1
|
Takamatsu R, Nakamura K, Chiyoda T, Tsuji K, Kawano R, Yoshimi N, Yamagami W, Nishihara H. Advancing Precision Oncology: Whole-Exome Sequencing in Endometrial Cancer Liquid-Based Cytology. Arch Pathol Lab Med 2025; 149:561-567. [PMID: 39396820 DOI: 10.5858/arpa.2024-0137-oa] [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: 08/28/2024] [Indexed: 10/15/2024]
Abstract
CONTEXT.— Diagnostic strategies for endometrial cancer have been evolving, with cytologic analysis being considered a key method in integrated oncologic diagnostics because of its less invasive nature and adaptability to various assessments. Liquid-based cytology (LBC) has emerged as a promising method for intact DNA preservation; it exhibits improved efficiency in advanced sequencing applications such as next-generation sequencing. However, despite the use of LBC in panel assays, its application in whole-exome sequencing (WES) for comprehensive genomic profiling remains underexplored. OBJECTIVE.— To investigate whether molecular classification is possible based on WES using DNA derived from LBC specimens. DESIGN.— We combined WES with targeted gene panel analysis to compare genomic findings of LBC and traditional tissue samples obtained from 7 cases of endometrial cancer. We investigated pathogenic mutations, tumor mutational burden, and microsatellite instability, and achieved molecular classification with high accuracy. RESULTS.— We found a substantial concordance between LBC and traditional tissue samples in terms of pathogenic mutation detection, with a 95% match in the LBC samples and 94% in the tissue samples. Notably, our results highlight the importance of combining WES with panel-based analysis in identifying the ultramutated status of a case that had been missed during panel analysis. CONCLUSIONS.— Our findings emphasize the potential of LBC samples in the precise and noninvasive genomic analysis of cases of endometrial cancer and offer a new avenue for developing diagnostic and therapeutic strategies in precision oncology.
Collapse
Affiliation(s)
- Reika Takamatsu
- From the Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (Takamatsu, Nakamura, Kawano, Yoshimi, Nishihara)
| | - Kohei Nakamura
- From the Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (Takamatsu, Nakamura, Kawano, Yoshimi, Nishihara)
- the Department of Obstetrics and Gynecology, Kumagaya General Hospital, Kumagaya, Saitama, Japan (Nakamura)
| | - Tatsuyuki Chiyoda
- the Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (Chiyoda, Tsuji, Yamagami)
| | - Kosuke Tsuji
- the Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (Chiyoda, Tsuji, Yamagami)
| | - Ryutaro Kawano
- From the Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (Takamatsu, Nakamura, Kawano, Yoshimi, Nishihara)
| | - Naoki Yoshimi
- the Division of Diagnostic Pathology, Okinawa Red Cross Hospital, Naha, Okinawa, Japan (Yoshimi)
| | - Wataru Yamagami
- the Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (Chiyoda, Tsuji, Yamagami)
| | - Hiroshi Nishihara
- From the Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan (Takamatsu, Nakamura, Kawano, Yoshimi, Nishihara)
| |
Collapse
|
2
|
Kjær A, Kristjánsdóttir N, Juul RI, Nordentoft I, Birkenkamp-Demtröder K, Ahrenfeldt J, Strandgaard T, Radif D, Hodgson D, Abbosh C, Aerts HJWL, Agerbæk M, Jensen JB, Birkbak NJ, Dyrskjøt L. Low T cell diversity associates with poor outcome in bladder cancer: A comprehensive longitudinal analysis of the T cell receptor repertoire. Cell Rep Med 2025; 6:102101. [PMID: 40315845 DOI: 10.1016/j.xcrm.2025.102101] [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: 08/30/2024] [Revised: 11/20/2024] [Accepted: 04/09/2025] [Indexed: 05/04/2025]
Abstract
T cells are crucial effector cells in the endogenous defense against cancer, yet the clinical impact of their quantity, diversity, and dynamics remains underexplored. Here, we investigate the clinical relevance of the T cell receptor (TCR) repertoire in patients with bladder cancer. In advanced-stage disease, low pre-treatment peripheral TCR diversity is associated with worse overall survival (p = 0.024), particularly when coupled with low circulating T cell fractions (p = 0.00049). These low-diversity repertoires are dominated by hyper-expanded clones that persist throughout treatment. Further longitudinal analysis reveals reductions in TCR diversity after treatment, indicating adverse effects on the immune system. In early-stage disease, immunotherapy increases TCR diversity in patients with good outcomes. Furthermore, single-cell sequencing identifies most hyper-expanded clones as cytotoxic T cells, while non-expanded clones are predominantly naive T cells. Overall, this highlights TCR diversity as a promising biomarker, offering opportunities for tailored oncological treatments to enhance clinical outcomes.
Collapse
Affiliation(s)
- Asbjørn Kjær
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Nanna Kristjánsdóttir
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Randi Istrup Juul
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Iver Nordentoft
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Karin Birkenkamp-Demtröder
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Johanne Ahrenfeldt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Deema Radif
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Darren Hodgson
- Cancer Biomarker Development, Oncology R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Christopher Abbosh
- Cancer Biomarker Development, Oncology R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Hugo J W L Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Mads Agerbæk
- Department of Oncology, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; Department of Urology, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Nicolai J Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark.
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark.
| |
Collapse
|
3
|
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
|
4
|
Ziogas DC, Foteinou D, Theocharopoulos C, Martinos A, Petsiou DP, Anastasopoulou A, Gogas H. State-of-the-art in Metastatic Uveal Melanoma Treatment: A 2025 Update : How to treat Metastatic Uveal Melanoma in 2025. Curr Oncol Rep 2025:10.1007/s11912-025-01684-0. [PMID: 40380030 DOI: 10.1007/s11912-025-01684-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2025] [Indexed: 05/19/2025]
Abstract
PURPOSE OF REVIEW Uveal melanoma (UM) is the most common intraocular malignancy in adults, representing a rare but aggressive melanoma subtype with a distinct molecular landscape, unique metastatic behavior and limited therapeutic options in the metastatic setting. This review provides an in-depth analysis of the latest evidence on the evolving treatment landscape of metastatic UM. RECENT FINDINGS For liver-only metastatic disease, locoregional therapies provide significant benefit compared to systemic therapies. The recent approval of tebentafusp-tebn, a bispecific gp100 peptide-HLA-directed CD3 T-cell engager, marks a pivotal advancement for HLA-A*02:01-positive patients with unresectable/metastatic UM, demonstrating a clinically significant survival benefit. Several clinical studies are currently active, examining emerging locoregional and systemic treatments for metastatic UM, with promising early data. Despite effective local disease control through radiotherapy and enucleation, approximately 50% of patients develop metastatic disease, predominantly in the liver, with a median survival of less than one year. The approval of tebentafusp represents a landmark achievement in UM treatment, while promising experimental combinations have demonstrated clinical utility in late phase clinical trials, offering hope for further improvement in patient survival.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Dimitra Foteinou
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalampos Theocharopoulos
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Martinos
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dioni-Pinelopi Petsiou
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Amalia Anastasopoulou
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
5
|
Liu J, Su Y, Zhang C, Dong H, Yu R, Yang X, Tian Y, Feng Y, Zhang J, Shi M, Wang C, Li W, Liu J, He L, Yang X, Liu H. NCOA3 impairs the efficacy of anti-PD-L1 therapy via HSP90α/EZH2/CXCL9 axis in colon cancer. Int Immunopharmacol 2025; 155:114579. [PMID: 40215778 DOI: 10.1016/j.intimp.2025.114579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized colon cancer treatment, but their efficacy is largely restricted by the limited presence of CD8+ cytotoxic T lymphocytes (CTLs). However, the specific genetic alterations that impact the CD8+ CTL infiltration in colon cancer remain poorly understood. Here, we analyzed clinical and multi-omics data from the Memorial Sloan-Kettering Cancer Center (MSKCC) ICIs-treated and The Cancer Genome Atlas (TCGA) colon adenocarcinoma (COAD) cohorts to screen the key mutations that may influence the efficacy of immunotherapy. We found that patients with NCOA3 mutations exhibit better response to immunotherapy and higher CD8+ CTL infiltration. In vitro and in vivo experiments revealed that mutant NCOA3 increases the efficacy of anti-PD-L1 and CD8+ CTL recruitment by upregulating C-X-C motif chemokine ligand 9 (CXCL9), which is dependent on its impaired intrinsic histone acetyltransferase activity. Mechanistically, wild-type NCOA3 as histone acetyltransferase upregulates Heat shock protein 90 alpha (HSP90α) by enhancing histone H3 lysine 27 acetylation (H3K27ac) at its promoter region. Increased HSP90α stabilizes Enhancer of zeste homolog 2 (EZH2), which then increase the histone H3 lysine 27 trimethylation (H3K27me3) at the CXCL9 promoter region, thereby suppressing the expression of CXCL9. Targeted inhibition of NCOA3 by small molecular inhibitor SI-2 improves the efficacy of PD-L1 blockade therapy. NCOA3 could serve as a novel biomarker and potential target to improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Jiaqi Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yixi Su
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Chi Zhang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Haiyan Dong
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Runfeng Yu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xin Yang
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yu Tian
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yanchun Feng
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jingdan Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Mengchen Shi
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Chen Wang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Weiqian Li
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jun Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Lingyuan He
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiangling Yang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| |
Collapse
|
6
|
He M, Ji C, Li Z, Chen S, Gao J, Shen L, Zhang C. Circulating tumor DNA predicts clinical benefits of immune checkpoint blockade in HER2-negative patients with advanced gastric cancer. Gastric Cancer 2025:10.1007/s10120-025-01621-x. [PMID: 40372586 DOI: 10.1007/s10120-025-01621-x] [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: 09/25/2024] [Accepted: 04/29/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are becoming more prominent in the treatment of gastric cancer (GC). However, predictive biomarkers of response to ICIs in HER2-negative patients remain incompletely understood. METHODS A total of 47 patients diagnosed with HER2-negative advanced GC who underwent ICI regimens were eligible for this study. Plasma samples with paired white blood cells prior to treatments were collected from these 47 patients. Variations of circulating tumor DNA (ctDNA) was evaluated by next-generation sequencing followed by its significance analysis. RESULTS A total of 658 somatic mutations involving 203 genes were identified in all ctDNA. Mutations in MEN1, MLH1, CEBPA, ATR, GNAQ, and FOXL2 genes were more frequent in responders (P < 0.05). Compared with wild-type patients, patients with CEBPA or IRS2 mutations had prolonged median progression-free survival (mPFS, P = 0.0056). Patients with co-occurring mutations in IRS2/CEBPA, IRS2/POLD1, TP53/PIK3CA, or POLD1/CEBPA had longer mPFS compared with others (P = 0.003; 0.006; 0.0166; 0.0315; respectively). Both alteration of CDKN2A alone and co-mutations with MSH6 were significantly associated with superior overall survival (OS, P = 0.0289; 0.0355; respectively). In addition, higher on-treatment ctDNA concentration or variant allele frequency (VAF) were associated with poorer response (P < 0.05). Additionally, the increased molecular alterations of POLE, FGFR2 and MDC1 seemed to indicate the acquired resistance to ICIs. CONCLUSIONS Variation signatures captured by ctDNA as well as the kinetics of ctDNA could predict the clinical benefits of ICB in HER2-negative GC patients, which was worth further validated in large cohort.
Collapse
Affiliation(s)
- Mei He
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Congcong Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhiwei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shiqing Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, 201114, China
| | - Jing Gao
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Cheng Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
7
|
Kim M, Gu W, Iwakawa RK, Kina S, Nakajima T, Higuchi T, Ogawa M, Suzuki K, Tsushima Y, Yokoo S. Integrative Analysis of 18F-FDG PET Radiomics and mRNA Expression in Recurrent/Metastatic Oral Squamous Cell Carcinoma: A Cross-Sectional Study. Mol Imaging Biol 2025:10.1007/s11307-025-02012-5. [PMID: 40369386 DOI: 10.1007/s11307-025-02012-5] [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: 10/08/2024] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND This study explored the relationship between mRNA expression profiles obtained through next-generation sequencing (NGS) and 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) texture analysis in patients with treatment-resistant oral squamous cell carcinoma (OSCC) who were treated with molecular-targeted drugs. We analyzed the correlation between 18F-FDG PET texture features and NGS data in a small cohort of five patients with recurrent or metastatic OSCC who received molecular-targeted drugs after surgery. Patients were categorized into two groups based on treatment response: responders (n = 3) and non-responders (n = 2). To validate our findings, we examined transcriptomic data from 292 OSCC patients in The Cancer Genome Atlas (TCGA) database. RESULTS The gene ankyrin repeat and SOCS box containing two (ASB2) was significantly overexpressed in non-responders and strongly correlated with specific PET radiomic features, including GLRLM_GLNU, GLRLM_RLNU, and GLZLM_GLNU (p < 0.05). High ASB2 expression was also associated with poor prognosis in OSCC patients (p < 0.05) and decreased overall survival, as shown by Kaplan-Meier analysis of the TCGA database (p = 0.017). CONCLUSIONS Integrating ASB2 expression data with 18F-FDG PET texture features could potentially improve the prediction of treatment outcomes in treatment-resistant OSCC patients undergoing molecular-targeted therapy.
Collapse
Affiliation(s)
- Mai Kim
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Wenchao Gu
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Reika Kawabata- Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Japan
| | - Shinichiro Kina
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Japan
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Takahito Nakajima
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masaru Ogawa
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Keisuke Suzuki
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Satoshi Yokoo
- Department of Oral and Maxillofacial Surgery, and Plastic Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
8
|
Gonzalez MF. Well-Differentiated Adenocarcinoma With Papillary Architecture, Focal Residual Cilia, Apical Snouts, and CHEK2 and p53 Mutations. Cytopathology 2025. [PMID: 40369890 DOI: 10.1111/cyt.13508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 04/15/2025] [Accepted: 05/03/2025] [Indexed: 05/16/2025]
Abstract
Ciliated adenocarcinomas are rare non-terminal respiratory unit-type lung adenocarcinomas characterised by ciliated cells with nuclear atypia and glandular or papillary architecture. A few cases of ciliated adenocarcinoma of the lung have been reported. These adenocarcinomas are negative for TTF1 and have either KRAS (25%) or EGFR (9%) mutations. A case of a well-differentiated adenocarcinoma with papillary architecture, focal residual cilia, apical snouts, and CHEK2 and p53 mutations diagnosed at stage IV(T4NxM0) is reported. The article describes the cytomorphological features of this tumour, the potential pitfalls, diagnostic limitations, challenges identified in cytology specimens, and the necessary work-up for a definitive diagnosis. The differential diagnosis of papillary lesions of the lung and the clinical significance of CHEK2 mutations are also discussed in this article.
Collapse
Affiliation(s)
- Maria F Gonzalez
- Temple University Hospital and Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Pecora G, Mancini C, Mazzilli R, Zamponi V, Telese S, Scalera S, Maugeri-Saccà M, Ciuffreda L, De Nicola F, Fanciulli M, La Salvia A, Mancini M, Vecchione A, Siciliani A, Ibrahim M, Bellavia D, Isidori AM, Faggiano A, Mancini R, De Vitis C. Genetic insight into lung neuroendocrine tumors: Notch and Wnt signaling pathways as potential targets. J Transl Med 2025; 23:538. [PMID: 40361150 PMCID: PMC12076951 DOI: 10.1186/s12967-025-06442-1] [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: 02/04/2025] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The molecular landscape of lung neuroendocrine neoplasms is still poorly characterized, making it difficult to develop a molecular classification and personalized therapeutic approaches. Significant clinical heterogeneity of these malignancies has been highlighted among poorly differentiated histotypes and within the subgroup of well-differentiated neuroendocrine tumors (NET). Currently, the main prognostic factors of lung NET include stage, histotype, grade, peripheral location, and demographic parameters. To gain deeper insights into the genomic underpinnings of lung NETs, we conducted a pilot investigation to uncover potential genetic mutations and copy number variations (CNVs) implicated in their pathogenesis. METHODS Formalin-fixed, paraffin-embedded intraoperative tumor biopsies and matched peripheral blood mononuclear cell samples were collected from six consecutive patients with lung NETs. The whole exome sequencing (WES) was performed to profile germline and somatic mutations, identify novel genetic alterations, and detect CNVs. Clinical and pathological data were systematically documented at diagnosis and during follow-up. RESULTS The WES analysis identified a subset of mutations shared between germline and somatic; some were of particular clinical interest as they were associated with tumor proliferation and potential therapeutic targets such as the genes KDM5C, ATR, COL7A1, NOTCH4, PTPRS, SMO, SPEN, SPTA1, TAF1. These mutations were predominantly linked to chromatin remodeling and were involved in critical oncogenic pathways such as Notch and Wnt signaling. CONCLUSIONS This pilot study highlights the potential role of NGS analysis on solid biopsy in the assessment of the mutational profile of lung NET. A comparison of germline and somatic mutations is critical to identifying putative tumor driver mutations. In perspective, the enrichment of a subpopulation of cancer cells in the blood, with one or more specific mutations, is information of enormous clinical relevance, either for prognosis or therapeutic decisions. Translational studies on large prospective series are required to establish the role of liquid biopsy in lung NET.
Collapse
Affiliation(s)
- Giulia Pecora
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, ENETS Center of Excellence, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Camilla Mancini
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, ENETS Center of Excellence, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Rossella Mazzilli
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, ENETS Center of Excellence, Rome, Italy
| | - Virginia Zamponi
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, ENETS Center of Excellence, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Stefano Telese
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Stefano Scalera
- Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcello Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ludovica Ciuffreda
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, Translational Research Area, IRCCS Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Francesca De Nicola
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, Translational Research Area, IRCCS Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Maurizio Fanciulli
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies, Translational Research Area, IRCCS Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Anna La Salvia
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), Rome, Italy
| | - Massimiliano Mancini
- Morphologic and Molecular Pathology Unit, S. Andrea University Hospital, Rome, Italy
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital-Sapienza University of Rome, Rome, Italy
- Morphologic and Molecular Pathology Unit, S. Andrea University Hospital, Rome, Italy
| | | | - Mohsen Ibrahim
- Department of Thoracic Surgery, Sant'Andrea University Hospital, Rome, Italy
| | - Diana Bellavia
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Antongiulio Faggiano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, ENETS Center of Excellence, Rome, Italy.
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital-Sapienza University of Rome, Rome, Italy
- Morphologic and Molecular Pathology Unit, S. Andrea University Hospital, Rome, Italy
| | - Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital-Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Wang Z, Dai Z, Gao Y, Zhao Z, Li Z, Wang L, Gao X, Qiu Q, Qiu X, Liu Z. Development of a machine learning-based predictive risk model combining fatty acid metabolism and ferroptosis for immunotherapy response and prognosis in prostate cancer. Discov Oncol 2025; 16:744. [PMID: 40355680 PMCID: PMC12069205 DOI: 10.1007/s12672-025-02484-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Prostate cancer (PCa) remains a leading cause of cancer-related mortality, necessitating robust prognostic models and personalized therapeutic strategies. This study integrated bulk RNA sequencing, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics to construct a prognostic model based on genes shared between ferroptosis and fatty acid metabolism (FAM). Using the TCGA-PRAD dataset, we identified 73 differentially expressed genes (DEGs) at the intersection of ferroptosis and FAM, of which 19 were significantly associated with progression-free survival (PFS). A machine learning-based prognostic model, optimized using the Lasso + Random Survival Forest (RSF) algorithm, achieved a high C-index of 0.876 and demonstrated strong predictive accuracy (1-, 2-, and 3-year AUCs: 0.77, 0.75, and 0.78, respectively). The model, validated in the DFKZ cohort, stratified patients into high- and low-risk groups, with the high-risk group exhibiting worse PFS and higher tumor mutation burden (TMB). Functional enrichment analysis revealed distinct pathway activities, with high-risk patients showing enrichment in immune-related and proliferative pathways, while low-risk patients were enriched in metabolic pathways. Immune microenvironment analysis revealed heightened immune activity in high-risk patients, characterized by increased infiltration of CD8 + T cells, regulatory T cells, and M2 macrophages, alongside elevated TIDE scores, suggesting immune evasion and resistance to immunotherapy. In contrast, low-risk patients exhibited higher infiltration of plasma cells and neutrophils and demonstrated better responses to immune checkpoint inhibitors (ICIs). Spatial transcriptomics and scRNA-seq further elucidated the spatial distribution of model genes, highlighting the central role of macrophages in mediating risk stratification. Additionally, chemotherapy sensitivity analysis identified potential therapeutic agents, such as Erlotinib and Picolinic acid, for low-risk patients. In vitro experiments showed that overexpression of CD38 in the PC-3 cell line led to elevated lipid peroxidation (C11-BODIPY) and reactive oxygen species (ROS), suggesting increased cell ferroptosis. These findings provide a comprehensive framework for risk stratification and personalized treatment in PCa, bridging molecular mechanisms with clinical outcomes.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhihong Dai
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yuren Gao
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhongxiang Zhao
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhen Li
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Liang Wang
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiang Gao
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiuqiu Qiu
- Department of Urology, Gaohou People's Hospital, Maoming, 525200, China.
| | - Xiaofu Qiu
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510317, China.
| | - Zhiyu Liu
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
11
|
Kouhen F, El Ghanmi A, Inghaoun H, Miftah H, Ghazi B, Badou A. The promise of PD1/PDL1 targeted immunotherapy in locally advanced cervical cancer: a game-changer for patients outcome? Front Immunol 2025; 16:1573576. [PMID: 40433369 PMCID: PMC12106400 DOI: 10.3389/fimmu.2025.1573576] [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: 02/09/2025] [Accepted: 04/14/2025] [Indexed: 05/29/2025] Open
Abstract
Locally advanced cervical cancer remains a significant therapeutic challenge, with high rates of recurrence and metastasis despite advances in chemoradiation. Immunotherapy, particularly immune checkpoint inhibitors targeting the PD-1/PD-L1 axis, has emerged as a promising strategy to enhance treatment efficacy. This review explores the integration of immunotherapy with standard chemoradiation, highlighting the potential of PD-1 inhibitors, such as pembrolizumab, in improving progression-free survival (PFS) among high-risk patients. Furthermore, the role of predictive biomarkers, including microsatellite instability (MSI) and tumor mutational burden (TMB), is examined to refine patient selection and personalize therapeutic approaches. Emerging strategies, including the use of nivolumab, ipilimumab, and maintenance immunotherapy, are also discussed. While preliminary clinical data are encouraging, further research is required to optimize treatment combinations, establish robust patient selection criteria, and enhance long-term outcomes in cervical cancer management.
Collapse
Affiliation(s)
- Fadila Kouhen
- Mohammed VI Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Rabat, Morocco
- Laboratory of Neurooncology, Oncogenetic and Personalized Medicine, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Department of Radiotherapy, International University Hospital Cheikh Khalifa, Casablanca, Morocco
| | - Adil El Ghanmi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura, Morocco
| | - Hanane Inghaoun
- Laboratory of Neurooncology, Oncogenetic and Personalized Medicine, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
| | - Hayat Miftah
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Casablanca, Morocco
| | - Bouchra Ghazi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura, Morocco
| | - Abdallah Badou
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Casablanca, Morocco
| |
Collapse
|
12
|
Saito S, Kato S, Arai U, En A, Tsunezumi J, Mizushima T, Tateishi K, Adachi N. HR eye & MMR eye: one-day assessment of DNA repair-defective tumors eligible for targeted therapy. Nat Commun 2025; 16:4239. [PMID: 40355434 PMCID: PMC12069580 DOI: 10.1038/s41467-025-59462-2] [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: 07/04/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Homologous recombination (HR) and mismatch repair (MMR) act as guardians of the human genome, and defects in HR or MMR are causative in at least a quarter of all malignant tumors. Although these DNA repair-deficient tumors are eligible for effective targeted therapies, fully reliable diagnostic strategies based on functional assay have yet to be established, potentially limiting safe and proper application of the molecular targeted drugs. Here we show that transient transfection of artificial DNA substrates enables ultrarapid detection of HR and MMR. This finding led us to develop a diagnostic strategy that can determine the cellular HR/MMR status within one day without the need for control cells or tissues. Notably, the accuracy of this method allowed the discovery of a pathogenic RAD51D mutation, which was missed by existing companion diagnostic tests. Our methods, termed HR eye and MMR eye, are applicable to frozen tumor tissues and roughly predict the response to therapy. Overall, the findings presented here could pave the way for accurately assessing malignant tumors with functional defects in HR or MMR, a step forward in accelerating precision medicine.
Collapse
Affiliation(s)
- Shinta Saito
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Shingo Kato
- Department of Clinical Cancer Genomics, Yokohama City University Hospital, Yokohama, 236-0004, Japan
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Usaki Arai
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Atsuki En
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Jun Tsunezumi
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan
| | - Taichi Mizushima
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Kensuke Tateishi
- Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Noritaka Adachi
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, Yokohama, 236-0027, Japan.
| |
Collapse
|
13
|
Farmanbar A, Kneller R, Firouzi S. The Use of Mutational Signatures to Decipher the Inter-Relationship of Reactive Oxygen Species with Homologous Recombination and Non-Homologous End-Joining Deficiencies as Well as Their Effects on APOBEC Mutagenesis in Breast Cancer. Cancers (Basel) 2025; 17:1627. [PMID: 40427126 DOI: 10.3390/cancers17101627] [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: 04/04/2025] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Defective DNA repair systems result in the accumulation of mutations, loss of genomic integrity, and eventually cancer. Following initial malignant transformation due to specific DNA damage and defective DNA repair, cancer cells become reliant upon other DNA repair pathways for their survival. The co-occurrence of specific repair deficiencies brings catastrophic outcomes such as cell death for cancer cells and thus holds promise as a potential therapeutic strategy. Exploring the co-occurrence and mutual exclusivity of mutational signatures provides valuable knowledge regarding combinations of defective repair pathways that are cooperative and confer selective advantage to cancer cells and those that are detrimental and cannot be tolerated by them. Methods: Taking advantage of mutational signature profiling, we analyzed whole-genome sequences of 1014 breast cancers to reveal the underlying mutational processes and their interrelationships. Results: We found an inverse relationship between deficiencies of homologous recombination (HRd) and non-homologous end joining (NHEJd) with reactive oxygen species (ROS). Moreover, HRd and NHEJd co-occurred with APOBEC but were mutually exclusive with mismatch repair deficiency (MMRd) and ROS. Our analysis revealed that SBS8 and SBS39 signatures of currently unknown etiology correlate with NHEJd. ID1 and ID2 signatures co-occur with ROS and have mutual exclusivity with HRd, SBS8, SBS39 and NHEJd. The ID4 signature, with currently unknown etiology, has mutual exclusivity with HRd and NHEJd and co-occurred with ROS. On the other hand, the ID15 signature, with currently unknown etiology, co-occurred with SBS8, SBS39, HRd, NHEJd and DBS2, while having an inverse relationship with MMRd and ROS. Comparing the mutational signatures of HRd and non-HRd TNBC genomes reveals the unique presence of ROS signatures in non-HRd tumors and the lack of ROS signature in HRd tumors. Conclusion: Taken together, these analyses indicate the possible application of mutation signatures and their interactions in advancing patient stratification and suggest appropriate therapies targeting the make-up of individual tumors' mutational processes. Ultimately, this information provides the opportunity to discover promising synthetic lethal candidates targeting DNA repair systems.
Collapse
Affiliation(s)
- Amir Farmanbar
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
| | - Robert Kneller
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan
| | - Sanaz Firouzi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
14
|
Jeon JE, Chen KT, Madison R, Schrock AB, Sokol E, Levy MA, Rozenblit M, Huang RSP, Pusztai L. Genomic landscape and homologous recombination repair deficiency signature in stage I-III and de novo stage IV primary breast cancers. Oncologist 2025; 30:oyaf089. [PMID: 40421962 DOI: 10.1093/oncolo/oyaf089] [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: 09/24/2024] [Accepted: 03/26/2025] [Indexed: 05/28/2025] Open
Abstract
PURPOSE We compared genomic alterations and a homologous recombination deficiency (HRD) signature (HRDsig) in primary tumors from stage I-III to those in de novo stage IV breast cancers and from stage I-III cancers with early (<2 years after diagnosis) versus late (>2 years) recurrence. METHODS De-identified genomic and clinical data of primary breast cancers (stage I-III N = 910, stage IV N = 783) from the United States, the nationwide clinico-genomic database of Flatiron Health and Foundation Medicine were analyzed. Genomic results included the mutation status of 324 cancer-related genes and HRDsig, a DNA scar-based measure of HRD. RESULTS No significant differences were observed in the frequencies of genomic alterations across disease stages, or between stage I-III cancers with early versus late relapse. Overall, the most prevalent biomarkers were PIK3CA mutation and HRDsig positivity. HRDsig positivity was observed in 82% of germline or somatic g/sBRCA1/2 or germline PALB2 (gPALB2) mutated cancers, 13.1% in cancers with other HR-repair (HRR) gene alterations (ATM, BARD1, BRIP1, CDK12, CHEK1, CHEK2, FANCL, somatic PALB2 (sPALB2), RAD51B, RAD51C, RAD51D and RAD54L), and also in 16.5% of HRR wild-type cancers. HRDsig positivity was observed across receptor subtypes and was the highest in TNBC (30%), followed by ER+/HER2- cancers (17%), then HER2 + cancers (8.7%). CONCLUSIONS Early-stage (I-III) and de novo stage IV breast cancers shared a similar prevalence of targetable genomic alterations and overall genomic landscape. HRDsig identified approximately 16% of breast cancers without g/sBRCA/gPALB2 alteration that might potentially benefit from PARP inhibitors or platinum-based treatments and should be tested in future clinical studies.
Collapse
Affiliation(s)
- June E Jeon
- Department of Internal Medicine, Greenwich Hospital, Greenwich, CT, USA
| | | | | | | | | | | | | | | | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
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
|
16
|
Kakimi K, Sugie T. Why combine and why neoadjuvant? Tumor immunological perspectives on chemoimmunotherapy in triple-negative breast cancer. Breast Cancer 2025:10.1007/s12282-025-01707-5. [PMID: 40327275 DOI: 10.1007/s12282-025-01707-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by limited targeted therapies and high recurrence rates. While immune checkpoint inhibitors (ICIs) have shown promise, their efficacy as monotherapy is limited. Clinically, ICIs demonstrate significant benefit primarily when combined with chemotherapy, particularly in the neoadjuvant setting for early-stage TNBC, which yields superior outcomes compared to adjuvant therapy. This review elucidates the tumor immunological principles underlying these observations. We discussed how the suppressive tumor microenvironment (TME), progressive T cell exhaustion, and associated epigenetic scarring constrain ICI monotherapy effectiveness. Crucially, we highlight the immunological advantages of the neoadjuvant approach: the presence of the primary tumor provides abundant antigens, and intact tumor-draining lymph nodes (TDLNs) act as critical sites for ICI-mediated priming and expansion of naïve and precursor exhausted T cells. This robust activation within TDLNs enhances systemic anti-tumor immunity and expands the T cell repertoire, a process less effectively achieved in the adjuvant setting after tumor resection. These mechanisms provide a strong rationale for the improved pathological complete response (pCR) rates and event-free survival observed with neoadjuvant chemoimmunotherapy, as demonstrated in trials like KEYNOTE-522. We further explore the implications for adjuvant therapy decisions based on treatment response, the challenges of ICI resistance, the need for predictive biomarkers, management of immune-related adverse events (irAEs), and future therapeutic directions. Understanding the dynamic interplay between chemotherapy, ICIs, T cells, and the TME, particularly the role of TDLNs in the neoadjuvant context, is essential for optimizing immunotherapy strategies and improving outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Onohigashi, Osakasayama, Osaka, 589-8511, Japan.
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| | - Tomoharu Sugie
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| |
Collapse
|
17
|
Pakola SA, Clubb JHA, Santos JM, Havunen R, Lassus H, Cervera-Carrascon V, Hemminki A. Oncolytic adenoviruses in platinum-resistant ovarian cancer: a new era in immunotherapy? Expert Opin Biol Ther 2025:1-4. [PMID: 40322861 DOI: 10.1080/14712598.2025.2501731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Affiliation(s)
- Santeri Artturi Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - James Hugo Armstrong Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Joao Manuel Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Heini Lassus
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
18
|
Takano Y, Mizuno K, Iwase M, Morita S, Torii N, Kikumori T, Ando Y. Tumor mutational burden status and clinical characteristics of invasive lobular carcinoma of the breast. Breast Cancer 2025:10.1007/s12282-025-01706-6. [PMID: 40314919 DOI: 10.1007/s12282-025-01706-6] [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: 12/04/2024] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND High tumor mutational burden (TMB-H) is an established biomarker for a favorable response to immune checkpoint inhibitors. However, tumor mutational burden (TMB) in invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) has not been sufficiently investigated. METHODS We collected data of patients with ILC or IDC from the Center for Cancer Genomics and Advanced Therapeutics database between June 2019 and August 2023. Furthermore, we examined the clinicopathological factors and TMB status. RESULTS Patients with ILC (n = 170) had a median TMB score of 4.00 mut/Mb (interquartile range, 2.00-7.14 mut/Mb), whereas those with IDC (n = 2598) had a score of 3.90 mut/Mb (2.00-6.00 mut/Mb). TMB-H was more common in patients with ILC than in those with IDC (18.2% vs. 10.1%, P < 0.001), particularly in the ER+ /HER2- subtype. Multivariate analysis revealed that the pathological diagnosis of ILC (P = 0.006), tissue samples collected from metastatic sites (P < 0.001), and older age (50 years, P < 0.001) were independent factors for TMB-H. CONCLUSIONS Patients with ILC were more likely to have TMB-H than those with IDC. The findings of this study would be invaluable in selecting treatment strategies for patients with ILC.
Collapse
Affiliation(s)
- Yuko Takano
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan.
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan.
| | - Kazuyuki Mizuno
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Madoka Iwase
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Sachi Morita
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Nao Torii
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Toyone Kikumori
- Department of Breast and Endocrine Surgery, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
19
|
Kang I, Naghi L, Yost SE, Mortimer J. Clinical Actionability of Molecular Targets in Multi-Ethnic Breast Cancer Patients: A Retrospective Single-Institutional Study. Mol Diagn Ther 2025; 29:393-405. [PMID: 40192953 PMCID: PMC12062054 DOI: 10.1007/s40291-025-00777-7] [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] [Accepted: 03/03/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Precision oncology is making remarkable advancements in optimizing patient care by personalizing treatments. To date, the US Food and Drug Administration (FDA) has approved poly(ADP-ribose) polymerase inhibitors (PARPi) olaparib (Lynparza, AstraZeneca and Merck) and talazoparib (Talzenna, Pfizer Oncology Together™) for germline or somatic BRCA1/2-mutated metastatic breast cancer (BC) patients, and PI3K inhibitor alpelisib (Piqray, Novartis) plus fulvestrant for patients with hormone receptor-positive human epidermal growth factor receptor 2-negative (HR+HER2-) PIK3CA-mutated advanced BC. In addition, the FDA approved capivasertib (Trucap, AstraZeneca) for HR+HER2- locally advanced or metastatic BC patients with one or more AKT1, PIK3CA, or PTEN alterations. Finally, the FDA recently approved elacestrant (Orserdu, Stemline Therapeutics, Inc.) for postmenopausal patients with ER+ HER2- ESR1-mutated advanced or metastatic BC with disease progression following at least one line of endocrine therapy. METHODS This study presents a single institutional retrospective review of genomic reports of patients with BC. Analysis of genomic reports of 1361 BC sequencing reports was performed for 1010 patients with BC from 2013 to 2023 (23% of patients had multiple reports). Eligible patients had at least one primary or metastatic tumor. Multiple sequencing platforms were used for FFPE specimens including Tempus xT targeted next-generation sequencing (NGS), Foundation One Medicine, HopeSeq, Ashion Analytics GEM ExTra, and Exact Sciences Oncomap. Liquid biopsies were performed by Guardant, Tempus, and Foundation One Medicine. Chart reviews were performed to collect patient characteristics. BRCA1/2-mutated, metastatic BC patients who initiated treatment with olaparib or talazoparib, and PIK3CA-mutated, HR+ metastatic BC patients who initiated treatment with alpelisib were reported. In addition, patients with ESR1 or AKT1/PIK3CA/PTEN mutations were identified. Clinical outcomes, including progression-free survival (PFS) and overall survival (OS) were analyzed for BRCA1/2 and PIK3CA-mutated patients who received PARPi or alpelisib. Survival curves were generated using the Kaplan Meier method. RESULTS A cohort of 1010 BC patients with 1361 genomic reports was identified. A total of 935/1361 (69%) specimens were formalin-fixed paraffin-embedded (FFPE) tumor biopsies and 426/1361 (31%) were liquid biopsies. Receptor status included 65% HR+HER2-, 8% HR+HER2+, 4% HR-HER2+, and 23% TNBC. Racial and ethnic distribution of these patients included 50% non-Hispanic White, 26% Hispanic, 17% Asian, 6% African American, 1% other (Native Hawaiian or Other Pacific Islander, American Indian or Alaska Native, or unknown). Sequencing platforms included 30% Tempus xT, 31% Foundation One, 10% HopeSeq, 20% GEM ExTra, and 9% Exact Sciences. Liquid biopsies included 79% Guardant, 20% Tempus, and 1% Foundation One. Of 1010 patients, the most common mutations were TP53 (44%), PIK3CA (38%), ESR1 (14%), PTEN (12%), CCND1 (11%), FGFR1 (10%), CDH1 (10%), ERBB2 (9%), MYC (9%), FGF3 (8%), GATA3 (8%), FGF19 (8%), FGF4 (7%), ARID1A (6%), RB1 (5%), BRCA2 (5%), MAP3K1 (4%), AKT1 (4%), NF1 (4%), MLL3 (4%), ZNF703 (4%), CDKN2A (4%), BRCA1 (4%), MCL1 (3%), ATM (3%), PALB2 (1%), and CHEK2 (1%). The majority of reports with tumor mutation burden (TMB) results (97%) had low or intermediate TMB. A total of 784 actionable mutations in 1010 patients were reported, including 381/1010 (38%) PIK3CA; 144/1010 (14%) ESR1; 122/1010 (12%) PTEN; 48/1010 (5%) BRCA2; 36/1010 (4%) BRCA1; 41/1010 (4%) AKT1; and 12/1010 (1%) PALB2. Of the 96/1010 (10%) patients with BRCA1, BRCA2, or PALB2 mutations not including variants of uncertain significance (VUS), 33/96 (34.4%) received olaparib and 3/96 (3%) received talazoparib in the metastatic setting, and 28 were eligible for response (one had toxicity, two were lost to follow-up, and two went to hospice). Median PFS was 9.0 months and median OS was 21.8 months for patients receiving PARPi. Of the 381/1010 (38%) patients with PIK3CA mutations, 84/381 (22%) received alpelisib and 41 were eligible for response (22 had toxicity, 13 were discontinued, six were lost to follow-up, and two went to hospice). Median PFS was 7.9 months and median OS was 31.2 months for patients receiving alpelisib. A total of 544/1010 (54%) patients had AKT1, PIK3CA, or PTEN mutations which are now FDA approved for capivasertib in HR+HER2- metastatic BC patients. In addition, 144/1010 (14%) patients had ESR1 mutations which are FDA approved for elacestrant in HR+HER2- metastatic BC patients. CONCLUSIONS In this study, a total of 784 clinically actionable mutations were reported for 1010 patients with genomic sequencing. Of these, 96/1010 (10%) patients had at least one actionable mutation in homologous recombination repair genes (BRCA1, BRCA2, PALB2) and 36/96 (37.5%) patients received PARP inhibitors (33 olaparib and three talazoparib). In addition, 381/1010 (38%) patients had at least one clinically actionable PIK3CA mutation, and 84/381 (22%) received alpelisib. Additionally, 544/1010 (54%) of patients had either AKT1 (41/1010), PIK3CA (381/1010), or PTEN (122/1010) alterations that were FDA approved in November 2023 for capivasertib in the treatment of HR+HER2- metastatic BC (MBC) patients. Furthermore, 144/1010 (14%) patients in this study had at least one ESR1 mutation, a clinically actionable mutation that was FDA approved in January 2023 for elacestrant in the treatment of ER+HER2- MBC patients (44% detected by liquid biopsy). Future studies are needed to determine the efficacy of elacestrant and capivasertib for patients with these mutations, and to tailor strategies for optimal patient quality of life and cancer outcome.
Collapse
Affiliation(s)
- Irene Kang
- Department of Medical Oncology, City of Hope National Medical Center, 1000 Fivepoint, Irvine, CA, 92618, USA.
| | - Leah Naghi
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Susan E Yost
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Joanne Mortimer
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
20
|
Hida T. Genomic profiling and personalized treatment strategies for skin malignancies: findings from the center for cancer genomics and advanced therapeutics database. Int J Clin Oncol 2025; 30:856-866. [PMID: 40156656 DOI: 10.1007/s10147-025-02755-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors and molecular-targeted therapies have dominated recent cancer treatment. However, these treatments face challenges, such as primary and acquired resistance, indicating that not all patients benefit from them. Therefore, the search for new molecular targets is crucial. In addition, immune checkpoint inhibitors have exhibited racial differences in their effectiveness for certain neoplasms. Hence, understanding the genomic landscape of cancers in various racial groups is important. In Japan, health insurance has covered comprehensive genomic profiling since 2019, and the Center for Cancer Genomics and Advanced Therapeutics (C-CAT) has accumulated genetic abnormalities along with clinical data of patients with various cancers. These data are crucial for advancing cancer research and drug development. This review discusses the genetic abnormalities of the major skin malignancies including melanoma, cutaneous squamous cell carcinoma (cSCC), and extramammary Paget's disease (EMPD), and proposes potential treatment strategies by comparing C-CAT data analysis with other genetic studies. The C-CAT data have emphasized unique genetic alterations in tumors of the Japanese population, particularly racial differences in tumor mutational burden in cutaneous melanoma and cSCC, indicating the importance of personalized treatment strategies that consider racial differences.
Collapse
Affiliation(s)
- Tokimasa Hida
- Department of Dermatology, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-Ku, Sapporo, 060-8543, Japan.
| |
Collapse
|
21
|
Brada MD, Karakulak T, Schraml P, Haberecker M, Rutishauser D, Ross JS, Eberli D, Moch H. AXL and SRC in clear cell renal cell carcinoma: absence of mutations, rare alternative splicing events, but association of protein expression with poor prognosis. J Pathol Clin Res 2025; 11:e70028. [PMID: 40260977 PMCID: PMC12012843 DOI: 10.1002/2056-4538.70028] [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: 09/07/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025]
Abstract
Novel treatment options for metastatic renal cell carcinomas (RCC) include specific MET inhibitors, GAS6/AXL inhibitors, and SRC inhibitors. The interplay between c-MET, SRC, AXL expression, and their gene mutation patterns in different renal carcinoma subtypes is unclear. To improve the understanding of these signaling pathways, we analyzed c-MET, AXL, and SRC expression in 590 clear cell RCC (ccRCC) and 127 papillary RCC (pRCC) by immunohistochemistry and integrated sequencing data to investigate the frequency of MET, AXL, and SRC gene mutations, their expression levels, and the presence of splice variants. In TCGA and in Foundation Medicine, Inc. (FMI) datasets, AXL and SRC gene alterations were extremely rare (<2%) or absent in ccRCC (n = 531 and 2,781, respectively) and pRCC (n = 290 and 566, respectively). On the other hand, MET mutations or amplifications were found in 9.7% (TCGA) and 10.2% (FMI) of pRCC. We show that strong SRC staining intensity by immunohistochemistry is associated with high tumor stage, high grade, and shorter survival in ccRCC (p < 0.001 each). AXL expression correlates with high stage and grade in ccRCC (p < 0.001 each). Both SRC and AXL expression were independent prognostic parameters in multivariate analysis (p < 0.05). MET expression is associated with longer survival in pRCC (p < 0.05). Our TCGA data analysis aligns with SRC immunohistochemistry findings on tumor stage and shorter survival in ccRCC. TCGA expression data showed a moderate positive correlation between AXL and c-MET in pRCC. In addition, we identified alternative splicing events reported for AXL in pRCC, and MET and SRC in ccRCC, across various alternative splicing databases. In conclusion, we identified high SRC expression as a biomarker for poor prognosis of ccRCC. Our data demonstrate c-MET, AXL, and SRC signaling pathway interactions independent of c-MET, AXL, and SRC mutations in ccRCC.
Collapse
Affiliation(s)
- Muriel D Brada
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
| | - Tülay Karakulak
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Peter Schraml
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
| | - Martina Haberecker
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
| | - Dorothea Rutishauser
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
| | - Jeffrey S Ross
- Foundation Medicine, Inc.CambridgeMAUSA
- Department of PathologyUpstate Medical UniversitySyracuseNYUSA
- Department of UrologyUpstate Medical UniversitySyracuseNYUSA
- Department of Medicine (Oncology)Upstate Medical UniversitySyracuseNYUSA
| | - Daniel Eberli
- Department of UrologyUniversity Hospital ZurichZurichSwitzerland
- University of ZurichZurichSwitzerland
| | - Holger Moch
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
- University of ZurichZurichSwitzerland
| |
Collapse
|
22
|
Szabó IL, Emri G, Ladányi A, Tímár J. Clinical Applications of the Molecular Landscape of Melanoma: Integration of Research into Diagnostic and Therapeutic Strategies. Cancers (Basel) 2025; 17:1422. [PMID: 40361349 PMCID: PMC12071057 DOI: 10.3390/cancers17091422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The molecular landscape of cutaneous melanoma is complex and heterogeneous, and a deeper understanding of the genesis and progression of the tumor driven by genetic alterations is essential for the development of effective diagnostic and therapeutic strategies. Molecular diagnostics and the use of biomarkers are increasingly playing a role in treatment decisions. However, further research is urgently needed to elucidate the relationships between complex genetic alterations and the effectiveness of target therapies (although BRAF mutation is still the only targeted genetic alteration). Further research is required to exploit other targetable genetic alterations such as NRAS, KIT or rare mutations. Treatment guidelines for cutaneous melanoma are continually evolving based on data from recent and ongoing clinical trials. These advancements reflect changes mainly in the optimal timing of systemic therapy and the choice of combination therapies increasingly tailored to molecular profiles of individual tumors. Mono- or combination immunotherapies demonstrated unprecedented success of melanoma treatment; still, there is room for improvement: though several factors of primary or acquired resistance are known, they are not part of patient management as biomarkers. The novel developments of cancer vaccines to treat melanoma (melanoma-marker-based or personalized neoantigen-based) are encouraging; introduction of them into clinical practice without proper biomarkers would be the same mistake made in the case of first-generation immunotherapies.
Collapse
Affiliation(s)
- Imre Lőrinc Szabó
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (I.L.S.); (G.E.)
- HUN-REN-UD Allergology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Gabriella Emri
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (I.L.S.); (G.E.)
- HUN-REN-UD Allergology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Andrea Ladányi
- Department of Surgical and Molecular Pathology, National Institute of Oncology, 1122 Budapest, Hungary;
- National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary
| |
Collapse
|
23
|
Zhang X, Zhang P, Dong H, Li L, Lu L, Lv H, Yu X, Yu H. Homologous Recombination Deficiency Is Associated with Shorter Survival in Colorectal Cancer Patients. J Gastrointest Cancer 2025; 56:105. [PMID: 40261491 DOI: 10.1007/s12029-025-01231-x] [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] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) patients benefit more from immune checkpoint inhibitor therapy, but they only account for around 15% of all patients. The remaining patients still lack effective therapeutic biomarkers to predict their prognosis. METHODS We performed whole-exome sequencing (WES) to analyze 84 CRC specimens, classifying them into different groups based on their microsatellite status (MS), tumor mutation burden (TMB), homologous recombination deficiency (HRD) score, and clinicopathological features, which might be associated with clinical outcomes. Survival analysis and multivariable Cox regression modeling were employed to identify prognostic indicators. Comparative genomic profiling evaluated somatic mutations, copy number variations (CNVs), and pathway activation patterns across clinical subgroups. RESULTS The characteristics of the cohort (N = 84) revealed a median age of 52 years, with a male predominance (61.9%) and a majority of patients presenting with stage IV disease (77%). The HRD-high (HRD-H) subgroup accounted for 16.7%, while 19.0% of cases were microsatellite instability-high (MSI-H) and 22.6% were TMB-high (TMB-H). Multivariable analysis identified HRD-H as an independent predictor of overall survival (OS: HR = 0.19, 95% CI 0.12-0.94, p = 0.002). Comparative genomics demonstrated distinct mutation landscapes between HRD-H and HRD-low subgroups. In microsatellite-stable (MSS) patients, HRD-H status correlated with enriched SMAD4 mutations (p < 0.01) and differential activation of TGF-β/MYC signaling pathways compared to HRD-H-MSI counterparts. CONCLUSION HRD status serves as a novel independent prognostic biomarker in CRC. Our multi-parametric genomic framework delineates stratification-specific molecular signatures, advocating for HRD-integrated molecular diagnostics to optimize CRC management.
Collapse
Affiliation(s)
- Xuan Zhang
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, School of Medicine, Sir Runrun Shaw Hospital, Zhejiang University, Zhejiang, 310016, P.R. China
- Zhejiang Provincial Key Laboratory of Digital Medical Diagnostic Technology, DiAn Diagnostic Group Co., Ltd. Hangzhou 310020, Zhejiang, P.R. China
| | - Pan Zhang
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, 219 Miao Pu Road, Shanghai, 200135, China
| | - Hua Dong
- Zhejiang Provincial Key Laboratory of Digital Medical Diagnostic Technology, DiAn Diagnostic Group Co., Ltd. Hangzhou 310020, Zhejiang, P.R. China
| | - Lin Li
- Zhejiang Provincial Key Laboratory of Digital Medical Diagnostic Technology, DiAn Diagnostic Group Co., Ltd. Hangzhou 310020, Zhejiang, P.R. China
| | - Lingling Lu
- Zhejiang Provincial Key Laboratory of Digital Medical Diagnostic Technology, DiAn Diagnostic Group Co., Ltd. Hangzhou 310020, Zhejiang, P.R. China
| | - Hongyuan Lv
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, School of Medicine, Sir Runrun Shaw Hospital, Zhejiang University, Zhejiang, 310016, P.R. China
| | - Xin Yu
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, School of Medicine, Sir Runrun Shaw Hospital, Zhejiang University, Zhejiang, 310016, P.R. China.
| | - Hong Yu
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, School of Medicine, Sir Runrun Shaw Hospital, Zhejiang University, Zhejiang, 310016, P.R. China.
| |
Collapse
|
24
|
Aobo Z, Xiao Z, Chengfei X, Zhe X, Yingxue C, Chenhe Z, Fuan X, Fan Y, Mengmeng X, Feng Y, Wengang L. Combination of immune checkpoint inhibitors and anthracyclines as a potential first-line regimen for dedifferentiated liposarcoma: systematic review and meta-analysis. Cancer Immunol Immunother 2025; 74:179. [PMID: 40257618 PMCID: PMC12011665 DOI: 10.1007/s00262-025-04007-1] [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: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Dedifferentiated liposarcoma (DDLPS) is a rare and aggressive subtype of soft tissue sarcoma, characterized by limited treatment options and poor prognosis. Despite surgical resection being the only potentially curative treatment for localized DDLPS, the recurrence rate remains high, and systemic chemotherapy, typically anthracycline-based, shows limited efficacy in advanced stages. While immune checkpoint inhibitors (ICIs) have shown promise in various sarcoma subtypes, including DDLPS, their role as a first-line treatment remains unclear. METHODS We conducted a systematic meta-analysis to evaluate the efficacy of ICIs in treating patients with DDLPS. A total of 25 studies encompassing 245 patients were included. Data on overall response rate (ORR), progression-free survival, and grade III-V treatment-related adverse events were analyzed. We assessed treatment efficacy based on the line of therapy and treatment regimens, including ICI monotherapy, dual ICI therapy, and ICI combinations with other modalities. RESULTS The pooled ORR for all ICI-based treatments was 7%. First-line ICI therapy yielded a significantly higher ORR of 22%, compared to 4% in later-line treatment. The combination of ICI with anthracyclines demonstrated the highest ORR of 52%. In contrast, ICI regimens combined with trabectedin or other agents showed limited efficacy. Sensitivity analysis confirmed the stability of results, and publication bias was not detected. CONCLUSION This meta-analysis supports the potential role of ICIs, particularly in combination with anthracyclines, as a first-line therapeutic strategy for DDLPS. These results provide a foundation for future prospective studies aimed at optimizing immunotherapy approaches for this rare and challenging malignancy.
Collapse
Affiliation(s)
- Zhuang Aobo
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhou Xiao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xu Chengfei
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Fujian, 361102, China
| | - Xi Zhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chen Yingxue
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhang Chenhe
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xie Fuan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Fan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Mengmeng
- Department of Retroperitoneal Tumor Surgery, Peking University People's Hospital, Beijing, China.
| | - Ye Feng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Li Wengang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
25
|
QUAN JINGDAN, WAN ZIXIN, WU WEI, CAO XINYUAN, QIU JIAYUAN, LIU XIAOYE, ZHANG ZHIWEI. Classical biomarkers and non-coding RNAs associated with diagnosis and treatment in gastric cancer. Oncol Res 2025; 33:1069-1089. [PMID: 40296904 PMCID: PMC12034007 DOI: 10.32604/or.2025.063005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
One of the most prevalent malignant tumors worldwide, stomach cancer still has a high incidence and fatality rate in China, and the number of young people developing early-onset gastric cancer is steadily increasing. The 5-year survival rate of stomach cancer is typically 30%-35%, the prognosis is bad, the patients' quality of life is low, and the progression of advanced gastric cancer cannot be effectively managed despite the use of surgical surgery, chemotherapy, and other medicines. We urgently need molecular biomarkers with high specificity and sensitivity to increase the early gastric cancer detection rate, extend patient survival, and improve patient quality of life. The initial diagnosis of gastric cancer primarily depends on gastroscopy and biopsy, and invasive procedures cause significant discomfort to patients. Similar to this, treating advanced and metastatic stomach cancer is a pressing issue that requires attention. More and more immune checkpoint molecules have been discovered, and corresponding inhibitors are gradually being applied to clinical diagnosis and treatment. Recently, some non-coding RNAs have begun to be used as new targets for the treatment of gastric cancer. Some non-coding RNAs are highly present in the serum or urine of gastric cancer patients and can be used as diagnostic markers or prognostic indicators. Many clinical trials targeting non-coding RNAs have also shown good therapeutic effects. In general, targeting non-coding RNAs has shown good therapeutic effects. The biomarkers for gastric cancer detection and treatment are reviewed in this article, focusing on the new non-coding RNAs used in diagnosis, prognosis, and treatment. Patients with stomach cancer should have access to more precise and efficient diagnosis and treatment choices as a result of ongoing technological advancements and thorough research.
Collapse
Affiliation(s)
- JINGDAN QUAN
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - ZIXIN WAN
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - WEI WU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - XINYUAN CAO
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - JIAYUAN QIU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - XIAOYE LIU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - ZHIWEI ZHANG
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| |
Collapse
|
26
|
Wang D, Wang Y, Peng Y, Peng L. Utilizing multi-omics analysis, a new signature has been identified and validated for predicting prognosis and response to immunotherapy in lung squamous cell carcinoma, which is based on tumor mutation burden. Discov Oncol 2025; 16:539. [PMID: 40240626 PMCID: PMC12003222 DOI: 10.1007/s12672-025-02166-2] [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: 08/30/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Immunotherapy is used extensively in treating non-small cell lung cancer (NSCLC) patients. Nevertheless, in contrast to lung adenocarcinoma (LUAD), the endeavors to develop effective targeted treatments for lung squamous cell carcinoma (LUSC) have not yielded positive outcomes. Hence, it is crucial to discover biomarkers for immunotherapy and investigate more potent treatments, which is an immediate requirement for individuals with LUSC. The LUSC somatic mutation data were obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Multivariate analysis was performed to create a signature related to tumor mutation burden (TMB). Next, we utilized the CIBERSORT algorithm to assess the correlation between TMB and immune infiltrates. Additionally, we identified prognostic immune cells of LUSC through Kaplan-Meier analysis. The TCGA and ICGC cohorts covered a combined total of 11 genes that were frequently mutated. SYNE1 and TTN mutation correlated with an increased TMB and suggested a positive clinical outlook. A TMB-related signature (SYNE1 and TTN) was constructed based on this. The outlook for the high-risk group in LUSC was considerably poorer than the low-risk group (p = 0.004). In LUSC, there was a correlation between the TMB-related signature and immune infiltrates, and a positive response to anti-PD-L1 therapy was observed in individuals with low-risk scores. Furthermore, based on Kaplan-Meier analysis, plasma cells were identified as predictive immune cells in LUSC samples. In conclusion, the GSEA examination demonstrated that the TMB-associated signature stimulated immune system-related signaling pathways. To sum up, the TMB-associated signature could be marker to anticipate the immune reaction in individuals with LUSC.
Collapse
Affiliation(s)
- Dongguang Wang
- Department of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, China
- The Innovation and Entrepreneurship Education Center for Agricultural Biotechnology of Hunan Province, Loudi, 417000, China
| | - Yan Wang
- Department of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, China
- The Innovation and Entrepreneurship Education Center for Agricultural Biotechnology of Hunan Province, Loudi, 417000, China
| | - Yiqun Peng
- Department of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, China
- The Innovation and Entrepreneurship Education Center for Agricultural Biotechnology of Hunan Province, Loudi, 417000, China
| | - Liang Peng
- Department of Nephrology, The Second Affiliated Hospital of University of South China, Hengyang, 421001, China.
| |
Collapse
|
27
|
Zhang N, Li J, Ren Y, Xu Y. Comprehensive pan-cancer analysis of PPP1R3G reveals its diagnostic, prognostic, and immunotherapeutic implications. Discov Oncol 2025; 16:530. [PMID: 40232629 PMCID: PMC12000506 DOI: 10.1007/s12672-025-02361-1] [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: 12/04/2024] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND PPP1R3G, a regulatory subunit of protein phosphatase 1, plays a critical role in glycogen metabolism and has been implicated in various cancers. This study provides a comprehensive pan-cancer analysis of PPP1R3G, evaluating its expression, diagnostic and prognostic significance, and potential as a therapeutic target. METHODS We performed an extensive pan-cancer analysis of PPP1R3G using several databases to assess its expression and investigate its correlations with clinical outcomes. Our investigation included assessing PPP1R3G's impact on survival, its correlation with immune checkpoints and tumor stemness scores, and its prognostic significance. We also explored its relationship with immunomodulators, genomic profiles, and immunological characteristics, as well as its response to immunotherapy and involvement in various biological pathways. RESULTS PPP1R3G expression varied significantly across different cancers and correlated with both diagnostic and prognostic outcomes. Moreover, PPP1R3G was significantly linked to immune checkpoints, immunomodulators, prognosis, immunoregulatory genes, tumor stemness, cellular function, and immune infiltration across numerous cancer types. Further analysis of PPP1R3G-related gene enrichment, mutation profiles, RNA modifications, and genomic heterogeneity revealed that missense mutations were the predominant alteration affecting PPP1R3G. CONCLUSIONS Overall, the expression of PPP1R3G is closely associated with various cancers and may serve as a potential biomarker for cancer detection.
Collapse
Affiliation(s)
- Nie Zhang
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Jiaoyu Li
- Graduate School of Anhui Medical University, Hefei, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Hefei, China
| | - Yanzhi Ren
- Department of Cardiology, Shizhong District People's Hospital, Zaozhuang, China
| | - Yahui Xu
- Graduate School of Anhui Medical University, Hefei, China.
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
28
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
29
|
Oka T, Smith SS, Oliver-Garcia VS, Lee T, Son HG, Mortaja M, Azin M, Garza-Mayers AC, Huang JT, Nazarian RM, Horn TD, Demehri S. Epigenomic regulation of stemness contributes to the low immunogenicity of the most mutated human cancer. Cell Rep 2025:115561. [PMID: 40250424 DOI: 10.1016/j.celrep.2025.115561] [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: 05/14/2024] [Revised: 12/02/2024] [Accepted: 03/24/2025] [Indexed: 04/20/2025] Open
Abstract
Despite harboring the highest tumor mutational burden of all cancers, basal cell carcinoma (BCC) has low immunogenicity. Here, we demonstrate that BCC's low immunogenicity is associated with epigenomic suppression of antigen presentation machinery reminiscent of its cell of origin. Primary BCC had low T cell infiltrates and low human leukocyte antigen class I (HLA-I) expression compared with cutaneous squamous cell carcinoma (SCC) and normal keratinocytes. Forkhead box C1 (Foxc1), a regulator of quiescence in hair follicle stem cells, was expressed in BCC. Foxc1 bound to promoter of interferon regulatory factor 1 and HLA-I genes, leading to their deacetylation and reduced expression. A histone deacetylase inhibitor, entinostat, overcame Foxc1's effect and upregulated HLA-I in BCC. Topical entinostat plus imiquimod immunotherapy blocked BCC development in mice. Collectively, our findings demonstrate that low BCC immunogenicity is associated with a stem-like quiescent program preserved in the tumor cells, which can be blocked to enable BCC immunotherapy.
Collapse
Affiliation(s)
- Tomonori Oka
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sabrina S Smith
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Valeria S Oliver-Garcia
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Truelian Lee
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Heehwa G Son
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Mahsa Mortaja
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marjan Azin
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna C Garza-Mayers
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer T Huang
- Dermatology Section, Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rosalynn M Nazarian
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas D Horn
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Wang Z, Cheng L, Li G, Cheng H. Epithelial and macrophage cell interaction in cervical cancer through single-cell RNA-sequencing and spatial analysis. Front Immunol 2025; 16:1537785. [PMID: 40270962 PMCID: PMC12014682 DOI: 10.3389/fimmu.2025.1537785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Background Cervical cancer (CC) is a major global health issue, ranking sixth in cancer-related mortality. The tumor microenvironment (TME) plays a crucial role in tumor growth. This study explored the cellular composition and immunological landscape of CC using various genomic data sources. Methods Data from the Cancer Genome Atlas and Gene Expression Omnibus were analyzed, including single-cell RNA sequencing, spatial transcriptome analysis, and survival data. Gene set variation analysis (GSVA) identified pathways in CD8+ cells, macrophages, and epithelial cells. Immunohistochemistry assessed marker expression in CC and normal tissues. Tumor immune dysfunction and exclusion (TIDE) scores differentiated high- and low-macrophage groups. Cell-cell communication analyses highlighted interactions between macrophages and epithelial cells. Results Macrophage markers correlated with overall survival (OS) and disease-free survival (DFS). Epithelial cell subgroups 1 and 4, along with CD8+ T cells, were associated with OS. TIDE scores varied between groups. Specific ligand-receptor interactions were found between macrophages and epithelial cell subgroup 1. Triptolide was effective in epithelial cell subgroup 1, while memantine was more effective in macrophages. Conclusion Epithelial-macrophage interactions in the TME are crucial for CC progression and treatment, offering a potential immunotherapeutic strategy.
Collapse
Affiliation(s)
- Zhichao Wang
- Department of Pediatric Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Long Cheng
- Department of Intensive Care Unit, First Hospital of Jilin University, Changchun, Jilin, China
| | - Guanghui Li
- Department of Vascular Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huiyan Cheng
- Department of Obstetrics and Gynecology, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
31
|
Kou Z, Zhu S, Zhu J, Wang S, Zheng Y, Zhou S, Si Z, Zhu H. Multi-omics analysis identifies DLX4 as a novel biomarker for diagnosis, prognosis, and immune infiltration: from pan-cancer to renal cancer. Discov Oncol 2025; 16:467. [PMID: 40186710 PMCID: PMC11972278 DOI: 10.1007/s12672-025-02258-z] [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: 11/04/2024] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND DLX4 is involved in the regulation of embryonic development, but its function in cancer remains unclear. Here, we conducted a pan-cancer analysis to investigate the molecular mechanisms of DLX4, with a particular emphasis on its role in renal cancer. METHODS A comprehensive analysis of DLX4 was performed, focusing on differences in expression, prognostic value, somatic mutations, methylation modifications, and immune landscapes across various cancer types using multiple databases. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were utilized to explore the potential biological functions. Additionally, we evaluated the expression profile, prognostic significance, and immune infiltration of DLX4 in Kidney Renal Clear Cell Carcinoma (KIRC). The effect of DLX4 on KIRC was further validated by Spatial Transcriptomics, Real-time PCR (RT-PCR), and Immunohistochemistry experiments. RESULTS DLX4 was found to be upregulated in 26 cancer types and associated with poor prognosis. It was also correlated with tumor mutational burden (TMB), microsatellite instability, mismatch repair, and methylation, and was significantly enriched in pathways related to cell proliferation. In KIRC, DLX4 expression increased along with TMB and immune scores, likely due to the infiltration of regulatory T cells (Tregs) and T-helper 2 (Th2) cells. Spatial transcriptomics revealed a strong correlation between DLX4 localization and tumor cells. Experimental validation confirmed that DLX4 expression is significantly upregulated in renal cancer tissues. CONCLUSION Our study explored the mechanisms of DLX4 in pan-cancer, especially in renal clear cell carcinoma, identifying it as a promising biomarker and therapeutic target.
Collapse
Affiliation(s)
- Zengshun Kou
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, China
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Shuaizhi Zhu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, China
- Qingdao West Coast New Area District Hospital, Qingdao, China
| | - Jiaxi Zhu
- Faculty of Arts & Science, University of Toronto - St. George Campus, Toronto, Canada
| | - Shufei Wang
- College of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yu Zheng
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, China
| | - Shengjie Zhou
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, China
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Zi'ang Si
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, China
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, Shandong, China.
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
32
|
Braganca Xavier C, Guardia GDA, Alves JPB, Lopes CDH, Awni BM, Campos EF, Jardim DL, Galante PAF. Identifying predictors of overall survival among patients with TMB-low metastatic cancer treated with immune checkpoint inhibitors. Oncologist 2025; 30:oyaf078. [PMID: 40285678 PMCID: PMC12032576 DOI: 10.1093/oncolo/oyaf078] [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: 10/31/2024] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have significantly advanced cancer therapy, yet their efficacy in tumors with low tumor mutational burden (TMB) remains suboptimal. In this study, we aimed to elucidate the impact of somatic mutations on overall survival (OS) in TMB-low patients treated with ICIs and to explore the potential for personalized treatment selection through machine learning. METHODS We conducted a comprehensive analysis of 1172 TMB-low (TMB < 10 mutations per megabase) patients with cancer receiving ICIs, examining the association between specific gene mutations and OS. Additionally, we developed a decision tree model (DTM) to predict OS based on clinical features and tumor mutational profiles. RESULTS Our findings reveal that mutations in DAXX, HLA-A, H3C2, IGF1R, CTNNB1, SMARCA4, KMT2D, and TP53 are significantly associated with poorer survival outcomes in the multivariate analysis. Remarkably, for renal cell carcinoma (RCC) patients, VHL mutations predicted improved OS following ICI even when adjusted for age, sex, and microsatellite instability (MSI) status in both multivariate analysis and the DTM model. CONCLUSIONS These results reinforce the prevailing notion that TMB alone does not predict ICI response, highlighting the critical role of individual gene mutations in TMB-low tumors under ICI therapy. Furthermore, our study demonstrates the promise of machine learning models in optimizing ICI treatment decisions, paving the way for more precise and effective therapeutic strategies in this patient population.
Collapse
Affiliation(s)
- Camila Braganca Xavier
- MD Anderson Cancer Center, Houston, TX, 77030, United States
- Hospital Sírio-Libanês, São Paulo, SP, 01308-050, Brazil
| | | | | | | | - Beatriz M Awni
- Hospital Sírio-Libanês, São Paulo, SP, 01308-050, Brazil
| | | | - Denis L Jardim
- Hospital Sírio-Libanês, São Paulo, SP, 01308-050, Brazil
- Oncoclínicas&CO - Medica Scientia Innovation Research (MedSir), São Paulo, SP, 04538-132, Brazil
| | | |
Collapse
|
33
|
Li X, Wang H, Li X, Zeng M, He Z, Song L, Chen Z, Tang X, Wang A. An antibody-dependent cellular phagocytosis-related gene signature predicts survival and response to immunotherapy in stomach adenocarcinoma. Medicine (Baltimore) 2025; 104:e42079. [PMID: 40193680 PMCID: PMC11977745 DOI: 10.1097/md.0000000000042079] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 04/09/2025] Open
Abstract
Antibody-dependent cellular phagocytosis (ADCP) is an immune biological process and plays a biological role in the clearance of tumor cells and the response to immune checkpoint inhibitors. However, the effects of ADCP on stomach adenocarcinoma (STAD) remain unclear. Clinical and genomic data were extracted from multiple datasets. The ADCP-related signature was established using Cox least absolute shrinkage and selection operator regression. Expression of the C5a receptor also known as complement component 5a receptor 1 in the tumor and adjacent-normal tissues was calculated using immunohistochemistry staining. Validation of the signature was conducted in the training and validation cohorts by Cox regression and log-rank tests. Furthermore, the immune infiltrates, the tumor immune dysfunction and exclusion score, and tumor mutation burden score were calculated using the corresponding algorithms, and Mann-Whitney U tests were used to evaluate the differences between groups. Seventy-three hub genes with predictive performance were identified to establish an ADCP-related signature. Accordingly, a 27-gene signature was established, C5a receptor also known as complement component 5a receptor 1, one of the signature genes, had higher expression in tumors than adjacent-normal samples, and its predictive performance was validated in the GSE84437 and The Cancer Genome Atlas cohorts. We found that the ADCP-related signature is an excellent prognostic predictor of STAD. Moreover, the molecular characteristics and some indices of response to immunotherapy differed between the high- and low-risk groups. We constructed a 27-gene signature that is associated with the prognosis and response to STAD-based immunotherapy and provide insights into the biological mechanisms underlying this predictive function.
Collapse
Affiliation(s)
- Xiaochuan Li
- Department of Colorectal and Anorectal Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Hongjian Wang
- General Surgery Department of Yunfu People’s Hospital, Yunfu, China
| | - Xiaofeng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Miaoen Zeng
- Department of Gastroenterology, Fogang County People’s Hospital, Qingyuan, China
| | - Zhuguang He
- Department of Oncology, Zhaoqing First People’s Hospital, Zhaoqing, China
| | - Linjie Song
- Department of Colorectal and Anorectal Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Zhiming Chen
- Department of Integrated Traditional Chinese and Western Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xinyue Tang
- Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ang Wang
- Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
34
|
Ferraro DA, Bisig B, Rotzinger DC, Pareja F, Missiaglia E, Voutsadakis I, Homicsko K, Digklia A. Case Report: Lasting complete response to pembrolizumab in mismatch repair-deficient cardiac sarcoma: a genomic characterization. Front Oncol 2025; 15:1485386. [PMID: 40248199 PMCID: PMC12003144 DOI: 10.3389/fonc.2025.1485386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Sarcomas are traditionally considered "cold" tumors with poor response to immunotherapy. However, evidence accumulating over the last years shows that immune checkpoint inhibitors (ICIs) may have a role in selected sarcoma patients according to predictive markers. Here, we report the case of a woman diagnosed with a primary cardiac undifferentiated sarcoma. Following failure of standard first line chemotherapy, high-throughput sequencing (HTS) revealed a high tumor mutational burden (TMB), pathogenic mutations in FAT1 and NOTCH2 and a microsatellite instability (MSI)-associated signature. Immunohistochemistry confirmed mismatch repair-deficiency (MMRd) and abundant CD8+ tumor-infiltrating lymphocytes (TILs), in the absence of tertiary lymphoid structures. The patient was, therefore, treated with the ICI pembrolizumab, reaching a complete response that continues to persist at last follow-up, more than seven years from initial diagnosis and nearly six years from initiation of ICI treatment. This case illustrates the importance of performing HTS in rare sarcomas given the availability of efficient therapies, such as those for tumors displaying high TMB or MMRd/MSI. In agreement with other reports, it supports the contention that MMRd/MSI status and high numbers of TILs are valuable predictive markers of response to immunotherapy in sarcomas.
Collapse
Affiliation(s)
- Daniela A. Ferraro
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
| | - Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - David C. Rotzinger
- Department of Radiology, CHUV University Hospital, Lausanne, Switzerland
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ioannis Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON, Canada
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Krisztian Homicsko
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Antonia Digklia
- Department of Medical Oncology, CHUV University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
35
|
Zhang Y, Li Q, Lan J, Xie G, Zhang G, Cui J, Leng P, Wang Y. Triple-negative breast cancer molecular subtypes and potential detection targets for biological therapy indications. Carcinogenesis 2025; 46:bgaf006. [PMID: 39977309 DOI: 10.1093/carcin/bgaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer associated with poor prognosis. While chemotherapy remains the conventional treatment approach, its efficacy is limited and often accompanied by significant toxicity. Advances in precision-targeted therapies have expanded treatment options for TNBC, including immunotherapy, poly (ADP-ribose) polymerase inhibitors, androgen receptor inhibitors, cell cycle-dependent kinase inhibitors, and signaling pathway inhibitors. However, the heterogeneous nature of TNBC contributes to variations in treatment outcomes, underscoring the importance of identifying intrinsic molecular subtypes for personalized therapy. Additionally, due to patient-specific variability, the therapeutic response to targeted treatments is inconsistent. This highlights the need to strategize patients based on potential therapeutic targets for targeted drugs to optimize treatment strategies. This review summarizes the classification strategies and immunohistochemical (IHC) biomarkers for TNBC subtypes, along with potential targets for identifying indications for targeted drug therapy. These insights aim to support the development of personalized treatment approaches for TNBC patients.
Collapse
Affiliation(s)
- Yanchuan Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinghua Li
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Lan
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, Institute of Breast Health Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Guojing Xie
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guangjie Zhang
- Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, China
| | - Junhao Cui
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
| | - Ping Leng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingshuang Wang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu, China
- Sichuan Key Laboratory of Medical Molecular Testing, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
36
|
He H, Yan M, Ye K, Shi R, Tong L, Zhang S, Zhu Y, Zhan R. Predicting Prognosis and Immunotherapy Response in Glioblastoma (GBM) With a 5-Gene CAF-Risk Signature. Cancer Rep (Hoboken) 2025; 8:e70158. [PMID: 40226936 PMCID: PMC11995297 DOI: 10.1002/cnr2.70158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAF) represent significant constituents within the extracellular matrix (ECM) across a range of cancers. Nevertheless, there exists a scarcity of direct proof concerning the function of CAF in glioblastoma (GBM). AIMS This study endeavors to probe the participation of CAF in GBM by developing and validating a CAF-risk signature and exploring its correlation with immune infiltration and immunotherapy responsiveness. METHODS AND RESULTS To fulfill these objectives, mRNA expression profiles of GBM samples and their corresponding clinical data were retrieved from two databases. First, stromal CAF-associated genes were identified by weighted gene co-expression network analysis (WGCNA). This method constructs co-expression networks and pinpoints gene modules with similar expression patterns to detect relevant genes. Subsequently, a CAF-risk signature was established via univariate and LASSO Cox regression analyses. Thereafter, gene set enrichment analysis (GSEA) and single-sample gene set enrichment analysis (ssGSEA) were carried out to investigate the underlying molecular mechanisms. The immune status was evaluated with several R packages, and the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm was utilized to assess the response to immunotherapy. Validation was performed using single-cell RNA sequencing (scRNA) datasets, the Cancer Cell Line Encyclopedia (CCLE), and the Human Protein Atlas (HPA). Eventually, a 5-gene (ITGA5, MMP14, FN1, COL5A1, and COL6A1) prognostic CAF model was constructed. Notably, immune infiltration analysis demonstrated a significant correlation between Treg, Macrophage, and CAF risk scores. Moreover, TIDE analysis suggested a decreased responsiveness to immunotherapy in high CAF-risk patients. In addition, GSEA showed significant enrichment of the transforming growth factor alpha (TGF-α), inflammatory response, and epithelial-mesenchymal transition (EMT) pathways in this subgroup. Finally, the validation through scRNA, CCLE, and HPA datasets confirmed these findings. CONCLUSION The 5-gene CAF-risk signature exhibited accurate prognostic predictions and efficiently evaluated clinical immunotherapy responses among GBM patients. These results offer robust evidence regarding the implication of CAF in GBM and underscore the potential clinical value of personalized anti-CAF therapies in conjunction with immunotherapy.
Collapse
Affiliation(s)
- Haifeng He
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Min Yan
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Ke Ye
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Rui Shi
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Luqing Tong
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Shengxiang Zhang
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yu Zhu
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Renya Zhan
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
37
|
Wu YF, Zhao Q. Letter to the editor-Comment on: "Intrahepatic cholangiocarcinoma with arterial phase hyperenhancement and specialized tumor microenvironment associated with good prognosis after radical resection: A single-center retrospective study". Surgery 2025; 180:108854. [PMID: 39395858 DOI: 10.1016/j.surg.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/04/2024] [Indexed: 10/14/2024]
Affiliation(s)
- Yin-Fang Wu
- Department of Gastrointestinal and Minimally Invasive Surgery, Shaoxing Second Hospital, Shaoxing, Zhejiang, China.
| | - Qi Zhao
- Department of Gastrointestinal and Minimally Invasive Surgery, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
38
|
Isaacson AL, Berry RS, Ulici V, Armstrong SM, Bena J, John I, Karunamurthy A, Billings SD, Dermawan JK, Goldblum J, Kilpatrick SE, Rubin BP, Fritchie KJ. Myxofibrosarcoma with epithelioid morphology: A clinicopathological study of 44 cases with emphasis on differential diagnosis. Histopathology 2025; 86:694-703. [PMID: 39610041 DOI: 10.1111/his.15373] [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: 09/28/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
AIMS Epithelioid myxofibrosarcoma (eMFS) is an aggressive morphological variant associated with high rates of local recurrence and metastatic disease. The clinicopathological understanding of this disease is currently limited to a few small case-series. METHODS AND RESULTS We reviewed 44 cases of eMFS and classified them based on the presence of focal (< 50%) or diffuse (> 50%) epithelioid morphology and Fédération Nationale des Centres de Lutte Contre le Cancer (FNCLCC) grade. The patients (28 males; 16 females) had a median age of 71 years (range = 14-90 years). The proximal extremity was the most common site (n = 21), followed by the trunk (n = 11), distal extremity (n = nine) and head/neck (n = two). Of cases with known depth of involvement (n = 41), 39 involved the subcutis, one was limited to the dermis and one limited to the skeletal muscle. Most cases (n = 34, 77%) demonstrated diffuse (> 50%) epithelioid morphology and were FNCLCC grade 3 (n = 29, 66%). Follow-up data were available for 22 patients. Two developed local recurrence and 10 developed metastases, frequently to regional lymph nodes. All metastatic tumours had a primary lesion with diffuse epithelioid morphology (P = 0.09). There was no association between grade and recurrent or metastatic disease (P = 0.67 and 0.90, respectively). Three cases initially diagnosed as eMFS, one in the neck and two in the axilla, were found to have NRAS Q61R mutations and a high tumour mutation burden and/or ultraviolet (UV)-light DNA mutational signature. CONCLUSIONS These findings suggest that UV-driven malignancies (including melanoma or sarcomatoid squamous cell carcinoma) may histologically mimic eMFS and should be considered in cases of eMFS presenting at atypical anatomical sites.
Collapse
Affiliation(s)
- Alexandra L Isaacson
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ryan S Berry
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Veronica Ulici
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Susan M Armstrong
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - James Bena
- Section of Biostatistics, Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Ivy John
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Steven D Billings
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Josephine K Dermawan
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - John Goldblum
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Scott E Kilpatrick
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Brian P Rubin
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Karen J Fritchie
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
39
|
Kang YJ, Luo Q, Worthington J, Kelly A, Cuff J, Zalcberg J, Canfell K, Steinberg J. Informing health system planning for biomarker-based treatment: statistical prevalence projections for solid cancers with key pan-tumour biomarkers (dMMR, MSI, high TMB) in Australia to 2042. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2025; 57:101537. [PMID: 40242465 PMCID: PMC12002892 DOI: 10.1016/j.lanwpc.2025.101537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/23/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025]
Abstract
Background Targeted cancer treatment based on mismatch repair deficiency (dMMR), microsatellite instability (MSI), or high tumour mutational burden (TMB) holds promise for improving patient outcomes, but presents substantial healthcare costs. Methods Using validated statistical methods, we projected 1-year to 5-year prevalence of individuals diagnosed with solid tumours exhibiting these biomarkers in Australia to 2042, for all solid cancers combined and 23 individual cancer types/groups, and separately for all stages combined, advanced disease at diagnosis (here, distant metastasis/lymph node involvement), and advanced disease after progression post-diagnosis. Findings The 5-year prevalence of individuals diagnosed with any solid cancer regardless of biomarker status is estimated to increase by 54·2%, from 438,346 in 2018 to 675,722 in 2042 (advanced disease at diagnosis: by 37·6% from 109,855 to 151,199), primarily due to population growth and ageing. The 5-year prevalence of individuals whose tumours exhibit a biomarker is estimated to increase accordingly, e.g. for advanced disease at diagnosis, from 3983 to 5448 for dMMR, from 2484 to 3553 for MSI, and from 13,310 to 17,893 for high TMB (representing 3·6%, 2·3% and 11·8% of 5-year prevalence of individuals with advanced disease at diagnosis, respectively; noting considerable overlap in the presence of these biomarkers). Interpretation We present the first long-term projections for cancer prevalence associated with key pan-tumour biomarkers in Australia, to inform health policy and healthcare planning for targeted therapies. Funding Medical Research Future Fund-Preventive and Public Health Research Initiative-2019 Targeted Health System and Community Organization Research Grant Opportunity (MRF1200535), Cancer Institute NSW Career Development Fellowship (2022/CDF1154), National Health and Medical Research Council of Australia Investigator Grant (APP1194679).
Collapse
Affiliation(s)
- Yoon-Jung Kang
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, NSW, 2011, Australia
| | - Qingwei Luo
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, NSW, 2011, Australia
| | - Joachim Worthington
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, NSW, 2011, Australia
| | - Anna Kelly
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, NSW, 2011, Australia
| | - Jeff Cuff
- Research Advocate, The Daffodil Centre, NSW, 2011, Australia
- Faculty of Science Biotech and Biomolecular Science, University of New South Wales, NSW, 2033, Australia
| | - John Zalcberg
- Department of Medical Oncology, Alfred Health and School of Public Health and Preventive Medicine, Faculty of Medicine, Monash University, VIC, 3004, Australia
| | - Karen Canfell
- School of Public Health, The University of Sydney, NSW, 2050, Australia
| | - Julia Steinberg
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, NSW, 2011, Australia
| |
Collapse
|
40
|
Laplante P, Rosa R, Nebot-Bral L, Goulas J, Pouvelle C, Nikolaev S, Silvin A, Kannouche PL. Effect of MisMatch repair deficiency on metastasis occurrence in a syngeneic mouse model. Neoplasia 2025; 62:101145. [PMID: 39985912 PMCID: PMC11905862 DOI: 10.1016/j.neo.2025.101145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Mismatch repair deficiency leads to high mutation rates and microsatellite instability (MSI-H), associated with immune infiltration and responsiveness to immunotherapies. In early stages, MSI-H tumors generally have a better prognosis and lower metastatic potential than microsatellite-stable (MSS) tumors, especially in colorectal cancer. However, in advanced stages, MSI-H tumors lose this survival advantage for reasons that remain unclear. We developed a syngeneic mouse model of MSI cancer by knocking out the MMR gene Msh2 in the metastatic 4T1 breast cancer cell line. This model mirrored genomic features of MSI-H cancers and showed reduction in metastatic incidence compared to their MSS counterparts. In MSI-H tumors, we observed an enrichment of immune gene-signatures that negatively correlated with metastasis incidence. A hybrid epithelial-mesenchymal signature, related to aggressiveness was detected only in metastatic MSI-H tumors. Interestingly, we identified immature myeloid cells at primary and metastatic sites in MSI-H tumor-bearing mice, suggesting that MMR deficiency elicits specific immune responses beyond T-cell activation.
Collapse
Affiliation(s)
- Pierre Laplante
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Reginaldo Rosa
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Laetitia Nebot-Bral
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Jordane Goulas
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Caroline Pouvelle
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Sergey Nikolaev
- Paris-Saclay Université, Inserm-U981, Gustave Roussy, Villejuif, France
| | - Aymeric Silvin
- Paris-Saclay Université, Inserm-U1015, Gustave Roussy, Villejuif, France
| | - Patricia L Kannouche
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France.
| |
Collapse
|
41
|
Yang L, Feng Y, Liu X, Zhang Q, Liu Y, Zhang X, Li P, Chen D. DYNC2H1 mutation as a potential predictive biomarker for immune checkpoint inhibitor efficacy in NSCLC and melanoma. Invest New Drugs 2025; 43:199-213. [PMID: 39934438 DOI: 10.1007/s10637-024-01495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 02/13/2025]
Abstract
Dynein cytoplasmic 2 heavy chain 1 (DYNC2H1) is reported to play a potential role in cancer immunotherapy. However, the association between DYNC2H1 mutation and the clinical benefit of immunotherapy in non-small cell lung cancer (NSCLC) and melanoma remains to be elucidated. We collected data from three public immune checkpoint inhibitor (ICI)-treated NSCLC cohorts (n = 137 in total) and seven ICI-treated melanoma cohorts (n = 418 in total) to explore the potential of DYNC2H1 mutation as a predictive biomarker. The clinical outcomes, including the objective response rate (ORR) and progression-free survival (PFS), of patients with DYNC2H1 mutations are significantly better than those of patients with wild-type DYNC2H1. Multivariate Cox regression analysis confirmed that DYNC2H1 mutation was an independent predictive factor for ICI efficacy in NSCLC and melanoma. In addition, DYNC2H1 mutation exhibited no prognostic value for NSCLC or melanoma. Tumour mutational burden (TMB) and tumour neoantigen burden (TNB) were significantly higher in patients with DYNC2H1 mutation than in those with wild-type DYNC2H1 in both NSCLC and melanoma cohort. The analysis of immune-related genes and immune cell enrichment revealed an association between DYNC2H1 mutation and increased immune infiltration, revealing a potential mechanism underlying the predictive role of DYNC2H1 mutation in immunotherapy efficacy. In conclusion, DYNC2H1 mutation serves as a predictive biomarker of ICI efficacy in NSCLC and melanoma.
Collapse
Affiliation(s)
- Lu Yang
- Department of Science and Technology, Nanjing Forestry University, Nanjing, 210037, China
| | - Yanlong Feng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xuewen Liu
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Qin Zhang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Yaqin Liu
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Xing Zhang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China
| | - Ping Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Dongsheng Chen
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd, Nanjing Simcere Medical Laboratory Science Co., Ltd, Nanjing, 210002, China.
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
42
|
Nourbakhsh M, Zheng Y, Noor H, Chen H, Akhuli S, Tiberti M, Gevaert O, Papaleo E. Revealing cancer driver genes through integrative transcriptomic and epigenomic analyses with Moonlight. PLoS Comput Biol 2025; 21:e1012999. [PMID: 40258059 PMCID: PMC12058160 DOI: 10.1371/journal.pcbi.1012999] [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: 11/13/2024] [Revised: 05/07/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer involves dynamic changes caused by (epi)genetic alterations such as mutations or abnormal DNA methylation patterns which occur in cancer driver genes. These driver genes are divided into oncogenes and tumor suppressors depending on their function and mechanism of action. Discovering driver genes in different cancer (sub)types is important not only for increasing current understanding of carcinogenesis but also from prognostic and therapeutic perspectives. We have previously developed a framework called Moonlight which uses a systems biology multi-omics approach for prediction of driver genes. Here, we present an important development in Moonlight2 by incorporating a DNA methylation layer which provides epigenetic evidence for deregulated expression profiles of driver genes. To this end, we present a novel functionality called Gene Methylation Analysis (GMA) which investigates abnormal DNA methylation patterns to predict driver genes. This is achieved by integrating the tool EpiMix which is designed to detect such aberrant DNA methylation patterns in a cohort of patients and further couples these patterns with gene expression changes. To showcase GMA, we applied it to three cancer (sub)types (basal-like breast cancer, lung adenocarcinoma, and thyroid carcinoma) where we discovered 33, 190, and 263 epigenetically driven genes, respectively. A subset of these driver genes had prognostic effects with expression levels significantly affecting survival of the patients. Moreover, a subset of the driver genes demonstrated therapeutic potential as drug targets. This study provides a framework for exploring the driving forces behind cancer and provides novel insights into the landscape of three cancer sub(types) by integrating gene expression and methylation data.
Collapse
Affiliation(s)
- Mona Nourbakhsh
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Yuanning Zheng
- Department of Biomedical Data Science, Stanford Center for Biomedical Informatics Research, Palo Alto, California, United States of America
| | - Humaira Noor
- Department of Biomedical Data Science, Stanford Center for Biomedical Informatics Research, Palo Alto, California, United States of America
| | - Hongjin Chen
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Subhayan Akhuli
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Matteo Tiberti
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Olivier Gevaert
- Department of Biomedical Data Science, Stanford Center for Biomedical Informatics Research, Palo Alto, California, United States of America
| | - Elena Papaleo
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| |
Collapse
|
43
|
Zhou DDX, Dalrymple J, Klingberg D, Lin FPY, Lord SJ, Cooper WA, Zaheed M, Simes RJ, John T, Lee CK. Clinical Impact of Somatic Genomic Variants of Oncogenes and Tumor Suppressor Genes in Previously Treated Advanced Non-Small Cell Lung Cancer. JCO Precis Oncol 2025; 9:e2400673. [PMID: 40239138 DOI: 10.1200/po-24-00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/18/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
PURPOSE Next-generation sequencing in non-small cell lung cancer (NSCLC) identifies somatic genomic variants (SGVs) in cancer susceptibility genes (CSGs). We hypothesized that SGVs would be associated with poorer overall survival (OS) but greater benefit with immune checkpoint inhibitors over chemotherapy. We investigated the prevalence and predictive value of SGVs, using data from OAK and POPLAR trials comparing atezolizumab with docetaxel. METHODS We curated a list of SGVs (excluding TP53, EGFR, ALK, and ROS1) on the basis of CSGs associated with tumorigenesis. We classified participants as SGV mutant or wild-type using baseline plasma analyzed by the FoundationOne Liquid CDx assay. Cox regression analyses and interaction tests between SGV status and treatment were performed. RESULTS Of 762 participants, 29% harbored an SGV. The SGV mutant group had worse OS (hazard ratio [HR], 1.28, 95% CI, 1.06 to 1.54), and within each treatment arm (docetaxel: HR, 1.31; atezolizumab: HR, 1.27). In the atezolizumab arm, the SGV mutant group compared with wild-type had worse OS in the PD-L1 high (HR, 1.31 [95% CI, 0.59 to 2.91]) and low (HR, 1.38 [95% CI, 0.98 to 1.93]) subgroups. SGV with missense, splice, and nonsense mutations had significantly worse OS than wild-type in the docetaxel arm (log-rank P = .01) but not in the atezolizumab arm (log-rank P = .33). SGV status did not predict greater OS benefit with atezolizumab over docetaxel (interaction P = .67). CONCLUSION In advanced NSCLC after chemotherapy progression, plasma-detected SGVs are common, and associated with inferior OS. Plasma SGV status should be considered as a stratification factor in future trials.
Collapse
Affiliation(s)
- Deborah Di-Xin Zhou
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- St George Hospital, Kogarah, NSW, Australia
| | | | | | - Frank Po-Yen Lin
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Sarah J Lord
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW Health Pathology, Camperdown, NSW, Australia
- Faculty of Medicine and Health and Western Sydney University School of Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Milita Zaheed
- Prince of Wales Hereditary Cancer Centre, Randwick, NSW, Australia
| | - Robert John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Chee Khoon Lee
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- St George Hospital, Kogarah, NSW, Australia
| |
Collapse
|
44
|
Dykes KC, Ball ED. A review of antibody-based immunotherapy clinical trials for adult acute myeloid leukemia (AML): monoclonal antibodies (mAbs) and beyond. Expert Opin Biol Ther 2025; 25:345-362. [PMID: 40069930 DOI: 10.1080/14712598.2025.2479014] [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: 11/20/2024] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
INTRODUCTION Antibody-based immunotherapies are a class of therapeutics under active investigation in clinical trials for the treatment of acute myeloid leukemia (AML). Our review provides a comprehensive examination of trials published to date, focusing on recurrent challenges and promising aspects of antibody-based therapeutics. AREAS COVERED We described antibody-based immunotherapies for AML, specifically, an overview of the most prominent antigen targets in published clinical trials investigating monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor therapies. Manuscripts and abstracts describing clinical trials investigating antibody-based therapies for AML published through December 2024, identified by searching Google Scholar and PubMed, were included. EXPERT OPINION Antibody-based immunotherapies for AML have encountered limitations, including imperfect target antigens with significant associated toxicity such as myelosuppression, in addition to challenges specific to the AML patient population. The majority of trials have targeted CD33, CD123, CD371 (CLL1/Clec12), and CD47. For successful implementation of antibody-based therapeutics in AML treatment, future directions require creative applications of antibody-based therapeutics specifically engineered to minimize limiting toxicities and tailoring of therapies for this unique patient population.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/drug therapy
- Immunotherapy/methods
- Immunotherapy/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/pharmacology
- Adult
- Clinical Trials as Topic
- Antibodies, Bispecific/administration & dosage
Collapse
Affiliation(s)
- Kaitlyn C Dykes
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Edward D Ball
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
45
|
Geng Y, Yin T, Li Y, He K, Zou B, Yu J, Sun X, Zhang T, Teng F. Computed Tomography-Based Radiomics and Genomics Analyses for Survival Prediction of Stage III Unresectable Non-Small Cell Lung Cancer Treated With Definitive Chemoradiotherapy and Immunotherapy. Mol Carcinog 2025; 64:733-743. [PMID: 39835605 PMCID: PMC11890425 DOI: 10.1002/mc.23883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
The standard therapy for locally unresectable advanced non-small cell lung cancer (NSCLC) is comprised of chemoradiotherapy (CRT) before immunotherapy (IO) consolidation. However, how to predict treatment outcomes and recognize patients that will benefit from IO remain unclear. This study aimed to identify prognostic biomarkers by integrating computed tomography (CT)-based radiomics and genomics. Specifically, our research involved 165 patients suffering from unresectable Stage III NSCLC. Cohort 1 (IO following CRT) was divided into D1 (n = 74), D2 (n = 32), and D3 (n = 26) sets, and the remaining 33 patients treated with CRT alone were grouped in D4. According to the CT images of primary tumor regions, radiomic features were analyzed through the least absolute shrinkage and selection operator (LASSO) regression. The Rad-score was figured out to forecast the progression-free survival (PFS). According to the Rad-score, patients were divided into high and low risk groups. Next-generation sequencing was implemented on peripheral blood and tumor tissue samples in the D3 and D4 cohorts. The maximum somatic allele frequency (MSAF) about circulating tumor DNA levels was assessed. Mismatch repair and switching/sucrose non-fermenting signaling pathways were significantly enriched in the low-risk group compared to the high-risk group (p < 0.05). Moreover, patients with MSAF ≥ 1% and those showing a decrease in MSAF after treatment significantly benefited from IO. This study developed a radiomics model predicting PFS after CRT and IO in Stage III NSCLC and constructed a radio-genomic map to identify underlying biomarkers, supplying valuable insights for cancer biology.
Collapse
Affiliation(s)
- Yuxin Geng
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Tianwen Yin
- Cancer CenterUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
- Institute of Radiation OncologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Precision Radiation OncologyWuhanChina
| | - Yikun Li
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Kaixing He
- Department of Breast SurgeryShengli Oilfield Central HospitalDongyingShandongChina
| | - Bingwen Zou
- Department of Radiation OncologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Jinming Yu
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
- Cancer CenterUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
- Institute of Radiation OncologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Precision Radiation OncologyWuhanChina
| | - Xiao Sun
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Tao Zhang
- Cancer CenterUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
- Institute of Radiation OncologyUnion Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Precision Radiation OncologyWuhanChina
| | - Feifei Teng
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
46
|
Zhang F, Wu X, Jiao T, Du H, Guo Q, Cui C, Chi Z, Sheng X, Jiang D, Zhang Y, Wu J, Kong Y, Si L. Genomic characterization reveals distinct mutational landscape of acral melanoma in East Asian. J Genet Genomics 2025; 52:525-538. [PMID: 39798666 DOI: 10.1016/j.jgg.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025]
Abstract
Acral melanoma, the most common melanoma subtype in East Asia, is associated with a poor prognosis. This study aims to comprehensively analyze the genomic characteristics of acral melanoma in East Asians. We conduct whole-genome sequencing of 55 acral melanoma tumors and perform data mining with relevant clinical data. Our findings reveal a unique mutational profile in East Asian acral melanoma, characterized by fewer point mutations and structural variations, a higher prevalence of NRAS mutations, and a lower frequency of BRAF mutations compared to patients of European descent. Notably, we identify previously underestimated ultraviolet radiation signatures and their significant association with BRAF and NRAS mutations. Structural rearrangement signatures indicate distinct mutational processes in BRAF-driven versus NRAS-driven tumors. We also find that homologous recombination deficiency with MAPK pathway mutations correlated with poor prognosis. The structural variations and amplifications in EP300, TERT, RAC1, and LZTR1 point to potential therapeutic targets tailored to East Asian populations. The high prevalence of whole-genome duplication events in BRAF/NRAS-mutated tumors suggests a synergistic carcinogenic effect that warrants further investigation. In summary, our study provides important insights into the genetic underpinnings of acral melanoma in East Asians, creating opportunities for targeted therapies.
Collapse
Affiliation(s)
- Fenghao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Xiaowen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Tao Jiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Haizhen Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Qian Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China
| | - Dezhi Jiang
- Beijing Kanghui Biotechnology Co. LTD, Beijing 100101, China
| | - Yuhong Zhang
- Clinical Research Division of Berry Oncology Corporation, Beijing 102206, China
| | - Jiayan Wu
- Beijing Kanghui Biotechnology Co. LTD, Beijing 100101, China; Clinical Research Division of Berry Oncology Corporation, Beijing 102206, China; Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, Fujian 350020, China.
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China.
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Research Institute, Beijing 100142, China.
| |
Collapse
|
47
|
Chen L, Xiao H, Jiang J, Li B, Liu W, Huang W. The KMeansGraphMIL Model: A Weakly Supervised Multiple Instance Learning Model for Predicting Colorectal Cancer Tumor Mutational Burden. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:671-679. [PMID: 39800053 DOI: 10.1016/j.ajpath.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 01/15/2025]
Abstract
Colorectal cancer (CRC) is one of the top three most lethal malignancies worldwide, posing a significant threat to human health. Recently proposed immunotherapy checkpoint blockade treatments have proven effective for CRC, but their use depends on measuring specific biomarkers in patients. Among these biomarkers, tumor mutational burden (TMB) has emerged as a novel indicator, traditionally requiring next-generation sequencing for measurement, which is time-consuming, labor intensive, and costly. To provide an economical and rapid way to predict patients' TMB, the KMeansGraphMIL model was proposed based on weakly supervised multiple-instance learning. Compared with previous weakly supervised multiple-instance learning models, KMeansGraphMIL leveraged both the similarity of image patch feature vectors and the spatial relationships between patches. This approach improved the model's area under the receiver operating characteristic curve to 0.8334 and significantly increased the recall to 0.7556. Thus, this study presents an economical and rapid framework for predicting CRC TMB, offering the potential for physicians to quickly develop treatment plans and saving patients substantial time and money.
Collapse
Affiliation(s)
- Linghao Chen
- Radiology Department, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Huiling Xiao
- Radiology Department, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jiale Jiang
- Department of Medical Imaging, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Bing Li
- Department of Medical Imaging, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Weixiang Liu
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen, China.
| | - Wensheng Huang
- Radiology Department, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
48
|
Wang CW, Muzakky H, Lee YC, Chung YP, Wang YC, Yu MH, Wu CH, Chao TK. Interpretable multi-stage attention network to predict cancer subtype, microsatellite instability, TP53 mutation and TMB of endometrial and colorectal cancer. Comput Med Imaging Graph 2025; 121:102499. [PMID: 39947084 DOI: 10.1016/j.compmedimag.2025.102499] [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: 10/15/2024] [Revised: 12/12/2024] [Accepted: 01/22/2025] [Indexed: 03/03/2025]
Abstract
Mismatch repair deficiency (dMMR), also known as high-grade microsatellite instability (MSI-H), is a well-established biomarker for predicting the immunotherapy response in endometrial cancer (EC) and colorectal cancer (CRC). Tumor mutational burden (TMB) has also emerged as an important quantitative genomic biomarker for assessing the efficacy of immune checkpoint inhibitors. Although next-generation sequencing (NGS) can be used to assess MSI and TMB, the high costs, low sample throughput, and significant DNA requirements make NGS impractical for routine clinical screening. In this study, an interpretable, multi-stage attention deep learning (DL) network is introduced to predict pathological subtypes, MSI, TP53 mutations, and TMB directly from low-cost, routinely used histopathological whole slide images of EC and CRC slides. Experimental results showed that this method consistently outperformed seven state-of-the-art approaches in cancer subtyping and molecular status prediction across EC and CRC datasets. Fisher's Least Significant Difference test confirmed a strong correlation between model predictions and actual molecular statuses (MSI, TP53, and TMB) (p<0.001). Furthermore, Kaplan-Meier disease-free survival analysis revealed that CRC patients with model-predicted high TMB had significantly longer disease-free survival than those with low TMB (p<0.05). These findings demonstrate that the proposed DL-based approach holds significant potential for directly predicting immunotherapy-related pathological diagnoses and molecular statuses from routine WSIs, supporting personalized cancer immunotherapy treatment decisions in EC and CRC.
Collapse
Affiliation(s)
- Ching-Wei Wang
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan.
| | - Hikam Muzakky
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Yu-Ching Lee
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Yu-Pang Chung
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Yu-Chi Wang
- Department of Gynecology and Obstetrics, Tri-Service General Hospital, Taipei, Taiwan; Department of Gynecology and Obstetrics, National Defense Medical Center, Taipei, Taiwan
| | - Mu-Hsien Yu
- Department of Gynecology and Obstetrics, Tri-Service General Hospital, Taipei, Taiwan; Department of Gynecology and Obstetrics, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Hua Wu
- Department of Pathology, Tri-Service General Hospital, Taipei, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology, Tri-Service General Hospital, Taipei, Taiwan; Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
49
|
Zeng J, Wu Z, Luo M, Chen Z, Xu X, Xie G, Chen Q, Bai W, Xiao G, Xie J. Identification of a long non-coding RNA signature associated with cuproptosis for prognosis and immunotherapy response prediction in patients with lung adenocarcinoma. Discov Oncol 2025; 16:432. [PMID: 40163162 PMCID: PMC11958909 DOI: 10.1007/s12672-025-02092-3] [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: 06/12/2024] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD), the most common histotype of lung cancer, exhibits high heterogeneity due to molecular variations. Cuproptosis is a newly discovered type of cell death that is linked to copper metabolism and long non-coding RNAs (lncRNAs) may play a significant role in this process. We conducted a comprehensive analysis of lncRNA related to cuproptosis and identified a CRLscore to predict the prognosis and immune landscape for LUAD patients. METHODS The LUAD patient cohort obtained from TCGA database was divided into training and validation sets. A range of statistical methods were employed to identify lncRNAs associated with cuproptosis. Multivariate Cox regression was then utilized to develop the CRLscore, which was further used to construct and evaluate a nomogram. Additionally, we investigated the biological functions, gene mutations, and immune landscape. RESULTS A CRLscore, comprising six cuproptosis-related lncRNAs, was developed to stratify patients into high- and low-risk groups. The CRLscore demonstrated its ability to independently predict prognosis in both the training set and the validation set. Utilizing the CRLscore, we constructed a nomogram that exhibited favorable predictive efficiency. Furthermore, the cuproptosis-related lncRNAs exhibited associations with important signaling pathways such as p53 signaling, MYC Targets V1, and G2M Checkpoint. Notably, the CRLscore displayed substantial differences in somatic mutations and immune landscape. Finally, qRT-PCR results showed the significant differential expression of five cuproptosis-related lncRNAs between LUAD and normal cells. CONCLUSION The CRLscore could serve as a potential prognostic indicator and may predict the response to immunotherapy in LUAD patients.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhenyu Wu
- Department of Urology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Meijuan Luo
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhibo Chen
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Xie Xu
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Guijing Xie
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Quhai Chen
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Wenjie Bai
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Gang Xiao
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| | - Jianjiang Xie
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
50
|
Leng X, Chen H, Chen G. Construction and validation of a reliable disulfidptosis-related lncRNAs signature of the subtype, prognostic, and immune landscape in bladder cancer. Discov Oncol 2025; 16:418. [PMID: 40153109 PMCID: PMC11953504 DOI: 10.1007/s12672-025-02174-2] [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: 11/16/2024] [Accepted: 03/18/2025] [Indexed: 03/30/2025] Open
Abstract
BACKGROUND Bladder cancer (BLCA) is one of the most frequently-diagnosed tumors globally. Disulfidptosis represents a critical framework for cell death mechanism in cancer therapy. Our study constructed a predictive model utilizing disulfidptosis-related lncRNAs (DRLs) to provide value in evaluating diagnosis, drug sensibility, and prognosis of BLCA patients. METHODS The study data of BLCA patients retrieved from TCGA-BLCA database. Cox and LASSO regression analysis were used to identify DRLs. Kaplan-Meier survival analysis, ROC curve, and nomograms were constructed to assess and forecast survival events. GSEA were performed to illustrate relevant enrichments results. Tumor mutation burden (TMB), immune status, and drug sensitivity were assessed. Muscle invasive bladder cancer (MIBC) tumor and tumor-adjacent normal tissues samples were collected in our department to validate the DRLs expression levels by RT-PCR. RESULTS Overall, nine DRLs (AL590428.1, LSAMP-AS1, LINC01184, LINC-PINT, AC023825.2, AC010331.1, AC009716.1, AC104785.1, AC008764.6) were identified. These DRLs were used to calculate risk scores and create a prognostic model. ROC revealed higher diagnostic efficiency of the model than other clinical characteristics. Nomogram was constructed using the risk scores, age, and tumor stage, which showed excellent predictive power and was verified by calibration graph. BLCA patients were further classified into high-risk group and low-risk group using median risk score as the cut-off value. The high-risk group showed lesser TMB levels and developed worse prognosis. GSEA of the high-risk group identified pathways associated with BLCA progression such as WNT signaling pathway. Immune cells including CD4+ and CD8+ T cells, and immune-related function like T cell co-stimulation also showed remarkable differences between two risk groups. Furthermore, IC50 values of twelve drugs such as Sorafenib, Nilotinib, and Navitoclax were significantly higher in the high-risk group. RT-PCR results revealed that 9 DRLs expression levels were statistically significant between tumor tissues samples and tumor-adjacent normal tissues samples. The expression trends of these DRLs in clinical tissues samples were the same as the findings in TCGA dataset. CONCLUSION Based on this study, it would be advisable to identify the key DRLs with potential prognostic value in BLCA to enhance the evaluation of clinical outcomes in this context.
Collapse
Affiliation(s)
- Xiaoping Leng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Han Chen
- Department of Urological Oncology Surgery, Chongqing University Cancer Hospital (Chongqing Cancer Institute & Chongqing Cancer Hospital), Chongqing, China
| | - Gang Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|