gregor Hutter
- Brain Tumor Immunotherapy and Biology Lab, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurosurgery, University Hospital of Basel, Basel, Switzerland
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zurich, Schlieren, Switzerland.
- InCephalo AG, Allschwil, Switzerland.
| |
Collapse
|
14
|
Wagner J, Oldenburg J, Nath N, Simm S. Explainable AI Model Reveals Informative Mutational Signatures for Cancer-Type Classification. Cancers (Basel) 2025; 17:1731. [PMID: 40507213 PMCID: PMC12153866 DOI: 10.3390/cancers17111731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/08/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: The prediction of cancer types is primarily reliant on driver genes and their specific mutations. The advancement in novel omics technologies has led to the acquisition of additional genetic data. When integrated with artificial intelligence models, there is considerable potential for this to enhance the accuracy of cancer diagnosis. As mutational signatures can provide insights into repair mechanism malfunctions, they also have the potential for more accurate cancer diagnosis. Methods: First, we compared unsupervised and supervised machine learning approaches to predict cancer types. We employed deep and artificial neural network architectures with an explainable component like layerwise relevance propagation to extract the most relevant features for the cancer-type prediction. Ten-fold cross-validation and an extensive grid search were used to optimize the neural network architecture using driver gene mutations, mutational signatures and topological mutation information as input. The PCAWG dataset was used as input to discriminate between 17 primary sites and 24 cancer types. Results: Overall, our approach showed that the most relevant mutation information to discriminate between cancer types is increased by >10% using the whole genome or intergenic and intronic genome regions instead of exome information. Furthermore, the most relevant features for most cancer types, except for two, are in the mutational signatures and not the topological mutation information. Conclusions: Informative mutational signatures outperformed the prediction of cancer types in comparison to driver gene mutations and added a new layer of diagnostic information. As the degree of information within the mutational signatures is not solely based on the frequency of occurrence, it is even possible to separate cancer types from the same primary site by the different relevant mutations. Furthermore, the comparison of informative mutational signatures allowed the cancer-type assignment of specific impaired repair mechanisms.
Collapse
Affiliation(s)
- Jonas Wagner
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.W.); (J.O.)
| | - Jan Oldenburg
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.W.); (J.O.)
| | - Neetika Nath
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.W.); (J.O.)
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, 17475 Greifswald, Germany; (J.W.); (J.O.)
- Institute of Bioanalysis, Department of Applied Sciences, Coburg University of Applied Sciences and Arts, 96450 Coburg, Germany
| |
Collapse
|
15
|
Carr A, Jackson JB, Coldren C, Chandra P, Koohestani F, Shiller M, Auber R. Tumor diagnosis recharacterization enabled by comprehensive genomic profiling to guide precision medicine strategy. NPJ Precis Oncol 2025; 9:149. [PMID: 40399445 PMCID: PMC12095656 DOI: 10.1038/s41698-025-00942-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 05/11/2025] [Indexed: 05/23/2025] Open
Abstract
Comprehensive genomic profiling (CGP) via next-generation sequencing is standard clinical practice for advanced and metastatic cancers in the U.S. and can help identify clinically actionable alterations in patients who may benefit from targeted therapies. CGP can also complement clinicopathological findings and in certain cases, may lead to diagnostic recharacterization resulting in more precise therapeutic strategies. Here, we highlight examples where molecular findings resulted in tumor re-evaluation and subsequent recharacterization. Twenty-eight cases where CGP results were inconsistent with initial pathological diagnosis and clinical presentation were selected for secondary clinicopathological review to explore alternative diagnostic explanations more consistent with the genomic results. Genomic profiling identified clinically actionable and prognostic variants leading to more accurate therapeutic recommendations based on the updated diagnoses highlighting the value of CGP beyond biomarker detection for therapy selection and supporting its complementary use in diagnostic confirmation to unveil opportunities for precision medicine strategies.
Collapse
Affiliation(s)
- Ann Carr
- PathGroup, Nashville, TN, 37217, USA.
| | | | | | | | | | | | | |
Collapse
|
16
|
Arnadottir GA, Jonsson H, Hartwig TS, Gruhn JR, Møller PL, Gylfason A, Westergaard D, Chan ACH, Oddsson A, Stefansdottir L, Roux LL, Steinthorsdottir V, Swerford Moore KH, Olafsson S, Olason PI, Eggertsson HP, Halldórsson GH, Walters GB, Stefansson H, Gudjonsson SA, Palsson G, Jensson BO, Fridriksdottir R, Petersen JF, Helgason A, Norddahl GL, Rohde PD, Saemundsdottir J, Magnusson OT, Halldorsson BV, Bliddal S, Banasik K, Gudbjartsson DF, Nyegaard M, Sulem P, Thorsteinsdottir U, Hoffmann ER, Nielsen HS, Stefansson K. Sequence diversity lost in early pregnancy. Nature 2025:10.1038/s41586-025-09031-w. [PMID: 40399685 DOI: 10.1038/s41586-025-09031-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/16/2025] [Indexed: 05/23/2025]
Abstract
Every generation, the human genome is shuffled during meiosis and a single fertilized egg gives rise to all of the cells of the body1. Meiotic errors leading to chromosomal abnormalities are known causes of pregnancy loss2,3, but genetic aetiologies of euploid pregnancy loss remain largely unexplained4. Here we characterize sequence diversity in early pregnancy loss through whole-genome sequencing of 1,007 fetal samples and 934 parental samples from 467 trios affected by pregnancy loss (fetus, mother and father). Sequenced parental genomes enabled us to determine both the parental and meiotic origins of chromosomal abnormalities, detected in half of our set. It further enabled us to assess de novo mutations on both homologous chromosomes from parents transmitting extra chromosomes, and date them, revealing that 6.6% of maternal mutations occurred before sister chromatid formation in fetal oocytes. We find a similar number of de novo mutations in the trios affected by pregnancy loss as in 9,651 adult trios, but three times the number of pathogenic small (<50 bp) sequence variant genotypes in the loss cases compared with adults. Overall, our findings indicate that around 1 in 136 pregnancies is lost due to a pathogenic small sequence variant genotype in the fetus. Our results highlight the vast sequence diversity that is lost in early pregnancy.
Collapse
Affiliation(s)
| | | | - Tanja Schlaikjær Hartwig
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Jennifer R Gruhn
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Loof Møller
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | - David Westergaard
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Andrew Chi-Ho Chan
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jesper Friis Petersen
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital-North Zealand, Hillerød, Denmark
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | | | | | - Palle Duun Rohde
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | | | - Bjarni V Halldorsson
- deCODE genetics/Amgen, Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | - Sofie Bliddal
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Karina Banasik
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Mette Nyegaard
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | | | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eva R Hoffmann
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Henriette Svarre Nielsen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavik, Iceland.
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
17
|
Rebello RJ, Posner A, Dong R, Prall OWJ, Sivakumaran T, Mitchell CB, Flynn A, Caneborg A, Mitchell C, Kanwal S, Fedele C, Webb S, Fisher K, Wong HL, Balachander S, Zhu W, Nicolson S, Dimitriadis V, Wilcken N, DeFazio A, Gao B, Singh M, Collins IM, Steer C, Warren M, Karanth N, Xu H, Fellowes A, Hicks RJ, Stewart KP, Shale C, Priestley P, Dawson SJ, Vissers JHA, Fox SB, Schofield P, Bowtell D, Hofmann O, Grimmond SM, Mileshkin L, Tothill RW. Whole genome sequencing improves tissue-of-origin diagnosis and treatment options for cancer of unknown primary. Nat Commun 2025; 16:4422. [PMID: 40393956 PMCID: PMC12092688 DOI: 10.1038/s41467-025-59661-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
Genomics can inform both tissue-of-origin (TOO) and precision treatments for patients with cancer of unknown primary (CUP). Here, we use whole genome and transcriptome sequencing (WGTS) for 72 patients and show diagnostic superiority of WGTS over panel testing (386-523 genes) in 71 paired cases. WGTS detects all reportable DNA features found by panel as well as additional mutations of diagnostic or therapeutic relevance in 76% of cases. Curated WGTS features and a CUP prediction algorithm (CUPPA) trained on WGTS data of known cancer types informs TOO in 71% of cases otherwise undiagnosed by clinicopathology review. WGTS informs treatments for 79% of patients, compared to 59% by panel testing. Finally, WGS of cell-free DNA (cfDNA) from patients with a high cfDNA tumour fraction (>7%), enables high-likelihood CUPPA predictions in 41% of cases. WGTS is therefore superior to panel testing, broadens treatment options, and is feasible using routine pathology samples and cfDNA.
Collapse
Affiliation(s)
- Richard J Rebello
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Atara Posner
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Ruining Dong
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Owen W J Prall
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Tharani Sivakumaran
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Camilla B Mitchell
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Aidan Flynn
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Alex Caneborg
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sehrish Kanwal
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Clare Fedele
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Samantha Webb
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Krista Fisher
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Hui-Li Wong
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Shiva Balachander
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Wenying Zhu
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Shannon Nicolson
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Voula Dimitriadis
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Nicholas Wilcken
- The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Anna DeFazio
- The Westmead Institute for Medical Research, Sydney, NSW, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Bo Gao
- Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
| | - Madhu Singh
- Department of Medical Oncology, Barwon Health Cancer Services, Geelong, VIC, Australia
| | - Ian M Collins
- Department of Medical Oncology, Southwest HealthCare, Warrnambool and Deakin University, Geelong, VIC, Australia
| | - Christopher Steer
- Border Medical Oncology, Albury Wodonga Regional Cancer Centre, Albury NSW, Australia and UNSW School of Clinical Medicine, Rural Clinical Campus, Albury, NSW, Australia
| | - Mark Warren
- Department of Medical Oncology, Bendigo Health, Bendigo, VIC, Australia
| | - Narayan Karanth
- Division of Medicine, Alan Walker Cancer Centre, Darwin, NT, Australia
| | - Huiling Xu
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Andrew Fellowes
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rodney J Hicks
- The St Vincent's Hospital Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Kym Pham Stewart
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | | | | | - Sarah-Jane Dawson
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Joseph H A Vissers
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Penelope Schofield
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Psychology, and Iverson Health Innovation Research Institute, Swinburne University, Melbourne, VIC, Australia
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, VIC, Australia
| | - David Bowtell
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Oliver Hofmann
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Sean M Grimmond
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Linda Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Richard W Tothill
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia.
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
18
|
Song Y, Liu T, Hao Q, Fang Q, Gong X, Li Y, Tian Z, Wei H, Wang M, Wang J, Cheng T, Mi Y. Comprehensive omics-based classification system in adult patients with B-cell acute lymphoblastic leukemia. Mol Oncol 2025. [PMID: 40388565 DOI: 10.1002/1878-0261.70053] [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: 12/05/2024] [Revised: 02/06/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is a highly heterogeneous disease with a challenging prognosis, particularly in adult patients. We enrolled 88 adult B-ALL patients with transcriptomic and mutation profiles for classification system identification, and a comprehensive system for B-ALL patients (COMBAT) was developed. COMBAT stratified patients into three cohorts: (1) COMBAT1, characterized by high stem/myeloid antigen expression, low immune infiltration, high infiltration of endothelial cells, and hypo-CIMP (CpG island methylator phenotype); (2) COMBAT2, defined as an inflamed subtype with immune exhaustion, moderate myeloid antigen expression, and hypo-CIMP; and (3) COMBAT3, marked by proliferative profiles with MYC pathway activation and hypomethylation at enhancer regions in patients characterized by CIMP. The molecular features of the three COMBATs were verified in two external cohorts, the GSE34861 (N = 194) and GSE66005 (N = 109) datasets. In univariate analysis, only COMBAT classification presented significance for OS, and patients of COMBAT3 presented significantly superior survival than COMBAT1/2 in Ph-negative ALL. Ph-negative ALL patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) in the COMBAT3 group showed better overall survival (OS) than those in the COMBAT1-2 groups (estimated 3-year OS: 100% vs. 65.6%, P = 0.034), suggesting a prognostic benefit of this subtype. In summary, the COMBAT system redefines the characteristics of adult B-ALL subtypes and guides the selection of allo-HSCT for Ph-negative patients.
Collapse
Affiliation(s)
- Yang Song
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Qishan Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qiuyun Fang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaoyuan Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Hui Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yingchang Mi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
19
|
Park J, Joung JG, Lim MC, Lee J, Kim BG, Kim JW, Shin SJ, Kim S, Park E, Choi CH, Kim HS, Park SY, Lee JY. Neoadjuvant Chemotherapy with Dual Immune Checkpoint Inhibitors for Advanced-Stage Ovarian Cancer: Final Analysis of TRU-D Phase II Nonrandomized Clinical Trial. Clin Cancer Res 2025; 31:1865-1876. [PMID: 40043003 DOI: 10.1158/1078-0432.ccr-24-3753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/12/2025] [Accepted: 03/03/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE This open-label, investigator-initiated, phase II study was conducted to evaluate the safety, survival, and neoadjuvant outcomes of neoadjuvant chemotherapy (NAC) combined with dual immune checkpoint inhibitors in advanced-stage epithelial ovarian cancer (EOC). PATIENTS AND METHODS Between June 2019 and July 2021, 45 patients with unresectable stage III to IV EOC were enrolled. The patients received three cycles of NAC combined with durvalumab and tremelimumab. All patients underwent interval debulking surgery and received three cycles of durvalumab and adjuvant chemotherapy, followed by 12 cycles of durvalumab as maintenance therapy. The primary endpoint was the 12-month progression-free survival (PFS) rate; the secondary endpoints were the objective response rate after NAC, a chemotherapy response score, pathologic complete response, overall survival, and safety. The preplanned exploratory analyses assessed the lymphocyte infiltration, PD-L1 expression, and genomic profiles of pretreatment tumors. RESULTS The 12-month PFS rate was 65.9% [95% confidence interval (CI), 52.8-not estimated (NE)], whereas the 24- and 30-month PFS rates were 38.6% (95% CI, 26.7-NE) and 36.4% (95% CI, 24.7-NE), respectively. After NAC, the objective response rate was 86.7%, whereas 14 patients (31.1%) had a chemotherapy response score of three, and five (11.1%) achieved pathologic complete response. The 30-month overall survival rate was 87.7%. The most common grade ≥3 adverse event was neutropenia (26.7%). In an exploratory analysis, patients with pre-NAC tumors showing PD-L1 (combined positive score) ≥1, high Mutation Signature 3, and a high extracellular matrix signature demonstrated improved PFS outcomes. CONCLUSIONS NAC combined with dual immune checkpoint inhibitors is feasible for advanced-stage EOC and shows promising activity with a durable clinical response.
Collapse
Affiliation(s)
- Junsik Park
- Department of Obstetrics and Gynecology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Je-Gun Joung
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, South Korea
| | - Jungbok Lee
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Byoung-Gie Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - So Jin Shin
- Department of Obstetrics and Gynecology, Keimyung University Dongsan Medical Center, Daegu, South Korea
| | - Sunghoon Kim
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunhyang Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Yoon Park
- Center for Gynecologic Cancer, National Cancer Center, Goyang, South Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
20
|
Mortusewicz O, Haslam J, Gad H, Helleday T. Uracil-induced replication stress drives mutations, genome instability, anti-cancer treatment efficacy, and resistance. Mol Cell 2025; 85:1897-1906. [PMID: 40378828 DOI: 10.1016/j.molcel.2025.04.015] [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: 01/15/2025] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 05/19/2025]
Abstract
Uracil incorporation into DNA, as a result of nucleotide pool imbalances or cytosine deamination (e.g., through APOBEC3A/3B), can result in replication stress and is the most common source of mutations in cancer and aging. Despite the critical role of uracil in genome instability, cancer development, and cancer therapy, only now is there emerging data on its impact on fundamental processes such as DNA replication and genome stability. Removal of uracil from DNA by base excision repair (BER) can generate a DNA single-strand break (SSB), which can trigger homologous recombination (HR) repair or replication fork collapse and cell death. Unprocessed uracil can also induce replication stress directly and independently of BER by slowing down replication forks, leading to single-stranded DNA (ssDNA) gaps. In this perspective, we review how genomic uracil induces replication stress, the therapeutic implications of targeting uracil-induced vulnerabilities, and potential strategies to exploit these mechanisms in cancer treatment.
Collapse
Affiliation(s)
- Oliver Mortusewicz
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna 171 65, Sweden
| | - James Haslam
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna 171 65, Sweden
| | - Helge Gad
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna 171 65, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Solna 171 65, Sweden.
| |
Collapse
|
21
|
Feng H, Zhao LY, Xu Z, Xie QF, Deng HJ, Yu J, Liu H. Homologous recombination deficiency and immunotherapy response in microsatellite-stable colorectal cancer: Evidence from a cohort study in China. World J Gastrointest Oncol 2025; 17:102767. [DOI: 10.4251/wjgo.v17.i5.102767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/14/2025] [Accepted: 03/07/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Patients with colorectal cancer (CRC) exhibiting microsatellite instability (MSI)-high generally demonstrate a favorable response to immunotherapy. In contrast, the efficacy of immunotherapy in microsatellite-stable (MSS) CRC patients is considerably restricted. This study sought to evaluate the effectiveness of immunotherapy in MSS patients characterized by homologous recombination deficiency (HRD) as opposed to those with homologous recombination proficiency (HRP).
AIM To investigate and compare the clinicopathological characteristics, treatment modalities, and outcomes between the HRD and HRP groups in CRC.
METHODS Next-generation sequencing was performed on 268 CRC patients to identify tumor-associated genetic alterations and assess their HRD scores and MSI status. Patients with HRD-related gene alterations or an HRD score ≥ 30 were classified into the HRD group, while the remaining patients were assigned to the HRP group. Clinical data, including staging and treatment regimens, were collected for analysis. Cox regression and Kaplan-Meier survival curves were employed to evaluate whether the HRD group demonstrated improved survival outcomes following immunotherapy treatment.
RESULTS Among the 268 patients, 64 were classified into the HRD group, which had a higher proportion of early-stage CRC diagnoses compared to the HRP group. Kaplan-Meier survival curves indicated significantly better survival rates in the HRD group compared to the HRP group across all cohorts, as well as among MSS patients treated with immunotherapy (P < 0.05).
CONCLUSION This study demonstrates that CRC patients with HRD have a more favorable prognosis and suggests that HRD status could serve as a predictive marker for immunotherapy response in MSS patients.
Collapse
Affiliation(s)
- Hao Feng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Li-Ying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Zhou Xu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Qing-Feng Xie
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hai-Jun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
22
|
Weijers DD, Hinić S, Kroeze E, Gorris MA, Schreibelt G, Middelkamp S, Mensenkamp AR, Bladergroen R, Verrijp K, Hoogerbrugge N, Wesseling P, van der Post RS, Loeffen JL, Gidding CE, van Kouwen MC, de Vries IJM, van Boxtel R, de Voer RM, Jongmans MC, Kuiper RP. Unraveling mutagenic processes influencing the tumor mutational patterns of individuals with constitutional mismatch repair deficiency. Nat Commun 2025; 16:4459. [PMID: 40368937 PMCID: PMC12078508 DOI: 10.1038/s41467-025-59775-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
Constitutional mismatch repair deficiency (CMMRD), caused by bi-allelic germline variants in mismatch repair (MMR) genes, is associated with high cancer incidence early in life. A better understanding of mutational processes driving sequential CMMRD tumors can advance optimal treatment. Here, we describe a genomic characterization on a representative collection of CMMRD-associated tumors consisting of 41 tumors from 17 individuals. Mutational patterns in these tumors appear to be influenced by multiple factors, including the affected MMR gene and tumor type. Somatic polymerase proofreading mutations, commonly present in brain tumors, are also found in a T-cell lymphoblastic lymphoma displaying associated mutational patterns. We show prominent mutational patterns in two second primary hematological malignancies after temozolomide treatment. Furthermore, an indel signature, characterized by one-base pair cytosine insertions in cytosine homopolymers, is found in 54% of tumors. In conclusion, analysis of sequential CMMRD tumors reveals diverse mutational patterns influenced by the affected MMR gene, tumor type and treatment history.
Collapse
Affiliation(s)
- Dilys D Weijers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Snežana Hinić
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Emma Kroeze
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Mark Aj Gorris
- Department of Medical BioSciences, Radboud university medical center, Nijmegen, The Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud university medical center, Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Medical BioSciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Sjors Middelkamp
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Arjen R Mensenkamp
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Reno Bladergroen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Kiek Verrijp
- Department of Medical BioSciences, Radboud university medical center, Nijmegen, The Netherlands
- Division of Immunotherapy, Oncode Institute, Radboud university medical center, Nijmegen, The Netherlands
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Amsterdam, The Netherlands
| | - Rachel S van der Post
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - Jan Lc Loeffen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Corrie Em Gidding
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Mariëtte Ca van Kouwen
- Department of Gastroenterology and Hepatology, Radboud university medical center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Medical BioSciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Richarda M de Voer
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Marjolijn Cj Jongmans
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roland P Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
23
|
Galvez-Cancino F, Navarrete M, Beattie G, Puccio S, Conde-Gallastegi E, Foster K, Morris Y, Sahwangarrom T, Karagianni D, Liu J, Lee AJX, Garyfallos DA, Simpson AP, Mastrokalos GT, Nannini F, Costoya C, Anantharam V, Cianciotti BC, Bradley L, Garcia-Diaz C, Clements M, Shroff A, Vahid Dastjerdi F, Rota EM, Sheraz S, Bentham R, Uddin I, Walczak H, Lladser A, Reading JL, Chester KA, Pule MA, Brennan PM, Marguerat S, Parrinello S, Peggs KS, McGranahan N, Lugli E, Litchfield K, Pollard SM, Quezada SA. Regulatory T cell depletion promotes myeloid cell activation and glioblastoma response to anti-PD1 and tumor-targeting antibodies. Immunity 2025; 58:1236-1253.e8. [PMID: 40280128 DOI: 10.1016/j.immuni.2025.03.021] [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: 06/26/2023] [Revised: 10/28/2024] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Glioblastoma is invariably lethal and responds poorly to immune checkpoint blockade. Here, we examined the impact of regulatory T (Treg) cell depletion on glioblastoma progression and immunotherapy responsiveness. In human glioblastoma, elevated Treg cell signatures correlated with poorer survival outcomes, with these cells expressing high levels of CD25. In Nf1-/-Pten-/-EGFRvIII+ glioblastoma-bearing mice, a single dose of non-interleukin-2 (IL-2) blocking (NIB) anti-CD25 (anti-CD25NIB) antibody depleted Treg cells and promoted CD8+ T cell clonal expansion and partial tumor control, further enhanced by programmed cell death-1 (PD1)-blockade. Treg cell depletion induced interferon-γ (IFN-γ)-dependent tumor microenvironment remodeling, increasing Fcγ receptor (FcγR) expression on intratumoral myeloid cells and enhancing phagocytosis. Combination of anti-CD25NIB with anti-EGFRvIII tumor-targeting antibodies resulted in complete tumor control. Anti-human CD25NIB treatment of glioblastoma patient-derived tumor fragments effectively depleted Treg cells and activated CD8+ T cells. These findings underscore the therapeutic relevance of Treg targeting in glioblastoma and unveil potent combination strategies for anti-CD25NIB based on innate cell activation.
Collapse
Affiliation(s)
- Felipe Galvez-Cancino
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK; Immune Regulation Laboratory, Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Mariela Navarrete
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Gordon Beattie
- CRUK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK; Bioinformatics Hub, UCL Cancer Institute, University College London, London, UK
| | - Simone Puccio
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Enrique Conde-Gallastegi
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Kane Foster
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Yasmin Morris
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Teerapon Sahwangarrom
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Despoina Karagianni
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Jiali Liu
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alvin J X Lee
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Dimitrios A Garyfallos
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alexander P Simpson
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Gerasimos-Theodoros Mastrokalos
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Francesco Nannini
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Cristobal Costoya
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Varshaa Anantharam
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | | | - Leanne Bradley
- Centre for Regenerative Medicine and Institute for Regeneration and Repair, & Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Claudia Garcia-Diaz
- Neurogenesis and Brain Cancer Group, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Melanie Clements
- Neurogenesis and Brain Cancer Group, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Aditya Shroff
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, London WC1E 6DD, UK
| | | | - Enrique Miranda Rota
- Recombinant Antibody Therapeutics Group, UCL Cancer Institute, London WC1E 6DD, UK
| | - Shahida Sheraz
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Robert Bentham
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Imran Uddin
- CRUK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, London WC1E 6DD, UK; Institute of Biochemistry I & CECAD Cluster of Excellence, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Alvaro Lladser
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - James L Reading
- Pre-Cancer Immunology Laboratory, Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kerry A Chester
- Recombinant Antibody Therapeutics Group, UCL Cancer Institute, London WC1E 6DD, UK
| | - Martin A Pule
- Research Department of Haematology, Cancer Institute, University College London, Paul O'Gorman Building, London WC1E 6DD, UK
| | - Paul M Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Samuel Marguerat
- Bioinformatics Hub, UCL Cancer Institute, University College London, London, UK
| | - Simona Parrinello
- Neurogenesis and Brain Cancer Group, Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6DD, UK
| | - Karl S Peggs
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Nicholas McGranahan
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Enrico Lugli
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Kevin Litchfield
- The Tumour Immunogenomics and Immunosurveillance (TIGI) Lab, UCL Cancer Institute, London WC1E 6DD, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine and Institute for Regeneration and Repair, & Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Sergio A Quezada
- Immune Regulation and Tumour Immunotherapy Laboratory, Cancer Immunology Unit, Research Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK.
| |
Collapse
|
24
|
Zhu Y, Gao Y, Huang X, Chen B, Wang X, Wu Y, Sun J, Huang X. Pan-cancer analysis reveals TRA16 as a master regulator of human carcinogenesis. Front Oncol 2025; 15:1543419. [PMID: 40432923 PMCID: PMC12106028 DOI: 10.3389/fonc.2025.1543419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction The nuclear receptor TR4 binding protein, TRA16, has been implicated in lung carcinogenesis; however, its broader role across diverse human cancers remains poorly understood. Understanding TRA16's involvement in cancer biology could uncover novel regulatory mechanisms and potential therapeutic targets. Methods We conducted a comprehensive pan-cancer analysis of TRA16 expression and function across multiple human malignancies. Gene co-expression networks, pathway enrichment, transcription factor analysis, organoid modeling, and intercellular communication profiling were employed. Tumor mutation burden (TMB) and microenvironmental features were also assessed in relation to TRA16 expression, stratified by TP53 mutation status. Results Correlation analysis identified the cell cycle as the top enriched pathway among TRA16-associated genes, with key transcription factors, including RB-E2F, MYC, and TP53, regulating genes co-expressed with TRA16. In liver cancer organoid models, TRA16 and its co-expressed genes were significantly upregulated. Intercellular communication analysis showed that TRA16-positive cells exhibited increased autocrine signaling and overall signaling activity. Importantly, patients with high TRA16 expression demonstrated elevated TMB and decreased stromal and immune features. Discussion These findings highlight TRA16 as a potential master regulator of oncogenic processes, contributing to tumor progression through coordinated regulation of cell cycle genes, intercellular signaling, and genomic instability. Our results provide new insights into TRA16's role across cancers and support its potential as a novel oncogene.
Collapse
Affiliation(s)
- Yanyan Zhu
- Department of Oncology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Yike Gao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoqing Huang
- Intelliphecy Center for Systems Medicine, Intelliphecy, Shenzhen, China
| | - Bowang Chen
- Intelliphecy Center for Systems Medicine, Intelliphecy, Shenzhen, China
- Department of Data Science, Intelliphecy, Nanjing, China
| | - Xinyi Wang
- Department of Dermatology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Ying Wu
- Department of Obstetrics and Gynecology, Southern University of Science and Technology Yantian Hospital, Shenzhen, China
| | - Jian Sun
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoyun Huang
- Intelliphecy Center for Systems Medicine, Intelliphecy, Shenzhen, China
- College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
25
|
Jazayeri B, Li R. Personalized neoantigen vaccines in urologic malignancies. NATURE CANCER 2025:10.1038/s43018-025-00944-z. [PMID: 40346293 DOI: 10.1038/s43018-025-00944-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Affiliation(s)
- Behzad Jazayeri
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Roger Li
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
26
|
Xu Y, Luo H, Wang J, Liu H, Chen L, Ji H, Deng Z, Liu X. CD103 + T Cells Eliminate Damaged Alveolar Epithelial Type II Cells Under Oxidative Stress to Prevent Lung Tumorigenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503557. [PMID: 40344646 DOI: 10.1002/advs.202503557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/20/2025] [Indexed: 05/11/2025]
Abstract
The nexus between aging-associated immune deteriorations and tumorigenesis of lung cancers remains elusive. In a mouse model with Med23 depletion in T cells (Med23 -/-), it is found a strong association between the decline of CD103+ T cells and spontaneous alveolar epithelial type II cell (AT2 cell)-originated lung adenocarcinomas. The reduction of CD103+ T cells in the lung results in an accumulation of AT2 cells bearing oxidative damages, which appears to be the major origin of the lung adenocarcinoma. Functional experiments reveal CD103+ T cells can eradicate oxidative-damage-bearing AT2 cells as well as ROS-dependent, KRAS (G12D)-driven tumorigenesis. In vitro co-cultures prove CD103+ T cells, especially CD103+ CD8+ T cells, exhibit a killing capacity that matches the oxidative stress level in the target cells. In aged animals, it is found the abundance of CD103+ CD8+ T cells in the lung declines with age, accompanied by an accumulation of oxidative-damage-bearing AT2 cells. Collectively, the study establishes the vital function of CD103+ T cells in surveilling epithelial cells under oxidative stress to prevent malignancies, and unravels a potential immuno-dysregulation in the aged lung which contributes to tumorigenesis.
Collapse
Affiliation(s)
- Yu Xu
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Haorui Luo
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Jiahao Wang
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Haifeng Liu
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Luonan Chen
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Hongbin Ji
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zimu Deng
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
- Zhongshan Institute for Drug discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Xiaolong Liu
- Key Laboratory of Multicellular Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China
- School of Life Science and Technology, ShanghaiTech University, 319 Yueyang Road, Shanghai, 200031, China
| |
Collapse
|
27
|
Zhang J, Song Z, Zhang Y, Zhang C, Xue Q, Zhang G, Tan F. Recent advances in biomarkers for predicting the efficacy of immunotherapy in non-small cell lung cancer. Front Immunol 2025; 16:1554871. [PMID: 40406096 PMCID: PMC12095235 DOI: 10.3389/fimmu.2025.1554871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/18/2025] [Indexed: 05/26/2025] Open
Abstract
Lung cancer continues to be the primary cause of cancer-related deaths globally, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all instances. Recently, immune checkpoint inhibitors (ICIs) have transformed the treatment approach for NSCLC, however, only a subset of patients experiences significant benefits. Therefore, identifying reliable biomarkers to forecast the efficacy of ICIs is crucial for ensuring the safety and effectiveness of treatments, becoming a major focus of current research efforts. This review highlights the recent advances in predictive biomarkers for the efficacy of ICIs in the treatment of NSCLC, including PD-L1 expression, tertiary lymphoid structures (TLS), tumor-infiltrating lymphocytes (TILs), tumor genomic alterations, transcriptional signatures, circulating biomarkers, and the microbiome. Furthermore, it underscores the pivotal roles of liquid biopsy, sequencing technologies, and digital pathology in biomarker discovery. Special attention is given to the predictive value of TLS, circulating biomarkers, and transcriptional signatures. The review concludes that the integration of multiple biomarkers holds promise for achieving more accurate efficacy predictions and optimizing personalized immunotherapy strategies. By providing a comprehensive overview of the current progress, this review offers valuable insights into biomarker-based precision medicine for NSCLC and outlines future research directions.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zehao Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanjie Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chentong Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
28
|
Saad A, Taylor A, Felder S, Helpman L, Bauer S, Shapira R, Levanon K, Korach J, Atamneh R, Breslauer S, Goldstein J, Peleg Hasson S. De-escalating first-line treatment in stage IVB or recurrent cervical cancer: outcomes of immunotherapy alone and systemic review. Oncologist 2025; 30:oyaf096. [PMID: 40421960 PMCID: PMC12107546 DOI: 10.1093/oncolo/oyaf096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 03/23/2025] [Indexed: 05/28/2025] Open
Abstract
INTRODUCTION Chemo-immunotherapy (IO) is the preferred first-line treatment for stage IVB or recurrent cervical cancer. However, limited data exist on the efficacy and safety of using IO-alone as a de-escalation strategy. We report outcomes from a case series of selected patients treated with IO-alone and review the feasibility of de-escalating first-line treatment. METHODS The authors conducted a literature review using Google Scholar and PubMed to identify reports using IO-alone as a de-escalation strategy across malignancies published between 1999 and December 2024 and also reviewed a cervical cancer database from a tertiary academic to identify patients with stage IVB or recurrent disease treated with IO-alone. The authors used the Kaplan-Meier method to estimate progression-free survival (PFS) and overall survival (OS). RESULTS Among 582 patients treated between 2015 and 2021, 18 met the inclusion criteria. The median age was 43 years (range 28-84); 67% had squamous cell carcinoma, 11% adenocarcinoma, and 80% expressed PD-L1. CPS scores were <1 in 20%, 1--10 in 33%, and >10 in 47%. Most patients had oligo-metastatic disease (83%). Treatment with IO-alone began a median of 7 months after platinum-based chemotherapy. Indications included prior adjuvant (44%) or neoadjuvant (22%) chemotherapy, clinical trial participation (11%), or patient preference (22%). Median PFS and OS were 27 months and 82 months, respectively. CONCLUSIONS These findings support the need for clinical trials evaluating IO-alone as a first-line treatment option for de-escalation in stage IVB or recurrent cervical cancer. Biomarker development is needed to better identify candidates for personalized therapy.
Collapse
Affiliation(s)
- Akram Saad
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Sheba Cancer Center and Institute of Oncology, Tel-Hashomer, Derech Sheba 2, Ramat Gan, Israel
| | - Alexandra Taylor
- The Royal Marsden, Department of Clinical Oncology, 203 Fulham Rd., London SW3 6JJ, United Kingdom
| | - Shira Felder
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Sheba Cancer Center and Institute of Oncology, Tel-Hashomer, Derech Sheba 2, Ramat Gan, Israel
| | - Limor Helpman
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Gynecologic Oncology, Sheba Medical Center, Tel-Hashomer, Derech Sheba 2, Ramat Gan, Israel
| | - Smadar Bauer
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Sheba Cancer Center and Institute of Oncology, Tel-Hashomer, Derech Sheba 2, Ramat Gan, Israel
| | - Ronnie Shapira
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Sheba Cancer Center and Institute of Oncology, Tel-Hashomer, Derech Sheba 2, Ramat Gan, Israel
| | - Keren Levanon
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Sheba Cancer Center and Institute of Oncology, Tel-Hashomer, Derech Sheba 2, Ramat Gan, Israel
| | - Jacob Korach
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Gynecologic Oncology, Sheba Medical Center, Tel-Hashomer, Derech Sheba 2, Ramat Gan, Israel
| | - Ronza Atamneh
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
| | - Samantha Breslauer
- Rappaport Faculty of Medicine, Technion Israel Institute of Technology, 1 Efron St. Bat Galim, Haifa 3525433, Israel
| | - Jeffrey Goldstein
- Tel Aviv Sourasky Medical Center, Department of Radiation Oncology, Weizmann St 6, Tel Aviv-Yafo, Israel
| | - Shira Peleg Hasson
- Faculty of Medicine, Tel Aviv University, P.O.B 39040 Ramat Aviv, Tel Aviv 69978, Israel
- Tel Aviv Sourasky Medical Center, Department of Medical Oncology, Weizmann St 6, Tel Aviv-Yafo, Israel
| |
Collapse
|
29
|
Lamberti G, Rihawi K, Mazzoni F, Riccardi F, Follador A, Tiseo M, Frassoldati A, Colantonio I, Bonetti A, Genova C, Giardina D, Bertolini F, Cinieri S, Pasello G, Brighenti M, Andrini E, Tognetto M, Boni L, Ardizzoni A. Carboplatin, etoposide, atezolizumab, and bevacizumab in the first-line treatment of patients with extensive stage small-cell lung cancer: the GOIRC-01-2019 CeLEBrATE study. J Immunother Cancer 2025; 13:e010694. [PMID: 40341031 PMCID: PMC12067786 DOI: 10.1136/jitc-2024-010694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND The addition of a programmed death-ligand 1 (PD-L1) inhibitor, either atezolizumab or durvalumab, to platinum-etoposide prolonged survival in a limited subset of patients with extensive-stage small-cell lung cancer (ES-SCLC). Preclinical studies demonstrated synergistic antitumor activity of combined vascular endothelial growth factor receptor and PD-L1 inhibition in SCLC. Since bevacizumab added to platinum-etoposide was safe and active in ES-SCLC, we investigated the efficacy of atezolizumab, bevacizumab, carboplatin, and etoposide as first-line treatment of ES-SCLC. METHODS The CeLEBrATE study is an Italian multicentric single-arm phase II trial of carboplatin (area under the curve 5 ml/min), etoposide (100 mg/sqm), bevacizumab (7.5 mg/kg), and atezolizumab (1,200 mg) every 3 weeks (q3w) for four to six courses, followed by bevacizumab and atezolizumab maintenance q3w in patients with ES-SCLC and no contraindications to immunotherapy or antiangiogenic therapy. Patients with asymptomatic brain metastases were eligible. Prophylactic cranial irradiation and consolidation thoracic external radiotherapy were not permitted while on study treatment. Primary endpoint was overall survival (OS) rate at 1 year. RESULTS 53 patients were enrolled (45.3% women, median age 65 years) and received at least one dose of study treatment. At a median follow-up time of 23.4 months (95% CI: 21.1 to 26.0), the 1-year OS rate was 61.8% (90% CI: 50.7% to 72.8%; p=0.04), with a median OS of 12.9 months (95% CI: 11.6 to 17.5). Median progression-free survival was 6.2 months (95% CI: 5.4 to 6.6) and objective response rate was 83.3% (95% CI: 69.8% to 92.5%). Grade 3-4 adverse events were reported in 34 patients (64.2%) leading to dose reductions in 24 (45.3%), and dose delays in 39 (73.9%) and 32 (69.6%) during the induction and maintenance phase, respectively. 19 (35.8%) treatment-related serious adverse events were reported. CONCLUSION The CeLEBrATE study met its primary objective demonstrating a signal of efficacy of bevacizumab, atezolizumab, carboplatin, and etoposide in the first-line treatment of patients with ES-SCLC. TRIAL REGISTRATION NUMBER GOIRC-01-2019 ML41241, Eudract Number: 2019-003798-2.
Collapse
Affiliation(s)
- Giuseppe Lamberti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| | - Karim Rihawi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| | - Francesca Mazzoni
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, Department of Oncology, Careggi University Hospital, Firenze, Italy
| | - Ferdinando Riccardi
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, Azienda Ospedaliera Cardarelli, Napoli, Italy
| | - Alessandro Follador
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Oncology, University Hospital Santa Maria Della Misericordia, Udine, Italy
| | - Marcello Tiseo
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Medicine and Surgery, University of Parma and Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Antonio Frassoldati
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Oncology, Azienda Ospedaliero Universitaria di Ferrara-Arcispedale Sant'Anna, Ferrara, Italy
| | - Ida Colantonio
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, S. Croce e Carle General Hospital, Cuneo, Italy
| | - Andrea Bonetti
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Oncology, "Mater Salutis" Hospital, Legnago, Italy
| | - Carlo Genova
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- AcademicOncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), Università degli Studi di Genova, Genova, Italy
| | - Donatella Giardina
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology, Ospedale Ramazzini di Carpi and Ospedale di Mirandola, Azienda Usl Modena, Carpi, Italy
| | - Federica Bertolini
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Saverio Cinieri
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, Hospital of Brindisi, Brindisi, Italy
| | - Giulia Pasello
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Matteo Brighenti
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Department, ASST Cremona, Cremona, Italy
| | - Elisa Andrini
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Michele Tognetto
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| | - Luca Boni
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Clinical Epidemiology Unit, IRCSS Ospedale Policlinico San Martino, Genova, Italy
| | - Andrea Ardizzoni
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| |
Collapse
|
30
|
Sun L, Gao X, Zhao Z, Zhu Y. Molecular subtypes and quantitative analysis of PD-L1 and tumor-associated immune cells in uterine carcinosarcoma. J Gynecol Oncol 2025; 36:36.e114. [PMID: 40405429 DOI: 10.3802/jgo.2025.36.e114] [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: 09/27/2024] [Revised: 01/24/2025] [Accepted: 04/03/2025] [Indexed: 05/24/2025] Open
Abstract
OBJECTIVE In the present study, molecular subtypes were determined, programmed death-ligand 1 (PD-L1) and tumor-associated immune cells (TAICs) were quantitatively detected, and their effect on prognosis in uterine carcinosarcoma (UCS) was analyzed. METHODS The study included 65 UCS cases. Direct sequencing of POLE exonuclease domain and immunohistochemistry of mismatch repair (MMR) deficiency proteins and p53 were used to stratify molecular subtypes. QuPath was used for quantitative immunohistochemical detection of PD-L1 and TAICs. The chi square test was used to determine the association between molecular subtypes and expression of PD-L1 and TAICs. The Kaplan-Meier method and Cox proportional hazards regression were used for plotting and survival analysis. RESULTS In 65 UCS cases, 1 case (1.5%) was POLE ultramutated (POLEmut) subtype, 11 cases (16.9%) were deficient MMR (dMMR) subtype, 32 cases (49.3%) were p53 mutant (p53mut) subtype, and 21 cases (32.3%) were nonspecific molecular profile (NSMP) subtype. The positive density of PD-L1 in tumor (p=0.022), CD8 in stroma (p=0.036), and CD163 in stroma (p=0.025) were significantly associated with molecular subtypes. The patients with POLEmut and dMMR subtypes had a relatively better prognosis trend than patients with NSMP and p53mut subtypes. The patients with high positive density of PD-L1 in tumor had significantly better prognosis; however, high positive density of CD163 in stroma showed significantly worse prognosis. CONCLUSION UCS could be classified into four molecular subtypes associated with prognosis. PD-L1 and M2 macrophages could effectively predict the prognosis of patients with UCS.
Collapse
Affiliation(s)
- Lili Sun
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Xiaozhuo Gao
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Zehua Zhao
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Yanmei Zhu
- Department of Pathology, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China.
| |
Collapse
|
31
|
Skelly DA, Graham JP, Cheng M, Furuta M, Walter A, Stoklasek TA, Yang H, Stearns TM, Poirion O, Zhang JG, Grassmann JDS, Luo D, Flynn WF, Courtois ET, Chang CH, Serreze DV, Menghi F, Reinholdt LG, Liu ET. Mapping the genetic landscape establishing a tumor immune microenvironment favorable for anti-PD-1 response. Cell Rep 2025; 44:115698. [PMID: 40343794 DOI: 10.1016/j.celrep.2025.115698] [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: 09/10/2024] [Revised: 01/03/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying host genetic factors modulating immune checkpoint inhibitor (ICI) efficacy is experimentally challenging. Our approach, utilizing the Collaborative Cross mouse genetic resource, fixes the tumor genomic configuration while varying host genetics. We find that response to anti-PD-1 (aPD1) immunotherapy is significantly heritable in four distinct murine tumor models (H2: 0.18-0.40). For the MC38 colorectal carcinoma system, we map four significant ICI response quantitative trait loci (QTLs) with significant epistatic interactions. The differentially expressed genes within these QTLs that define responder genetics are highly enriched for processes involving antigen processing and presentation, allograft rejection, and graft vs. host disease (all p < 1 × 10-10). Functional blockade of two top candidate immune targets, GM-CSF and IL-2RB, completely abrogates the MC38 transcriptional response to aPD1 therapy. Thus, our in vivo experimental platform is a powerful approach for discovery of host genetic factors that establish the tumor immune microenvironment propitious for ICI response.
Collapse
Affiliation(s)
- Daniel A Skelly
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - John P Graham
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | - Mayuko Furuta
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Andrew Walter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | | | - Timothy M Stearns
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Olivier Poirion
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ji-Gang Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Jessica D S Grassmann
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Diane Luo
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - William F Flynn
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Elise T Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; OB/Gyn Department, UConn Health, Farmington, CT 06032, USA
| | - Chih-Hao Chang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - David V Serreze
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Laura G Reinholdt
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.
| |
Collapse
|
32
|
Pepe D, Janssens X, Timcheva K, Marrón-Liñares GM, Verbelen B, Konstantakos V, De Groote D, De Bie J, Verhasselt A, Dewaele B, Godderis A, Cools C, Franco-Tolsau M, Royaert J, Verbeeck J, Kampen KR, Subramanian K, Cabrerizo Granados D, Menschaert G, De Keersmaecker K. Reannotation of cancer mutations based on expressed RNA transcripts reveals functional non-coding mutations in melanoma. Am J Hum Genet 2025:S0002-9297(25)00146-6. [PMID: 40359938 DOI: 10.1016/j.ajhg.2025.04.005] [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: 01/02/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
The role of synonymous mutations in cancer pathogenesis is currently underexplored. We developed a method to detect significant clusters of synonymous and missense mutations in public cancer genomics data. In melanoma, we show that 22% (11/50) of these mutation clusters are misannotated as coding mutations because the reference transcripts used for their annotation are not expressed. Instead, these mutations are actually non-coding. This, for instance, applies to the mutation clusters targeting known cancer genes kinetochore localized astrin (SPAG5) binding protein (KNSTRN) and BCL2-like 12 (BCL2L12), each affecting 4%-5% of melanoma tumors. For the latter, we show that these mutations are functional non-coding mutations that target the shared promoter region of interferon regulatory factor 3 (IRF3) and BCL2L12. This results in downregulation of IRF3, BCL2L12, and tumor protein p53 (TP53) expression in a CRISPR-Cas9 primary melanocyte model and in melanoma tumors. In individuals with melanoma, these mutations were also associated with a worse response to immunotherapy. Finally, we propose a simple automated method to more accurately annotate cancer mutations based on expressed transcripts. This work shows the importance of integrating DNA- and RNA-sequencing data to properly annotate mutations and identifies a number of previously overlooked and wrongly annotated functional non-coding mutations in melanoma.
Collapse
Affiliation(s)
- Daniele Pepe
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Xander Janssens
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Kalina Timcheva
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Grecia M Marrón-Liñares
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Benno Verbelen
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Vasileios Konstantakos
- Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for AI & Computational Biology (VIB.AI), Leuven, Belgium; VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Dylan De Groote
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jolien De Bie
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | | | - Barbara Dewaele
- Department of Human Genetics, KU Leuven, Leuven, Belgium; Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | | | - Charlotte Cools
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Mireia Franco-Tolsau
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jonathan Royaert
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jelle Verbeeck
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Kim R Kampen
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium; Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Karthik Subramanian
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - David Cabrerizo Granados
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Gerben Menschaert
- OHMX.bio NV, Evergem, Belgium; Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| | - Kim De Keersmaecker
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
33
|
Li Y, Song C, Wang H, Di W, Chen Y, Hu Y, Li P, Chen J, Ren Y, Gong J, Wang Q. Novel prognostic biomarkers in small cell lung cancer reveal mutational signatures, genomic mutations, and immune implications. Sci Rep 2025; 15:15592. [PMID: 40320401 PMCID: PMC12050310 DOI: 10.1038/s41598-025-00222-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
Small cell lung cancer (SCLC) is a highly malignant lung cancer subtype with a dismal prognosis and limited treatment options. This study aimed to identify new prognostic molecular biomarkers for SCLC and explore their immune-related implications for treatment strategies. We analyzed 200 SCLC samples via whole-exome sequencing (WES) and 313 samples by targeted sequencing. A smoking-related SBS4 mutational signature was linked to poorer prognosis and lower tumor mutational burden (TMB), while the APOBEC-mediated SBS13 signature was associated with better prognosis and higher TMB. We identified a molecular subtype with the worst outcomes and lowest TMB in both cohorts. Among 38 high-frequency mutated genes associated with SCLC prognosis, only UNC13A mutations were beneficial. Patients with UNC13A mutations had favorable immune infiltration and tumor immunogenicity. Additionally, TP53 splice site mutations were related to the worst survival outcomes. In conclusion, we discovered new molecular biomarkers for SCLC prognosis. Our findings on their immunological characteristics offer insights for developing novel SCLC treatment strategies.
Collapse
Affiliation(s)
- Yuting Li
- Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Chen Song
- Department of Hematology Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Haijun Wang
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Wenyu Di
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Yangyang Chen
- Department of Radiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Yuanyuan Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Peiheng Li
- Department of Radiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Jie Chen
- Department of Radiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China
| | - Yanfeng Ren
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Shandong Second Medical University, Baotong Xi Street, Weicheng District, Weifang, 261053, Shandong, China.
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Qinghua Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, China.
- Department of Health Statistics, Key Laboratory of Medicine and Health of Shandong Province, School of Public Health, Shandong Second Medical University, Baotong Xi Street, Weicheng District, Weifang, 261053, Shandong, China.
| |
Collapse
|
34
|
Rosenberger L, Hansmann L, Anastasopoulou V, Wolf SP, Drousch K, Moewes C, Feng X, Cao G, Huang J, Yew PY, Strønen E, Kato T, Saligrama N, Olweus J, Nakamura Y, Willimsky G, Blankenstein T, Schreiber H, Leisegang M. Selection of therapeutically effective T-cell receptors from the diverse tumor-bearing repertoire. J Immunother Cancer 2025; 13:e011351. [PMID: 40316304 PMCID: PMC12049912 DOI: 10.1136/jitc-2024-011351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/03/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND The development of T-cell receptor (TCR)-based T-cell therapies is hampered by the difficulties in identifying therapeutically effective tumor-specific TCRs from the natural repertoire of a patient's cancer-specific T cells. METHODS Here, we mimic experimentally near-patient conditions to analyze the T-cell repertoire in euthymic tumor-bearing mice responding to the H-2Kb-presented neoantigen p68S551F (mp68). We temporarily separated the time point of mp68 expression from that of cancer cell transplantation to exclude the influence of injection-induced inflammation on T-cell priming. Thus, the mp68-specific T-cell response could only develop after the acute inflammatory phase had subsided. RESULTS We found that mp68-specific TCRs isolated from either tumor-infiltrating T cells or spleens of mice immunized with mp68-expressing cancer cells are diverse and not inherently therapeutic when introduced into peripheral T cells and used for adoptive therapy of established tumors. While measuring short-term T-cell responses in vitro was unreliable for some TCRs in predicting their therapeutic failure, assessing the persistence of cancer cell destruction by TCR-modified T cells in long-term cultures accurately predicted therapeutic outcomes. A tumor-derived TCR with optimal function was also correctly identified with this approach when analyzing human TCRs that recognize the HLA-A2-presented neoantigen CDK4R24L. CONCLUSIONS We show that a neoantigen-directed T-cell response in tumor-bearing hosts comprises a diverse repertoire. Infiltration and expansion of certain T-cell clonotypes in the tumor do not necessarily correlate with therapeutic efficacy of their TCRs in adoptive therapy. We propose that analysis of persistent rather than immediate responses of TCR-modified T cells in vitro serves as a reliable parameter to identify TCRs that are therapeutically effective in vivo.
Collapse
Affiliation(s)
- Leonie Rosenberger
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Leo Hansmann
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vasiliki Anastasopoulou
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, Illinois, USA
| | - Kimberley Drousch
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christina Moewes
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Xinyi Feng
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Guoshuai Cao
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Jun Huang
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Poh Yin Yew
- Cancer Precision Medicine, Inc, Tokyo, Japan
| | - Erlend Strønen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Taigo Kato
- Department of Medicine, Center for Personalized Therapeutics, The University of Chicago, Chicago, Illinois, USA
| | - Naresha Saligrama
- Department of Neurology, Bursky Center for Human Immunology, and Immunotherapy Programs, Hope Center for Neurological Disorders, Center for Brain Immunology and Glia, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Yusuke Nakamura
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Gerald Willimsky
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology and Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
35
|
González‐Gil C, Lopes T, Morgades M, Fuster‐Tormo F, Montesinos P, Medina CR, Hermosín L, González‐Martínez T, Queipo M, González‐Campos J, Martínez‐Sánchez P, Díaz‐Beya M, Coll R, Maluquer C, Zamora L, Artola T, Vall‐Llovera F, Tormo M, Torrent A, Martínez‐Laperche C, Gil‐Cortés C, Barba P, Cervera M, Ribera J, Fernández‐Delgado M, Ayala R, Cladera A, Mateos MC, Vidal MJ, Feliu J, Torres A, Azaceta G, Calasanz MJ, Bigas A, Esteller M, Orfao A, Ribera JM, Genescà E. Genetic evolution and relapse-associated mutations in adult T-cell acute lymphoblastic leukemia patients treated in PETHEMA trials. Hemasphere 2025; 9:e70148. [PMID: 40420999 PMCID: PMC12104817 DOI: 10.1002/hem3.70148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 05/28/2025] Open
Abstract
Relapse is the main cause of treatment failure in T-cell acute lymphoblastic leukemia (T-ALL). Despite this, data from adult T-ALL patients treated with specific chemotherapeutic regimens that examine predictive markers and describe relapse mechanisms are scarce. In this study, we studied 74 paired diagnosis-relapse samples from 37 patients homogeneously treated with three consecutive measurable residual disease-oriented trials to identify genetic determinants involved in relapse in adult T-ALL. Analysis of single-nucleotide variants and copy number alterations consistently found N/KRAS mutations (20% relapsed cases) at diagnosis and at relapse (resistance profile). N/KRAS mut patients frequently relapse early during consolidation treatment. Relapse-specific mutations in NT5C2, NR3C1, SMARCA4, and TP53 (40% relapse cases) were not detected at diagnosis by conventional molecular techniques (relapse profile). However, single-cell-based analysis revealed a very minor clone containing the NT5C2(p.R367Q) variant at diagnosis. Patients with the NT5C2(p.R367Q) variant mostly relapse later during maintenance treatment. Tracking the NT5C2 variant by digital PCR confirm the expansion of the NT5C2 clone at maintenance treatment. Overall, our exploratory analysis suggests a role for these genetic events, most of which have already been described in pediatric cases, driving resistance associated to specific chemotherapeutic agents, contributing to the relapse of a high proportion of adult T-ALL patients (60%).
Collapse
Affiliation(s)
- Celia González‐Gil
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| | - Thaysa Lopes
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| | - Mireia Morgades
- Servei Hematologia Clínica, ICO‐Hospital Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| | - Francisco Fuster‐Tormo
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
- Present address:
Technology & Development TeamVeritas IntercontinentalBarcelonaSpain
| | | | | | - Lourdes Hermosín
- Servicio Hematología ClínicaHospital de JerezJerez de la FronteraSpain
| | | | - María‐Paz Queipo
- Servicio Hematología ClínicaHospital Virgen de la VictoriaMálagaSpain
| | | | | | - Marina Díaz‐Beya
- Servicio Hematología ClínicaHospital Clínic de BarcelonaBarcelonaSpain
| | - Rosa Coll
- Servei Hematologia ClínicaICO‐Hospital Josep TruetaGironaSpain
| | - Clara Maluquer
- Servei Hematologia ClínicaICO‐Hospital Duran i Reynals, Hospitalet del LlobregatBarcelonaSpain
| | - Lurdes Zamora
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
- Servei Hematologia Clínica, ICO‐Hospital Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| | - Teresa Artola
- Servicio Hematología ClínicaHospital Universitario de DonostiaDonostiaSpain
| | | | - Mar Tormo
- Hospital Clínico Universitario de Valencia, Instituto de Investigación INCLIVAValenciaSpain
| | - Anna Torrent
- Servei Hematologia Clínica, ICO‐Hospital Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| | | | | | - Pere Barba
- Servei Hematologia ClínicaHospital Universitari de la Vall d'HebronBarcelonaSpain
| | - Marta Cervera
- Servei d'Hematologia ClínicaICO‐Hospital Joan XXIIITarragonaSpain
| | - Jordi Ribera
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| | - Manuel Fernández‐Delgado
- Servicio Hematología ClínicaHospital General Universitario de CastellónCastellón de la PlanaSpain
| | - Rosa Ayala
- Servicio Hematología ClínicaHospital 12 de OctubreMadridSpain
| | - Antonia Cladera
- Servei Hematologia ClínicaHospital Son LLátzerPalma de MallorcaSpain
| | | | | | - Jesús Feliu
- Servicio Hematología ClínicaComplejo Hospitalario de San Millán y San PedroLogroñoSpain
| | - Ana Torres
- Servicio Hematología ClínicaHospital General de SegoviaSegoviaSpain
| | - Gemma Azaceta
- Servei Hematología ClínicaHospital Clínico Universitario Lozano BlesaZaragozaSpain
| | | | - Anna Bigas
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
- Programa d'Investigació del CancerInstitut Hospital del Mar d'Investigacions MèdiquesBarcelonaSpain
- CIBERONCBarcelonaSpain
| | - Manel Esteller
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
- CIBERONCBarcelonaSpain
| | - Alberto Orfao
- CIBERONCBarcelonaSpain
- Departamento de Medicina, Centro de Investigación del Cáncer (IBMCC‐CSIC/USAL), Instituto Biosanitario de Salamanca, CIBERONCUniversidad de SalamancaSalamancaSpain
| | - Josep Maria Ribera
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
- Servei Hematologia Clínica, ICO‐Hospital Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| | - Eulalia Genescà
- Institut d'Investigació contra la Leucemia Josep Carreras (IJC), Campus ICO‐Germans Trias i PujolUniversitat Autònoma de BarcelonaBadalonaSpain
| |
Collapse
|
36
|
Liu Y, Wang Q, Li Q, Ren P. Role of ELP6 in tumour progression and impact on ERK1/2 signalling pathway inhibitors in skin cutaneous melanoma. Oncol Lett 2025; 29:250. [PMID: 40177137 PMCID: PMC11962575 DOI: 10.3892/ol.2025.14996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Elongator acetyltransferase complex subunit 6 (ELP6), a subunit of the elongator complex, can increase the migratory potential of melanoma cells in vitro. However, the clinical relevance of ELP6 in patients with melanoma remains unclear. The present study aimed to investigate the role of ELP6 expression in melanoma progression and association with patient survival rates. Transcriptomic data from patients with melanoma available in The Cancer Genome Atlas, Gene Expression Profiling Interactive Analysis and cBioPortal databases were analysed to evaluate the associations between ELP6 expression levels and patient survival. In vitro experiments were conducted using short hairpin RNAs to downregulate ELP6, with a focus on cell viability, cell cycle regulation and the ERK1/2 signalling pathway. ELP6 expression levels were significantly elevated in patients with melanoma and were associated with poor survival outcomes. Knockdown of ELP6 resulted in decreased expression levels of p42 MAPK, reduced cell viability, G1 phase cell cycle arrest and led to reduced responsiveness to the MEK1/2 inhibitor U0126. ELP6 promotes melanoma progression via the ERK1/2 signalling pathway. Therefore, assessing ELP6 expression may offer potential therapeutic strategies for patients with melanoma.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Qinrong Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Qian Li
- Department of Pharmacy, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Peng Ren
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, P.R. China
| |
Collapse
|
37
|
Xiao Y, Jin W, Ju L, Fu J, Wang G, Yu M, Chen F, Qian K, Wang X, Zhang Y. Tracking single-cell evolution using clock-like chromatin accessibility loci. Nat Biotechnol 2025; 43:784-798. [PMID: 38724668 DOI: 10.1038/s41587-024-02241-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 04/10/2024] [Indexed: 05/18/2025]
Abstract
Single-cell chromatin accessibility sequencing (scATAC-seq) reconstructs developmental trajectory by phenotypic similarity. However, inferring the exact developmental trajectory is challenging. Previous studies showed age-associated DNA methylation (DNAm) changes in specific genomic regions, termed clock-like differential methylation loci (ClockDML). Age-associated DNAm could either result from or result in chromatin accessibility changes at ClockDML. As cells undergo mitosis, the heterogeneity of chromatin accessibility on clock-like loci is reduced, providing a measure of mitotic age. In this study, we developed a method, called EpiTrace, that counts the fraction of opened clock-like loci from scATAC-seq data to determine cell age and perform lineage tracing in various cell lineages and animal species. It shows concordance with known developmental hierarchies, correlates well with DNAm-based clocks and is complementary with mutation-based lineage tracing, RNA velocity and stemness predictions. Applying EpiTrace to scATAC-seq data reveals biological insights with clinically relevant implications, ranging from hematopoiesis, organ development, tumor biology and immunity to cortical gyrification.
Collapse
Affiliation(s)
- Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wan Jin
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
- Euler Technology, ZGC Life Sciences Park, Beijing, China
| | - Lingao Ju
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jie Fu
- Hong Kong University of Science and Technology, Hong Kong, China
| | - Gang Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mengxue Yu
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fangjin Chen
- High Performance Computing Center, Peking-Tsinghua College of Life Sciences, Peking University, Beijing, China
| | - Kaiyu Qian
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Yi Zhang
- Euler Technology, ZGC Life Sciences Park, Beijing, China.
| |
Collapse
|
38
|
Ma Z, Li M, Li F, Wu K, Wu X, Luo T, Gao N, Luo H, Sui Z, Yu Z, Jiang H, Shang X, Chen C, Yue J, Meng F, Duan X, Xu B. Multi-omics sequencing of gastroesophageal junction adenocarcinoma reveals prognosis-relevant key factors and a novel immunogenomic classification. Gastric Cancer 2025; 28:344-357. [PMID: 39883307 DOI: 10.1007/s10120-025-01585-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Gastroesophageal junction adenocarcinoma (GEJAC) exhibits distinct molecular characteristics due to its unique anatomical location. We sought to investigate effective and reliable molecular classification of GEJAC to guide personalized treatment. METHODS We analyzed the whole genomic, transcriptomic, T-cell receptor repertoires, and immunohistochemical data in 92 GEJAC patients and delineated the landscape of genetic and immune alterations. In addition to COSMIC nomenclature, the de novo nomenclature was also utilized to define signatures and investigate their correlation with survival. A novel molecular subtype was developed and validated in other cohorts. RESULTS We found 30 mutated driver genes, 7 novel genomic signatures, 3 copy-number variations, and 2 V-J gene usages related to prognosis that were not identified in previous study. A high frequency of COSMIC-SBS-384-1 and De novo-SV-32-A was associated with more neoantigen generation and a better survival. Using 19 molecular features, we identified three immune-related subtypes (immune inflamed, intermediate, and deserted) with discrete profiles of genomic signatures, immune status, and clinical outcome. The immune deserted subtype (27.2%) was characterized by an earlier KRAS mutation, worse immune reaction, and prognosis than the other two subtypes. The immune inflamed subtypes exhibited the highest levels of neoantigens, TCR/pMHC-binding strength, CD8 + T-cell infiltration, IFN-α/γ response pathways, and survival rate. CONCLUSIONS These results emphasize the immune reaction and prognostic value of novel molecular classifications based on multi-omics data and provide a solid basis for better management of GEJAC.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Biochemistry and Molecular Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Mengting Li
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310000, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fuqiang Li
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310000, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China
- BGI Genomics, Shenzhen, 518083, China
| | - Kui Wu
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310000, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China
- BGI Genomics, Shenzhen, 518083, China
| | - Xianxian Wu
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Tian Luo
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310000, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China
- BGI Genomics, Shenzhen, 518083, China
| | - Na Gao
- Department of Pathology, Key Laboratory of Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Huijuan Luo
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310000, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China
- BGI Genomics, Shenzhen, 518083, China
| | - Zhilin Sui
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Zhentao Yu
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Hongjing Jiang
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Xiaobin Shang
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Chuangui Chen
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Jie Yue
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Fianbiao Meng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xiaofeng Duan
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, 300060, China
| | - Bo Xu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and Chongqing University School of Medicine, 181 Hanyu Rd., Shapin District, Chongqing, 400030, China.
| |
Collapse
|
39
|
Klümper N, Cox A, Sjödahl G, Roghmann F, Bolenz C, Hartmann A, Grünwald V, Faltas BM, Hölzel M, Eckstein M. Pre-treatment metastatic biopsy: a step towards precision oncology for urothelial cancer. Nat Rev Urol 2025; 22:256-267. [PMID: 39472646 DOI: 10.1038/s41585-024-00951-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 05/10/2025]
Abstract
Early metastatic spread and clonal expansion of individual mutations result in a heterogeneous tumour landscape in metastatic urothelial cancer (mUC). Substantial molecular heterogeneity of common drug targets, such as membranous NECTIN4, FGFR3 mutations, PDL1 or immune phenotypes, has been documented between primary and metastatic tumours. However, translational and clinical studies frequently do not account for such heterogeneity and often investigate primary tumour samples that might not be representative in patients with mUC. We propose this as a potential factor for why many biomarkers for mUC have failed to be integrated into clinical practice. Fresh pre-treatment metastatic biopsies enable the capturing of prevailing tumour biology in real time. The characterization of metastatic tumour samples can improve response prediction to immunotherapy, the anti-NECTIN4 antibody-drug conjugate enfortumab vedotin and the FGFR inhibitor erdafitinib. Routine metastatic biopsy can thus improve the precision of identifying driver druggable alterations, thus improving treatment selection for patients with mUC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany.
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany.
| | - Alexander Cox
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
| | - Gottfrid Sjödahl
- Department of Translational Medicine, Division of Urological Research, Lund University, Lund, Sweden
| | - Florian Roghmann
- Department of Urology, Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Christian Bolenz
- Department of Urology and Paediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumour Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Bishoy M Faltas
- Department of Hematology/Oncology, Weill-Cornell Medicine, New York, NY, USA
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
40
|
Han L, Liu J, Zhang R, Cheng Y, Dong L, Wei L, Liu J, Wang K, Yu J. Insights From Nonsense-Mediated mRNA Decay for Prognosis in Homologous Recombination-Deficient Ovarian Cancer. Cancer Sci 2025; 116:1449-1463. [PMID: 40022542 PMCID: PMC12044663 DOI: 10.1111/cas.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Not all ovarian cancer patients with homologous recombination deficiency, especially those with germline BRCA mutations, can benefit from platinum-based and targeted therapy. Our study aimed to determine the value of nonsense-mediated mRNA decay, which targeted these mutations. The retrospective analysis of 797 ovarian cancer patients was performed using two public cohorts and one in-house cohort. We developed a prediction algorithm for nonsense-mediated mRNA decay to discriminate between trigger and escape status, finding that escape status indicated a better prognosis. Subsequently, we analyzed differential gene expression and functional pathways between the two statuses and filtered 8 genes associated with the cell cycle. Then the optimized key gene model was built using integrated machine learning algorithms (mean AUC > 0.89), which had a higher independent prognostic value for ovarian cancer with germline BRCA variants or homologous recombination deficiency than the nonsense-mediated mRNA decay algorithm. Furthermore, we classified patients into high- and low-risk groups by the machine learning model and found that the low-risk group had a better prognosis with higher drug response and immune levels of activated dendritic cells than the high-risk controls. Our findings provide a perspective based on nonsense-mediated mRNA decay and cell cycle pathways to distinguish subtypes of germline BRCA or homologous recombination deficiency.
Collapse
Affiliation(s)
- Lei Han
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Jialing Liu
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Runjiao Zhang
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Yanan Cheng
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Li Dong
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Lijuan Wei
- Cancer Prevention CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Juntian Liu
- Cancer Prevention CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Ke Wang
- Department of Gynecologic OncologyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Jinpu Yu
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| |
Collapse
|
41
|
Jian J, Yuan C, Hao H. Identifying key genes and functionally enriched pathways in acute myeloid leukemia by weighted gene co-expression network analysis. J Appl Genet 2025; 66:347-362. [PMID: 38977582 DOI: 10.1007/s13353-024-00881-0] [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: 02/23/2023] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 07/10/2024]
Abstract
Acute myeloid leukemia (AML) is characterized by the uncontrolled proliferation of myeloid leukemia cells in the bone marrow and other hematopoietic tissues and is highly heterogeneous. While with the progress of sequencing technology, understanding of the AML-related biomarkers is still incomplete. The purpose of this study is to identify potential biomarkers for prognosis of AML. Based on WGCNA analysis of gene mutation expression, methylation level distribution, mRNA expression, and AML-related genes in public databases were employed for investigating potential biomarkers for the prognosis of AML. This study screened a total of 6153 genes by analyzing various changes in 103 acute myeloid leukemia (AML) samples, including gene mutation expression, methylation level distribution, mRNA expression, and AML-related genes in public databases. Moreover, seven AML-related co-expression modules were mined by WGCNA analysis, and twelve biomarkers associated with the AML prognosis were identified from each top 10 genes of the seven co-expression modules. The AML samples were then classified into two subgroups, the prognosis of which is significantly different, based on the expression of these twelve genes. The differentially expressed 7 genes of two subgroups (HOXB-AS3, HOXB3, SLC9C2, CPNE8, MEG8, S1PR5, MIR196B) are mainly involved in glucose metabolism, glutathione biosynthesis, small G protein-mediated signal transduction, and the Rap1 signaling pathway. With the utilization of WGCNA mining, seven gene co-expression modules were identified from the TCGA database, and there are unreported genes that may be potential driver genes of AML and may be the direction to identify the possible molecular signatures to predict survival of AML patients and help guide experiments for potential clinical drug targets.
Collapse
Affiliation(s)
- Jimo Jian
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
- Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chenglu Yuan
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China
| | - Hongyuan Hao
- Qilu Hospital of Shandong University, Qingdao, 266035, Shandong, China.
| |
Collapse
|
42
|
Koh GCC, Nanda AS, Rinaldi G, Boushaki S, Degasperi A, Badja C, Pregnall AM, Zhao SJ, Chmelova L, Black D, Heskin L, Dias J, Young J, Memari Y, Shooter S, Czarnecki J, Brown MA, Davies HR, Zou X, Nik-Zainal S. A redefined InDel taxonomy provides insights into mutational signatures. Nat Genet 2025; 57:1132-1141. [PMID: 40210680 DOI: 10.1038/s41588-025-02152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Despite their deleterious effects, small insertions and deletions (InDels) have received far less attention than substitutions. Here we generated isogenic CRISPR-edited human cellular models of postreplicative repair dysfunction (PRRd), including individual and combined gene edits of DNA mismatch repair (MMR) and replicative polymerases (Pol ε and Pol δ). Unique, diverse InDel mutational footprints were revealed. However, the prevailing InDel classification framework was unable to discriminate these InDel signatures from background mutagenesis and from each other. To address this, we developed an alternative InDel classification system that considers flanking sequences and informative motifs (for example, longer homopolymers), enabling unambiguous InDel classification into 89 subtypes. Through focused characterization of seven tumor types from the 100,000 Genomes Project, we uncovered 37 InDel signatures; 27 were new. In addition to unveiling previously hidden biological insights, we also developed PRRDetect-a highly specific classifier of PRRd status in tumors, with potential implications for immunotherapies.
Collapse
Affiliation(s)
- Gene Ching Chiek Koh
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Arjun Scott Nanda
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Giuseppe Rinaldi
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Soraya Boushaki
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Andrea Degasperi
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Cherif Badja
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Andrew Marcel Pregnall
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Salome Jingchen Zhao
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Lucia Chmelova
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Daniella Black
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Laura Heskin
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - João Dias
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jamie Young
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Yasin Memari
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Scott Shooter
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jan Czarnecki
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Matthew Arthur Brown
- Genomics England, Queen Mary University of London, Dawson Hall, Charterhouse Square, London, UK
| | - Helen Ruth Davies
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Xueqing Zou
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Serena Nik-Zainal
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
43
|
Garcia NMG, Becerra JN, Srinivasan S, McKinney BJ, DiMarco AV, Wu F, Fitzgibbon M, Alvarez JV. APOBEC3 Activity Promotes the Survival and Evolution of Drug-Tolerant Persister Cells during EGFR Inhibitor Resistance in Lung Cancer. CANCER RESEARCH COMMUNICATIONS 2025; 5:825-840. [PMID: 40323013 DOI: 10.1158/2767-9764.crc-24-0442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/11/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
APOBEC mutagenesis is one of the most common endogenous sources of mutations in human cancer and is a major source of genetic intratumor heterogeneity. High levels of APOBEC mutagenesis are associated with poor prognosis and aggressive disease across diverse cancers, but the mechanistic and functional impacts of APOBEC mutagenesis on tumor evolution and therapy resistance remain relatively unexplored. To address this, we investigated the contribution of APOBEC mutagenesis to acquired therapy resistance in a model of EGFR-mutant non-small cell lung cancer. We find that inhibition of EGFR in lung cancer cells leads to a rapid and pronounced induction of APOBEC3 expression and activity. Functionally, APOBEC expression promotes the survival of drug-tolerant persister cells (DTP) following EGFR inhibition. Constitutive expression of APOBEC3B alters the evolutionary trajectory of acquired resistance to the EGFR inhibitor gefitinib, making it more likely that resistance arises through de novo acquisition of the T790M gatekeeper mutation and squamous transdifferentiation during the DTP state. APOBEC3B expression is associated with increased expression of the squamous cell transcription factor ΔNp63 and squamous cell transdifferentiation in gefitinib-resistant cells. Knockout of p63 in gefitinib-resistant cells reduces the expression of the ΔNp63 target genes IL-1α/β and sensitizes these cells to the third-generation EGFR inhibitor osimertinib. These results suggest that APOBEC activity promotes acquired resistance by facilitating evolution and transdifferentiation in DTPs and that approaches to target ΔNp63 in gefitinib-resistant lung cancers may have therapeutic benefit. SIGNIFICANCE APOBEC mutagenesis is a common source of genetic heterogeneity in cancer, and APOBEC mutational signatures are enriched in metastatic and drug-resistant tumors. However, the mechanisms through which APOBEC3 enzymes promote tumor evolution remain unknown. In this study, we show that APOBEC3 activity contributes to the development of therapy-resistant cancer cells by promoting evolution of DTP cells. These findings offer insights into the role of APOBEC mutagenesis in cancer progression.
Collapse
Affiliation(s)
- Nina Marie G Garcia
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Jessica N Becerra
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Sharan Srinivasan
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Brock J McKinney
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Ashley V DiMarco
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Feinan Wu
- Genomics and Bioinformatics, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Matthew Fitzgibbon
- Genomics and Bioinformatics, Fred Hutchinson Cancer Center, Seattle, Washington
| | - James V Alvarez
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|
44
|
Huang YE, Zhou S, Chen S, Chen J, Zhou X, Hou F, Liu H, Yuan M, Jiang W. Mutational signature-based biomarker to predict the response of immune checkpoint inhibitors therapy in cancers. Int J Biol Macromol 2025; 308:142585. [PMID: 40154701 DOI: 10.1016/j.ijbiomac.2025.142585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Patients have a limited response rate to immune checkpoint inhibitors (ICIs) therapy. Although several biomarkers have been proposed, their ability to accurately predict the response to ICIs therapy remains unsatisfactory. In addition, mutational signatures were validated to be associated with ICIs therapy. Therefore, we developed a mutational signature-based biomarker (MS-bio) to predict the response to ICIs therapy. Based on differentially mutated genes, we extracted six mutational signatures (single-base substitution (SBS)-A, SBS-B, SBS-C, SBS-D, double-base substitution (DBS)-A, and DBS-B) as MS-bio, and constructed a random forest (RF) model to predict the response. Internal and external validations consistently demonstrated the excellent predictive capability of MS-bio, with an accuracy reaching up to 0.82. Moreover, MS-bio exhibited superior performance compared to existing biomarkers. To further validate the accuracy of MS-bio, we explored its performance in The Cancer Genome Atlas (TCGA) cohort and found that the predicted responders were immunologically "hot". Finally, we found that SBS-C had the highest importance in prediction and was related to T cell differentiation. Overall, here we introduced MS-bio as a novel biomarker for accurately predicting the response to ICIs therapy, thereby contributing to the advancement of precision medicine.
Collapse
Affiliation(s)
- Yu-E Huang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Shunheng Zhou
- School of Computer Sciences, University of South China, Hengyang 421001, China
| | - Sina Chen
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Jiahao Chen
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Xu Zhou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Fei Hou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Haizhou Liu
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Wei Jiang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
45
|
Gunnarsson R, Yang M, Biloglav A, Lundin‐Ström KB, Lilljebjörn H, Castor A, Fioretos T, Olsson‐Arvidsson L, Paulsson K, Johansson B. Mutational signatures and kataegis in pediatric B-cell precursor acute lymphoblastic leukemia. Hemasphere 2025; 9:e70136. [PMID: 40395431 PMCID: PMC12088958 DOI: 10.1002/hem3.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 05/22/2025] Open
Affiliation(s)
- Rebeqa Gunnarsson
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
| | - Minjun Yang
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
| | - Andrea Biloglav
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
| | | | - Henrik Lilljebjörn
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
| | - Anders Castor
- Department of PediatricsSkåne University HospitalLundSweden
| | - Thoas Fioretos
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
- Department of Clinical Genetics, Pathology, and Molecular DiagnosticsSkåne University HospitalLundSweden
| | - Linda Olsson‐Arvidsson
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
- Department of Clinical Genetics, Pathology, and Molecular DiagnosticsSkåne University HospitalLundSweden
| | - Kajsa Paulsson
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
| | - Bertil Johansson
- Division of Clinical Genetics, Department of Laboratory MedicineLund UniversityLundSweden
- Department of Clinical Genetics, Pathology, and Molecular DiagnosticsSkåne University HospitalLundSweden
| |
Collapse
|
46
|
Cheng AP, Widman AJ, Arora A, Rusinek I, Sossin A, Rajagopalan S, Midler N, Hooper WF, Murray RM, Halmos D, Langanay T, Chu H, Inghirami G, Potenski C, Germer S, Marton M, Manaa D, Helland A, Furatero R, McClintock J, Winterkorn L, Steinsnyder Z, Wang Y, Alimohamed AI, Malbari MS, Saxena A, Callahan MK, Frederick DT, Spain L, Sigouros M, Manohar J, King A, Wilkes D, Otilano J, Elemento O, Mosquera JM, Jaimovich A, Lipson D, Turajlic S, Zody MC, Altorki NK, Wolchok JD, Postow MA, Robine N, Faltas BM, Boland G, Landau DA. Error-corrected flow-based sequencing at whole-genome scale and its application to circulating cell-free DNA profiling. Nat Methods 2025; 22:973-981. [PMID: 40217113 PMCID: PMC12077166 DOI: 10.1038/s41592-025-02648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/04/2025] [Indexed: 05/15/2025]
Abstract
Differentiating sequencing errors from true variants is a central genomics challenge, calling for error suppression strategies that balance costs and sensitivity. For example, circulating cell-free DNA (ccfDNA) sequencing for cancer monitoring is limited by sparsity of circulating tumor DNA, abundance of genomic material in samples and preanalytical error rates. Whole-genome sequencing (WGS) can overcome the low abundance of ccfDNA by integrating signals across the mutation landscape, but higher costs limit its wide adoption. Here, we applied deep (~120×) lower-cost WGS (Ultima Genomics) for tumor-informed circulating tumor DNA detection within the part-per-million range. We further leveraged lower-cost sequencing by developing duplex error-corrected WGS of ccfDNA, achieving 7.7 × 10-7 error rates, allowing us to assess disease burden in individuals with melanoma and urothelial cancer without matched tumor sequencing. This error-corrected WGS approach will have broad applicability across genomics, allowing for accurate calling of low-abundance variants at efficient cost and enabling deeper mapping of somatic mosaicism as an emerging central aspect of aging and disease.
Collapse
Affiliation(s)
- Alexandre Pellan Cheng
- New York Genome Center, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA.
- Département de Génie des Systèmes, École de Technologie Supérieure, Montréal, Québec, Canada.
- Axe Cancer, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.
| | - Adam J Widman
- New York Genome Center, New York, NY, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anushri Arora
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Aaron Sossin
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Srinivas Rajagopalan
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Nicholas Midler
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Rebecca M Murray
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Daniel Halmos
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Theophile Langanay
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Hoyin Chu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Giorgio Inghirami
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Catherine Potenski
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | | | - Dina Manaa
- New York Genome Center, New York, NY, USA
| | | | | | | | | | | | - Yohyoh Wang
- New York Genome Center, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Asrar I Alimohamed
- Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Murtaza S Malbari
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ashish Saxena
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Dennie T Frederick
- Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Lavinia Spain
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Michael Sigouros
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jyothi Manohar
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Abigail King
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Wilkes
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - John Otilano
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Juan Miguel Mosquera
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Samra Turajlic
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | | | - Nasser K Altorki
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jedd D Wolchok
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Ludwig Institute for Cancer Research, New York, NY, USA
| | - Michael A Postow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Bishoy M Faltas
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Genevieve Boland
- Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Dan A Landau
- New York Genome Center, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
47
|
Rana M, Liou KC, Thakur A, Nepali K, Liou JP. Advancing glioblastoma therapy: Learning from the past and innovations for the future. Cancer Lett 2025; 617:217601. [PMID: 40037502 DOI: 10.1016/j.canlet.2025.217601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Marred by a median survival of only around 12-15 months coupled with poor prognosis and effective therapeutic deprived drug armory, treatment/management of glioblastoma has proved to be a daunting task. Surgical resection, flanked by radiotherapy and chemotherapy with temozolomide, stands as the standard of care; however, this trimodal therapy often manifests limited efficacy due to the heterogeneous and highly infiltrative nature of GBM cells. In addition, the existence of the blood-brain barrier, tumor microenvironment, and the immunosuppressive nature of GBM, along with the encountered resistance of GBM cells towards conventional therapy, also hinders the therapeutic applications of chemotherapeutics in GBM. This review presents key insights into the molecular pathology of GBM, including genetic mutations, signaling pathways, and tumor microenvironment characteristics. Recent innovations such as immunotherapy, oncolytic viral therapies, vaccines, nanotechnology, electric field, and cancer neuroscience, as well as their clinical progress, have been covered. In addition, this compilation also encompasses a discussion on the role of personalized medicine in tailoring treatments based on individual tumor profiles, an approach that is gradually shifting the paradigm in GBM management. Endowed with the learnings imbibed from past failures coupled with the zeal to embrace novel/multidisciplinary approaches, researchers appear to be on the right track to pinpoint more effective and durable solutions in the context of GBM treatment.
Collapse
Affiliation(s)
- Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Ke-Chi Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
48
|
Liu N, Li X, Luo X, Liu B, Tang J, Xiao F, Wang W, Tang Y, Shu P, Zhang B, Chen Y, Qin D, Ma Q, Guo F, Tang X, Zhu D, Mei J, Chen W, Li D, Jiang L, Wang Y. Development and validation of machine learning models based on molecular features for estimating the probability of multiple primary lung carcinoma versus intrapulmonary metastasis in patients presenting multiple non-small cell lung cancers. Transl Lung Cancer Res 2025; 14:1118-1137. [PMID: 40386724 PMCID: PMC12082235 DOI: 10.21037/tlcr-24-875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/27/2025] [Indexed: 05/20/2025]
Abstract
Background Discrimination of multiple non-small cell lung cancers (NSCLCs) as multiple primary lung cancers (MPLCs) or intrapulmonary metastases (IPMs) is critical but remains challenging. The aim of this study is to develop and validate the machine learning (ML) models based on the molecular features for estimating the probability of MPLC or IPM for patients presenting multiple NSCLCs. Methods A total of 72 multiple NSCLCs patients with 157 surgical resection tumor lesions from January 2012 to January 2018 at two institutions were included for developing and testing models. Specifically, 46 patients with 103 tumors which were defined as definitive MPLC or IPM according to International Association for the Study of Lung Cancer (IASLC) criteria were used to develop models. They were spilt into training and validation sets using stratified random sampling and five-fold cross-validation. The developed models were tested in other 26 patients whose tumors were undetermined by traditional methods. Whole-exome sequencing (WES) was performed on all included tumor samples. Four molecular features were calculated to characterize tumors relatedness and served as model inputs, including genetic divergence, shared mutation number, Pearson correlation coefficient and early mutation number. Decision trees (DT), random forests (RF), and gradient boosting decision trees (GBDT) were employed, with performance assessed by areas under the curve (AUCs), accuracy, precision, recall, and F1 score in validation set. Disease-free survival (DFS) were used to evaluate model performance in test cohort. Clinical and genetic characteristics were then compared between MPLC and IPM populations. Results All of the four molecular features showed significant differences between MPLC and IPM patients in development cohort. That is, MPLC exhibited higher genetic divergence, lower shared mutation number, Pearson correlation and early mutation number than IPM (P<0.001). DT model, RF model and GBDT model were developed with these factors and achieved a mean AUC of 0.94 [standard deviation (SD) 0.09], 1.00 (SD 0.00) and 1.00 (SD 0.00) in validation set, respectively. DT model, RF model and GBDT model discriminated the undetermined multiple NSCLCs as MPLC (n=15) and IPM (n=11) consistently. MPLC identified by ML models had significantly prolonged DFS [hazard ratio =0.21; 95% confidence interval (CI): 0.04-1.0; P=0.04] than that of IPM. MPLC patients had a relative higher prevalence of family history of first-degree relatives with cancer, and more than half of these patients reported a family history of lung cancer. EGFR remains the most common mutated driver both in MPLC and IPM populations. Conclusions ML models based on the molecular features effectively distcriminate primary tumors from metastases in multiple NSCLCs, which improve the accuracy of multiple NSCLCs diagnosis and assist in clinical decision-making, particularly in challenging cases.
Collapse
Affiliation(s)
- Ning Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Luo
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, China
- School of Computer Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Bin Liu
- Department of Pulmonary Tumor Ward, Sichuan Cancer Hospital, Chengdu, China
| | - Jie Tang
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Xiao
- Genecast Biotechnology Co., Ltd., Wuxi, China
| | - Weiya Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Tang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Pei Shu
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Benxia Zhang
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Chen
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Diyuan Qin
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qizhi Ma
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fuchun Guo
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojun Tang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Daxing Zhu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiandong Mei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Weizhi Chen
- Genecast Biotechnology Co., Ltd., Wuxi, China
| | - Dan Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Shen C, Lin C, Qu F, Chen C, Shao Z, Jiang Y, Hu X, Di G. Genomic spectra of lymphovascular invasion in breast cancer. Chin J Cancer Res 2025; 37:138-153. [PMID: 40353081 PMCID: PMC12062984 DOI: 10.21147/j.issn.1000-9604.2025.02.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/25/2025] [Indexed: 05/14/2025] Open
Abstract
Objective Lymphovascular invasion (LVI) is a crucial step in metastasis and is closely associated with poor prognosis in patients with breast cancer. However, its clinical and molecular characteristics remain insufficiently defined. We aimed to identify molecular targets for LVI-positive (LVI+) breast cancer and predict patient prognosis via the analysis of genomic variations using targeted sequencing. Methods We established a large-scale targeted sequencing cohort of 4,079 breast cancer samples, which included 3,159 early-stage and locally advanced patients with available LVI statuses. Comparisons of somatic mutation frequencies and germline pathogenic/likely pathogenic (P/LP) mutation frequencies, mutational signature analyses, and mutual exclusivity and co-occurrence analyses were performed to identify key genomic features involved in LVI+ patients. Additionally, Kaplan-Meier survival analysis was conducted to further explore the prognostic value of co-mutations in LVI+ cases. Results We observed that LVI+ patients with the hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) and triple-negative breast cancer (TNBC) subtypes exhibited worse disease-free survival. Notably, HR+/HER2- and HER2+ breast cancer patients with LVI displayed distinct genomic features compared with LVI- tumors. Specifically, LVI+ HR+/HER2- tumors exhibited greater frequencies of somatic mutations in TP53 and ESR1, germline BRCA2 P/LP variations, and an enrichment of clock-like single-base substitution (SBS)1 mutational signatures. In contrast, LVI+ HER2+ tumors demonstrated a higher incidence of somatic PIK3CA mutations and increased activity of the apolipoprotein B mRNA editing enzyme catalytic polypeptide (APOBEC)-associated SBS2 signature. Furthermore, we revealed that the co-mutation of TP53 and NF1 could serve as a potential prognostic marker for LVI+ HR+/HER2- patients. Conclusions Our findings provide a comprehensive overview of the genomic characteristics of LVI in breast cancer, thereby offering insights that may help in refining precision treatment strategies for LVI+ breast cancer patients.
Collapse
Affiliation(s)
- Chuhan Shen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Caijin Lin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Feilin Qu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhiming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yizhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xin Hu
- Precision Cancer Medical Center, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Genhong Di
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
50
|
Tuffaha MZ, Castellano D, Colomé CS, Gutenkunst RN, Wahl LM. Nonhypermutator Cancers Access Driver Mutations Through Reversals in Germline Mutational Bias. Mol Biol Evol 2025; 42:msaf105. [PMID: 40415200 DOI: 10.1093/molbev/msaf105] [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: 01/07/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 05/27/2025] Open
Abstract
Cancer is an evolutionary disease driven by mutations in asexually reproducing somatic cells. In asexual microbes, bias reversals in the mutation spectrum can speed adaptation by increasing access to previously undersampled beneficial mutations. By analyzing tumors from 20 tissues, along with normal tissue and the germline, we demonstrate this effect in cancer. Nonhypermutated tumors reverse the germline mutation bias and have consistent spectra across tissues. These spectra changes carry the signature of hypoxia, and they facilitate positive selection in cancer genes. Hypermutated and nonhypermutated tumors thus acquire driver mutations differently: hypermutated tumors by higher mutation rates and nonhypermutated tumors by changing the mutation spectrum to reverse the germline mutation bias.
Collapse
Affiliation(s)
| | - David Castellano
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Claudia Serrano Colomé
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Ryan N Gutenkunst
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Lindi M Wahl
- Mathematics, Western University, London, ON, Canada N6A 5B7
| |
Collapse
|